You are on page 1of 243

Lakkis_FM_i-xiv r1.

qxd 3/29/07 1:26 PM Page i

1
2
3

Encapsulation and 4
5
6
7

Controlled Release 8
9
10

Technologies 11
12
13
14

in Food Systems 15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
S46
N47
Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page iii

1
2
3

Encapsulation and 4
5
6
7

Controlled Release 8
9
10

Technologies 11
12
13
14

in Food Systems 15
16
17
18
19
20
21
22
23
Edited by Jamileh M. Lakkis 24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
S46
N47
Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page iv

1 Jamileh M. Lakkis, Ph.D., has 14 years experience in the food, dietary supplements, and consumer
2 products industries. She served as Senior Project Manager at Pfizer/Cadbury-Schweppes, Morris
3 Plains, NJ, focusing on designing confectionery products as delivery systems for oral care benefits.
4 As a Senior Encapsulation Specialist for General Mills, Inc., Minneapolis, MN, Dr. Lakkis designed
5 several microencapsulation processes for stabilizing and masking the taste/aroma of a variety of
functional and nutraceutical actives for their applications in breakfast cereals, dairy, confections, and
6
shelf-stable bakery products. Her professional experience also includes engagements as Senior
7 Research Scientist at Land O’Lakes, Inc., Arden Hills, MN. Dr. Lakkis co-organized the first IFT
8 symposium on microencapsulation and controlled release applications in food systems. She is an
9 active member of the Controlled Release Society and serves on the society’s newsletter editorial
10 board representing the Consumer and Diversified Products Division.
11
12 ©2007 Blackwell Publishing
13 All rights reserved
14 Blackwell Publishing Professional
15 2121 State Avenue, Ames, Iowa 50014, USA
16
17 Orders: 1-800-862-6657
18 Office: 1-515-292-0140
19 Fax: 1-515-292-3348
20 Web site: www.blackwellprofessional.com
21 Blackwell Publishing Ltd.
22 9600 Garsington Road, Oxford OX4 2DQ, UK
23 Tel.: +44 (0)1865 776868
24
25 Blackwell Publishing Asia
26 550 Swanston Street, Carlton, Victoria 3053, Australia
27 Tel.: +61 (0)3 8359 1011
28 Authorization to photocopy items for internal or personal use, or the internal or personal use of specific
29 clients, is granted by Blackwell Publishing, provided that the base fee is paid directly to the Copyright
30 Clearance Center, 222 Rosewood Drive, Danvers, MA 01923. For those organizations that have been
31 granted a photocopy license by CCC, a separate system of payments has been arranged. The fee codes
32 for users of the Transactional Reporting Service is ISBN-13: 978-0-8138-2855-8/2007.
33
34 First edition, 2007
35 Library of Congress Cataloging-in-Publication Data
36
37 Encapsulation and controlled release technologies in food systems / edited by Jamileh M. Lakkis,
38 Ph. D.—1st ed.
39 p. cm.
40 Includes bibliographical references and index.
41 ISBN 978-0-8138-2855-8 (alk. paper)
42 1. Controlled release technology. 2. Microencapsulation. 3. Food—Analysis. I. Lakkis,
Jamileh M.
43
44 TP156.C64E53 2007
45 664'.024—dc22
46S 2007006839
47N
The last digit is the print number: 9 8 7 6 5 4 3 2 1
Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page v

1
2
3
4
5
I dedicate this book to LEBANON 6
Which had not been my country, I’d have chosen it to be 7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
S46
N47
Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page vii

Table of Contents 1
2
3
4
5
6
Dedication v 7
Contributors ix 8
Preface xi 9
Jamileh M. Lakkis 10
11
1. Introduction 1 12
Jamileh M. Lakkis 13
14
2. Improved Solubilization and Bioavailability of Nutraceuticals 15
in Nanosized Self-Assembled Liquid Vehicles 13 16
Nissim Garti, Eli Pinthus, Abraham Aserin, and Aviram Spernath 17
18
3. Emulsions as Delivery Systems in Foods 41 19
Ingrid A.M. Appelqvist, Matt Golding, Rob Vreeker, and Nicolaas Jan Zuidam 20
21
4. Applications of Probiotic Encapsulation in Dairy Products 83 22
Ming-Ju Chen and Kun-Nan Chen 23
24
5. Encapsulation and Controlled Release in Bakery Applications 113 25
Jamileh M. Lakkis 26
27
6. Encapsulation Technologies for Preserving and Controlling 28
the Release of Enzymes and Phytochemicals 135 29
Xiaoyong Wang, Yan Jiang, and Qingrong Huang 30
31
7. Microencapsulation of Flavors by Complex Coacervation 149 32
Curt Thies 33
34
8. Confectionery Products as Delivery Systems for Flavors, 35
Health, and Oral-Care Actives 171 36
Jamileh M. Lakkis 37
38
9. Innovative Applications of Microencapsulation in Food Packaging 201 39
Murat Ozdemir and Tugba Cevik 40
41
10. Marketing Perspective of Encapsulation Technologies 42
in Food Applications 213 43
Kathy Brownlie 44
45
Index 235 S46
N47

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

Contributors 1
2
3
4
5
6
Ingrid A.M. Appelqvist Nissim Garti 7
Unilever Food and Health Research Casali Institute of Applied Chemistry 8
Institute The Institute of Chemistry 9
Unilever R&D Vlaardingen The Hebrew University of Jerusalem 10
The Netherlands Jerusalem, Israel 11
Chapter 3 Nutralease Ltd., Mishor Adumim, Israel 12
Chapter 2 13
Abraham Aserin 14
Casali Institute of Applied Chemistry Matt Golding 15
The Institute of Chemistry Unilever Food and Health Research 16
The Hebrew University of Jerusalem Institute 17
Jerusalem, Israel Unilever R&D Vlaardingen 18
Nutralease Ltd., Mishor Adumim, Israel The Netherlands 19
Chapter 2 Chapter 3 20
21
Kathy Brownlie Qingrong Huang 22
Manager, Global Programme Department of Food Science 23
Frost & Sullivan Rutgers University 24
Oxford, England, UK New Brunswick, NJ 25
Chapter 10 Chapter 6 26
27
Tugba Cevik Nicolaas Jan Zuidam 28
Department of Chemical Unilever Food and Health Research 29
Engineering Institute 30
Section of Food Technology Unilever R&D Vlaardingen 31
Gebze Institute of Technology The Netherlands 32
Gebze-Kocaeli, Turkey Chapter 3 33
Chapter 9 34
Yan Jiang 35
Kun-Nan Chen Department of Food Science 36
Department of Mechanical Rutgers University 37
Engineering New Brunswick, NJ 38
Tung Nan Institute of Technology Chapter 6 39
Taipei, Taiwan 40
Chapter 4 Jamileh Lakkis 41
Senior Project Manager 42
Ming-Ju Chen Formerly with Pfizer/Cadbury-Schweppes 43
Department of Animal Science Morris Plains, NJ 44
National Taiwan University Chapter 1 45
Taipei, Taiwan Chapter 5 S46
Chapter 4 Chapter 8 N47

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

x Contributors

1 Murat Ozdemir Curt Thies


2 Department of Chemical Engineering Thies Technology
3 Section of Food Technology Henderson, Nevada
4 Gebze Institute of Technology Chapter 7
5 Gebze-Kocaeli, Turkey
6 Chapter 9 Rob Vreeker
7 Unilever Food and Health Research
8 Eli Pinthus Institute
9 Nutralease Ltd., Mishor Adumim, Israel Unilever R&D Vlaardingen
10 Adumim Food Ingredients The Netherlands
11 Mishor Adumim, Israel Chapter 3
12 Chapter 2
13 Xiaoyong Wang
14 Aviram Spernath Department of Food Science
15 Casali Institute of Applied Chemistry Rutgers University
16 The Institute of Chemistry New Brunswick, NJ
17 The Hebrew University of Jerusalem Chapter 6
18 Jerusalem, Israel
19 Chapter 2
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S
47N
Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page xi

Preface 1
2
3
4
5
6
Encapsulation and controlled release technologies have enjoyed their fastest growth in the 7
last two decades. These advances, pioneered by pharmaceutical companies, were a result 8
of: (1) the rapid change in drug development strategies to target specific organs or even 9
cells, (2) physicians’ growing concern about patient non-compliance, and (3) pharmaceuti- 10
cal companies desire to extend their market monopoly on new drugs for a certain period of 11
time as provided by the US and international patent laws. 12
Despite this progress, encapsulation and controlled release technologies have only been 13
recently adopted by the food industry. Food researchers and technologists have often been 14
confronted with the dilemma of how to translate all these advances from the drug arena into 15
practical applications in food systems. By searching the literature, one can find volumes of 16
books and specialized publications on encapsulation and controlled release technologies. 17
Unfortunately, most of these publications have dealt with theoretical aspects of these tech- 18
nologies with little emphasis on real applications in consumer and food products. 19
This book attempts to illustrate various aspects of encapsulation and controlled release 20
applications in food systems using practical examples. These examples will give the reader 21
an appreciation for the delicate art of designing encapsulated ingredients and the enormous 22
challenges in incorporating them into food formulations. Most of the practical examples in 23
this book were borrowed from the patent literature. This approach might be questioned 24
based on the fact that patents applications are never peer reviewed, but seems justifiable 25
considering the frantic effort by both industry and academia to protect their discoveries and 26
to gain limited-time monopoly on their innovations, thus limiting the availability of such 27
information in peer-reviewed articles. 28
This publication has several potential uses. It is a reference book for scientists in the 29
food, nutraceuticals and consumer products industries who are looking to introduce 30
microencapsulated ingredients into new or existing formulations. It is also a post-graduate 31
text designed to give students some comprehension of various aspects of encapsulation and 32
controlled release in food systems. 33
This book is organized in such a way that each chapter treats one major application of 34
encapsulation and controlled release technologies in foods. 35
Chapter 1 introduces the readers to various encapsulation and controlled release tech- 36
nologies, as well as release mechanisms, suitable for applications in foods, nutraceuticals 37
and consumer products. 38
Chapter 2 by Professor Nissim Garti and his collaborators discusses a novel approach to 39
encapsulation and controlled release via reverse microemulsion technique referred to as 40
nanosized self-assembled liquids (NSSL). Such systems are shown to provide exceptional 41
thermodynamic stability in a wide pH range. In addition to enhancing bioavailability of 42
functional active ingredients, NSSL systems, by virtue of their unique transparent appear- 43
ance, are excellent candidates for beverage applications. 44
Chapter 3, by Dr. Klaas-Jan Zuidam and co-workers, presents an elaborate approach to 45
understanding emulsions and their benefits as delivery systems in food applications. This S46
chapter discusses various mechanisms of emulsion stabilization and destabilization and N47

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

xii Preface

1 how they can best be designed for targeted delivery of flavors and functional ingredients in
2 the human gastrointestinal system.
3 Chapter 4 on encapsulation and controlled release of probiotics by Drs. Chen and Chen
4 reports on approaches for encapsulating probiotic bacteria in dairy products as well as in
5 the human gastrointestinal tract. This chapter also discusses novel optimization techniques
6 for stabilizing these beneficial bacteria and enhancing their survival rates.
7 Chapter 5, written by the editor of this book, highlights current approaches to encapsula-
8 tion and controlled release technologies for bakery products applications. Current encapsula-
9 tion practices such as hot-melt particle coating and spray chilling are discussed. Examples of
10 the performance of encapsulated leavening agents as well as sweeteners and flavors are
11 presented in shelf-stable bakery applications.
12 Chapter 6 on nanoencapsulation technology by Dr. Huang and his collaborators deals
13 with novel approaches to encapsulate enzymes and nutraceuticals. Specific examples are
14 presented on stabilization of phytochemicals and their enhanced bioavailability via incor-
15 poration into nanoemulsions and bioconjugation systems.
16 Chapter 7 on flavor encapsulation via complex coacervation is written by Dr. Curt Thies.
17 Discussion is focused on the basic principle of complex coacervation technique as a liquid–
18 liquid polymer phase separation phenomenon. Guidance on polymer selection and subse-
19 quent implications on the physicochemical properties of capsules as well as their release
20 behavior is provided.
21 Chapter 8, written by the editor of this book, details techniques used for delivering ther-
22 apeutic as well as functional actives and flavors via confectionery products. Technologies
23 and subsequent applications discussed in this chapter have wide applications in the food,
24 nutraceuticals, as well as pharmaceutical arenas. Mechanisms and challenges specific to
25 targeted release in upper gastrointestinal tract, especially the mouth and throat areas will be
26 described in great detail.
27 Chapter 9 discusses encapsulation and controlled release of actives in packaging appli-
28 cations by Dr. Ozdemir and collaborator. In this contribution, the authors provide examples
29 on embedding fragrances, pigments as well as antimicrobial and insect repellent agents
30 into food packaging films.
31 Chapter 10, authored by Ms. Kathy Brownlie, provides a marketing perspective of micro-
32 encapsulation technologies and their potential impact on the food industry. Ms. Brownlie
33 offers an in-depth assessment of market drivers as well as constraints that are still hindering
34 wider implementation of these technologies in food manufacturing.
35 This book has definitely surpassed my vision and expectations thanks to the contributors
36 that I am grateful to all of them for their expertise, commitment, and dedication. It is my
37 hope that this book will prove itself a useful source on encapsulation and controlled release
38 in a wide range of food and consumer product applications.
39 Many thanks to the editorial staff at Blackwell Publishing Co., especially to Mark
40 Barrett and Susan Engelken for their valuable help and advice throughout this project.
41 Last but not least, I would like to thank my parents who taught me the importance of
42 working hard, having clear goals, and standing for what I believe is right. It is a lesson that
43 guides me in everything I do.
44
45 Jamileh M. Lakkis
46S
47N
Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 1

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

1 Introduction 1
2
3
4
Jamileh M. Lakkis 5
6
7
8
9
The European Directive (3AQ19a) defines controlled release as a “modification of the rate 10
or place at which an active substance is released.” Such a modification can be made using 11
materials with specific barrier properties for manipulating the release of an active and to 12
provide unique sensory and/or functional benefits. 13
Addition of small amounts of nutrients to a food system, for example, may not affect its 14
properties significantly; however, incorporating high levels of the nutrient either to meet 15
certain requirements or to treat an ailment will most often result in unstable and often 16
unpalatable foods. Examples of such nutrients include fortification with calcium, vitamins, 17
polyunsaturated fatty acids, and so on, and the associated grittiness, medicinal and oxi- 18
dized taste, respectively. Different types of controlled-release systems have been formu- 19
lated to overcome these challenges and to provide a wide range of release requirements. 20
The two principal modes of controlled release are delayed and sustained release 21
(Figure 1.1). 22
23
• Delayed release is a mechanism whereby the release of an active substance is delayed 24
from a finite “lag time” up to a point when/where its release is favored and is no longer 25
hindered. Examples of this category include encapsulating probiotic bacteria for their 26
protection from gastric acidity and further release in the lower intestine, flavor release 27
upon microwave heating of ready-meals or the release of encapsulated sodium bicarbon- 28
ate upon baking of a dough or cake batter. 29
• Sustained release is a mechanism designed to maintain constant concentration of an 30
active at its target site. Examples of this release pattern include encapsulating flavors and 31
sweeteners for chewing gum applications so that their rate of release is reduced to main- 32
tain a desired flavor effect throughout the time of chewing. 33
34
A wide range of cores (encapsulants), wall-forming materials (encapsulating agents), and 35
technologies for controlling the interactions of ingredients in a given food system and for 36
manufacturing microcapsules and microparticles of different size, shape, and morphologi- 37
cal properties are commercially viable. 38
39
40
Wall-Forming Materials
41
Materials used in film coating or matrix formation include several categories: 42
43
1. Waxes and lipids: beeswax, candelilla and carnauba waxes, wax micro- and wax macro- 44
emulsions, glycerol distearate, natural and modified fats. 45
2. Proteins: gelatins, whey proteins, zein, soy proteins, gluten, and so on. All these proteins S46
are available both in native and modified forms. N47

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

2 Chapter 1

1
2
3
4
5 Sustained (long-lasting) release
6
7
8
9
10
11
Delayed release
12
13 time
14
15 Figure 1.1. Generic representation of “sustained” and “delayed” release profiles.
16
17
18
3. Carbohydrates: starches, maltodextrins, chitosan, sucrose, glucose, ethylcellulose, cel-
19
lulose acetate, alginates, carrageenans, chitosan, and so on.
20
4. Food grade polymers: polypropylene, polyvinylacetate, polystyrene, polybutadiene, and
21
so on.
22
23
24
25 Core Materials
26 Core materials include flavors, antimicrobial agents, nutraceutical and therapeutic actives,
27 vitamins, minerals, antioxidants, colors, acids, alkalis, buffers, sweeteners, nutrients,
28 enzymes, cross-linking agents, yeasts, chemical leavening agents, and so on.
29
30
31 Release Triggers
32
33 Encapsulation and controlled-release systems can be designed to respond to one or a com-
34 bination of triggers that can activate the release of the entrapped substance and to meet a
35 desired release target or rate. Triggers can be one or a combination of the following:
36
37 • temperature: fat/wax matrices
38 • moisture: hydrophilic matrices
39 • pH: enteric coating, emulsion coalescence, and others.
40 • Enzymes: enteric coating as well as a variety of lipid, starch and protein matrices.
41 • Shear: chewing, physical fracture, and grinding
42 • lower critical solution temperature (LCST) of hydrogels.
43
44 Payload is a term used to estimate the amount of active (core) entrapped in a given matrix
45 or wall material (shell). Payload is expressed as:
46S
47N Payload (%) = [(core)/(core + shell)] × 100
Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 3

Introduction 3

Entrapment of Actives in Food Matrices 1


2
Entrapment in an Amorphous Matrix 3
Encapsulation of active into an amorphous matrix, generally, involves melting a crystalline 4
polymer using heat and/or shear to transform the molecular structure into an amorphous 5
phase. The encapsulant is then incorporated into the metastable amorphous phase followed 6
by cooling to solidify the structure and form glass, thus restricting molecular movements. 7
Carbohydrates are excellent candidates for encapsulation applications due to the several 8
attributes possessed by them. 9
10
11
1. They form an integral part of many food systems. 12
2. They are cost-effective. 13
3. They occur in a wide range of polymer sizes. 14
4. They have desirable physicochemical properties such as solubility, melting, phase 15
change and so on. 16
17
Sucrose, maltodextrins, native and modified starches, polysaccharides, and gums have been 18
used in encapsulating flavors, minerals, vitamins, probiotic bacteria as well as pharmaceu- 19
tical actives. The unique helical structure of the amylose molecule, for example, makes 20
starch a very efficient vehicle for encapsulating molecules like lipids, flavors, and so on 21
(Conde-Petit et al., 2006). Some carbohydrates such as inulin and trehalose can provide 22
additional benefits for encapsulation applications. Inulin, for example, is a prebiotic ingre- 23
dient that can enhance survival of probiotic bacteria while trehalose serves as a support 24
nutrient for yeasts. 25
Two main technologies—spray drying and extrusion—have been used in large-scale 26
encapsulation applications into amorphous matrices, though using different mechanisms. 27
In spray drying, for example, the active is trapped within porous membranes of hollow 28
spheres, while in extrusion the goal is to entrap the active in a dense, impermeable glass. 29
Encapsulating actives via spray drying requires emulsifying the substrate into the encap- 30
sulating agent. This is important for flavor applications, in particular, considering the fact 31
that most flavors are made up of components of various chemistries (polarity, hydrophobic 32
to hydrophilic ratios), thus limiting their stability when dispersed or suspended in different 33
solvents. Hydrophobicity is one of the most critical attributes that can play a significant role 34
in determining flavors’ payload as well as their release in food systems. 35
The basic principle of spray drying has been adequately covered by Masters (1979). 36
Briefly, the process comprises atomizing a micronized (1–10 micron droplet size) emulsion 37
or suspension of an active and an encapsulating substance and further spraying the same 38
into a chamber. Drying takes place at relatively high temperatures (210°C inlet and 90°C 39
outlet), though the emulsion’s exposure to these temperatures lasts only for few seconds. 40
The process results in free flowing, low bulk density powders of 10–100 micron size. 41
Optimal payloads of 20% can be expected for flavors encapsulated in starch matrices. 42
Maltodextrins and sugars with lower molecular weight, due to their low viscosities and 43
inadequate emulsifying activities, result in lower flavor payloads. 44
Several factors can impact the efficiency of encapsulation via spray drying, mainly those 45
related to the emulsion (solid content, molecular weight, emulsion droplet size, and viscos- S46
ity) and to the process (feed flow rate, inlet/outlet temperature, gas velocity, and so on). N47
Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 4

4 Chapter 1

1 Release of flavors from spray-dried matrices takes place upon reconstitution of the dried
2 emulsion in the release medium, water most often. Reasonable prediction of the release
3 behavior should take into consideration the complex chemistry of flavors and the prevailing
4 partition and phase transport mechanisms between aqueous and non-aqueous phases
5 (Larbouss et al., 1991; Shimada et al., 1991).
6 Encapsulation into an amorphous matrix via extrusion has gained wide popularity in the
7 last two decades with applications ranging from entrapping flavors for their controlled
8 release to masking the grittiness of minerals and vitamins. Hot melt extrusion is a highly
9 integrated process with many unique advantages for encapsulation applications, namely:
10
11 1. Extruders are multifunctional systems (many unit operations) that can be manipulated
12 to provide desired processing temperature and shear rate profiles by varying screw
13 design, barrel heating, mixing speed, feed rate, moisture content, plasticizers, and so on.
14 2. Possibility of incorporating actives and other ingredients at different points of the extru-
15 sion process. Heat-labile actives, for example, can be incorporated via temperature-
16 controlled inlets toward the end of the barrel and their residence time in the extruder can
17 be minimized to avoid degradation of the active and to preserve its integrity.
18 3. Extruders are also formers—encapsulated products can be recovered in practically any
19 desired shape or size (pellets, rods, ropes, and so on).
20 4. Only very limited amount of water is needed to transform carbohydrates from their
21 native crystalline structure to amorphous glassy matrices in an extruder, thus limiting
22 the need for expensive downstream drying.
23 5. High payload—up to 30% can be expected when encapsulating solid actives in extruded
24 pellets.
25 6. Economics—attributes such as high throughput, continuous mode, and limited need
26 for drying make extrusion a very attractive process for manufacturing encapsulated
27 ingredients.
28
29 Figure 1.2 describes a typical melt extrusion encapsulation process. Carbohydrate (encapsu-
30 lating matrix), a mixture of sucrose and maltodextrin, is dry fed and melted by a combina-
31 tion of heat and shear in the extruder barrel so that the crystalline structure is transformed
32 into an amorphous phase. The encapsulant (flavor or other active) is added through an open-
33 ing in a cooled barrel situated toward the die to avoid flashing off of low boiling components.
34 The amorphous mixture exits the die in the form of a rope that can be cooled quickly by air
35 or liquid nitrogen to form a solid glassy material. The latter can be ground to a desired
36 particle size to form compact microparticles of high bulk density.
37 Using this technology, encapsulated products can be designed to achieve any desired tar-
38 get glass transition temperature by incorporating plasticizers (reduce Tg) or high-molecular
39 weight polymers (increase Tg). It should be cautioned that although glass transition and
40 associated microcapsule stability are clearly related to the material properties of the matrix
41 and rates of crystallization, there is growing evidence that in the glass transition region
42 small molecules are more mobile than might be expected from the high viscosity of the
43 matrix (Parker and Ring, 1995). Mechanism of degradation of molecules entrapped in a
44 glassy matrix is not fully understood but is speculated to be due to side-chain flexibility
45 (e.g. enzymes) and/or diffusion of small molecules such as water and oxygen through the
46S glassy matrix. Other deteriorative mechanisms may include Maillard reaction between the
47N active and the carrier matrix.
Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 5

Introduction 5

Active (powder, 1
Sugar blends dispersion, 2
(Dry feed) emulsion) 3
4
5
6
Amorphous 7
rope
8
EXTRUDER
9
10
Mixing & heating
Ground
11
microparticles 12
13
Figure 1.2. Encapsulation into amorphous carbohydrate matrices using hot melt extrusion. 14
15
16
Microcapsules manufactured via spray drying and extrusion may show structural imper- 17
fections, thus limiting their shelf life. While spray-dried microcapsules tend to have low 18
bulk density, extruded granules may show stickiness and clumping. In addition, the pres- 19
ence of exposed active on the microparticle surface may have detrimental consequences 20
such as drifts in the release profile and/or loss of active due to oxidation and other deterio- 21
rative processes. 22
A limited number of applications have employed freeze drying or other evaporative 23
techniques to form carbohydrate glasses from solution. Here, the removal of water mole- 24
cules takes place either by freezing the solution and subliming the ice as in freeze drying 25
or by evaporation. Freeze drying forms porous substrates due to transport of water vapor. 26
Unlike starches, sugars lack fixed molecular structure; thus they collapse upon freeze 27
drying. 28
Co-crystallization with sugars has been practiced in few unique situations but has 29
not found any commercial success. Crystalline sucrose is a poor flavor carrier but co- 30
crystallization with flavors forms aggregates of very small crystals that incorporate the fla- 31
vors either by inclusion within the crystals or by entrapment between them. 32
Release of actives from amorphous carbohydrate matrices takes place by subjecting the 33
matrix to moisture or high temperatures, that is, by bringing the matrix to a state above its 34
glass transition temperature. Microcapsules entrapped in amorphous structures are pre- 35
ferred for their ease of manufacturing, scalability and economics compared to other encap- 36
sulation technologies. Their usage has been adapted to a variety of food systems such as 37
surface sprinkle on breakfast cereals, hot instant drinks, soups, tea bags, chewing gum, 38
pressed tablets, and so on. 39
40
41
Complexation of Actives into Cyclodextrins 42
Entrapment of actives into cyclodextrins is a unique approach to microencapsulation that is 43
based on molecular selectivity. Cyclodextrins are cyclic oligosaccharides formed of vari- 44
ous numbers of α-(1,4) linked pyranose subunits. The 6-, 7-, and 8-numbered cyclic struc- 45
tures are referred to as α-, β-, and γ-cyclodextrins, respectively; these molecules vary in S46
their solubility, cavity size, and complexation properties (Table 1.1). N47
Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 6

6 Chapter 1

1 Table 1.1. Selected physicochemical properties of cyclodextrins (adapted from Martin Del Valle
2 2004)
3 Attribute -Cyclodextrin -Cyclodextrin -Cyclodextrin
4
5 Number of glucopyranose units 6 7 8
Molecular weight (g/mol) 972 1135 1297
6 Solubility in water at 25°C (% w/v) 14.5 1.85 23.2
7 Cavity diameter (Å) 4.7–5.3 6.0–6.5 7.5–8.3
8 Cavity volume (Å)3 174 262 427
9
10
Type and degree of complexation in cyclodextrins are determined by two main factors:
11
(1) steric fit of the guest (encapsulant) to the host (cyclodextrin) and (2) their thermody-
12
namic interactions, mainly hydrophobic type.
13
Generally, one guest molecule is included in one cyclodextrin molecule, although for
14
some molecules with low molecular weight, more than one guest molecule may fit into the
15
cavity (Figure 1.3). For molecules with large hydrodynamic radii, more than one cyclodex-
16
trin molecule may bind to the guest. In principle, only a portion of the molecule must fit
17
into the cavity to form a complex. As a result, one-to-one molar ratios are not always
18
achieved, especially with high- or low-molecular-weight guests.
19
Guest molecules in cyclodextrins are not permanently entrapped but occur in a dynamic
20
equilibrium. However, once a complex is formed and dried, it is very stable and often
21
results in very long shelf life (up to years at ambient temperatures under dry conditions).
22
Release of the complexed guest takes place by immersing the guest-host complex in aque-
23
ous media to dissolve the complex and further promoting the release of the guest when dis-
24
placed by water molecules.
25
A wide variety of molecules can be entrapped in cyclodextrins such as fats, flavors, col-
26
ors, and so on (Martin Del Valle, 2004; Parrish, 1988). Complexation of cyclodextrins with
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S
47N 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 7

Introduction 7

sweetening agents such as aspartame can also stabilize the molecule and improve its taste 1
as well as eliminate the bitter aftertaste of other sweeteners such as stevioside and gly- 2
cyrrhizin. Cyclodextrins can entrap undesirable substances such as cholesterol from prod- 3
ucts such as milk, butter, and eggs (Szetjli, 1998; Hedges, 1998). 4
5
6
Encapsulation in Microporous Matrices—Physical Adsorption 7
8
Physical adsorption can only be feasible when an active is adsorbed onto a large surface 9
area, microporous substrate, commonly referred to as molecular sieve. Examples of this 10
category include activated carbon (500–1400 m2/g) and amorphous silica (100–1000 m2/g) 11
(Cheremisinoff and Morresi, 1978). Despite their efficiency in entrapping volatiles, silica 12
and activated carbon usage in foods has been discouraged due to regulatory constraints and 13
is currently limited to packaging applications. The effectiveness of these materials is 14
demonstrated by extensive reduction in equilibrium vapor pressure which accompanies 15
physical adsorption of volatile flavors. 16
Micronized sugars have been used but with limited success in adsorption applications. 17
Dipping capillary-sized droplets of sucrose or lactose solution into liquid nitrogen followed 18
by freeze drying can produce amorphous spheres that have the ability to adsorb aromas. 19
Sorption of vapor causes these materials to revert to the more stable crystalline state with 20
accompanying loss of porosity. 21
22
23
Encapsulation in Fat- or Wax-Based Matrices 24
Entrapment of functional actives in fat-based matrices can be achieved using two main tech- 25
nologies, hot-melt fluid bed coating and spray congealing. Actives can best be entrapped via 26
mixing them with a fat/wax carrier followed by spray congealing. These technologies have 27
been adequately discussed in Chapter 5 which deals with the encapsulation of bakery 28
leavening agents. 29
30
31
32
Encapsulation in Emulsions and Micellar Systems
33
Encapsulation via micelles is a convenient approach to enhance the solubility of insoluble 34
or slightly soluble actives. This technique involves the simple entrapment of a hydrophobic 35
active in a hydrophilic shell material, thus rendering the particle or droplet soluble in aque- 36
ous media. This is no trivial matter when considering the problems with bioavailability of 37
hydrophobic drugs and nutritional actives (fat-soluble vitamins, fish oil, and a host of 38
water-insoluble drug actives). 39
A second important function of micelles is their small size which allows them to evade 40
the body’s screening mechanism, the reticuloendothelial system (RES). Recognition by 41
RES is the main reason for removal of many drug delivery vehicles from the blood before 42
reaching their target site (Sagalowicz et al., 2006). 43
Micelles serve as drug “reservoirs” or “microcontainers” that ultimately release drugs 44
via diffusional processes. An in-depth discussion on encapsulation into emulsion systems 45
can be found in Chapters 2 and 3 of this book by Professor Garti and Dr. Zuidam and their S46
respective coworkers. N47
Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 8

8 Chapter 1

1 Encapsulation in Cross-Linked or Coacervated Polymers


2
3 Coacervation, as defined by Speiser (1976), is a process of transferring macromolecules
4 with film properties from a solvated state via an intermediate phase, the coacervation
5 phase, into a phase in which a film is formed around each particle and then to a final phase
6 in which this film is solidified or hardened. Two types of coacervation processes are com-
7 monly used in encapsulation applications, namely simple and complex:
8
9 1. Simple coacervation is based on “salting out” of one polymer by addition of agents
10 (salts, alcohols) that have higher affinity to water than the polymer. It is essentially a
11 dehydration process whereby separation of the liquid phase results in the solid particles
12 or oil droplets becoming coated and eventually hardened into microcapsules.
13 2. Complex coacervation, on the other hand, is a process whereby a polyelectrolyte com-
14 plex is formed. It requires the mixing of two colloids at a pH at which one is negatively
15 charged and the other positively charged, leading to phase separation and formation of
16 enclosed solid particles or liquid droplets (Rabiskova and Valaskova, 1998).
17
18 Several parameters can impact the formation and integrity of coacervates such as the poly-
19 mers’ molecular weight, their w/w ratios, temperature, and processing time. Core materials
20 suitable for coacervation are solids and liquids that are water-insoluble so that the active
21 would not dissolve in the aqueous phase. One of the approaches to achieving high oil pay-
22 loads is by using hydrophobic surfactants (Rabiskova and Valeskova, 1998).
23 The release of actives from coacervated systems is primarily a function of the wall type
24 and its thickness (slower release with increased wall thickness). Chapter 7 of this book
25 presents an in-depth discussion on coacervation for flavor encapsulation applications.
26
27
Encapsulation into Hydrogel Matrices
28
29 Hydrogels are hydrophilic, three-dimensional network gels that can absorb much more
30 water than their own weight. Hydrogels consist of (a) polymers, (b) molecular linkers or
31 spacers, and (c) an aqueous solution. Basic high-molecular-weight polymers include poly-
32 saccharides, proteins, chitin, chitosans, hydrophilic polymers, and so on (Shahidi et al.,
33 2006). The affinity of hydrogels to aqueous media makes them ideal absorbing matrices for
34 food and agricultural actives.
35 The principle of encapsulation by hydrogels is simply to entrap an active substance
36 and to further release it via gel-phase changes in response to external stimuli. Grahm and
37 Mao (1996) categorized the types of materials that cannot be delivered via hydrogels as:
38 (i) extremely water-soluble actives due to the risk of uncontrollable quick release and
39 (ii) very high-molecular-weight substances due to the extremely slow release rate to
40 achieve a desired benefit.
41 Release of actives from hydrogels takes place via diffusion. The latter can be impacted
42 by various chemical and physical factors such as the prevailing chemical bonds (H-bonds,
43 ionic bonds, electrostatic interactions, and hydrophobic interactions) between the active
44 and the matrix. Physical factors include molecular size and conformation. Controlling
45 (extending) the release of an active in a hydrogel matrix can be achieved by decreasing the
46S hydrophilicity and/or diffusivity of the hydrogel structure or by covalently linking the
47N active to the carrier hydrogel matrix.
Lakkis_Ch_01_001-012 r1.qxd 3/30/07 6:23 PM Page 9

Introduction 9

Ideal hydrogels display a sharp phase transition upon swelling in an aqueous solvent 1
in response to environmental stimuli such as temperature, pH, electric field, and so on. 2
Release from hydrogels can be predicted from their LCT (lower critical solution tempera- 3
tures) values. As temperature increases to the hydrogel’s LCT, the hydrogel shrinks due to 4
dehydration. Below LCT, hydrogels can take up water thus increasing their swelling 5
(Ichikawa et al., 1996). 6
7
Overview of Release Mechanisms 8
9
Despite the far-reaching applications of encapsulation and controlled-release technologies 10
in many industries, predicting the release of encapsulated actives, especially in biological 11
systems (foods included), remains a challenge. In the human gastrointestinal tract (GIT), 12
for example, the release of microcapsules is a function of the physiological conditions, 13
presence of food as well as the physicochemical properties of the ingested dosage. 14
One of the essential requirements for predicting release mechanisms of microencapsu- 15
lated dosages is by identifying parameters involved in mass transport and diffusion of the 16
actives from a region of high concentration (dosage) to a region of low concentration in the 17
surrounding environment. 18
Encapsulation and controlled-release systems can be classified into two main types: 19
reservoir and matrix systems and, in some cases, combinations of both. 20
21
Reservoir-Type Systems 22
23
Reservoir-type systems are simply described as delivery devices where an inert membrane
24
surrounds an active agent which upon activation diffuses through the membrane at a finite
25
controllable rate (Figure 1.4a). Reservoir-type systems are capable of achieving zero-order
26
rates provided that constant thermodynamic activity is maintained inside the coating
27
material. Reservoir-type systems are subject to shifts to a “burst-like” mechanism due to
28
minor flaws in the membrane integrity.
29
30
Matrix Systems 31
32
Matrix or monolithic delivery systems can best be represented by microparticles prepared
33
by extrusion or fat-congealed capsules where the actives are dispersed in the encapsulating
34
35
36
37
(a) (b) (c)
Reservoir-type Matrix-type Combination-type
38
device device device 39
40
41
42
43
44
45
Figure 1.4. Schematic representation of encapsulation systems: (a) reservoir-type, (b) matrix-type,
and (c) combination-type.
S46
N47
Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 10

10 Chapter 1

1 medium (carbohydrate, fat, or other matrices). Matrix systems can be swellable (hydrogel)
2 or non-swellable. Compared to reservoir systems, matrix systems require less quality con-
3 trol, hence lower manufacturing cost (Figure 1.4b).
4
5
6 Combination Release Mechanism
7
8 Examples of this category can best be illustrated by congealed microcapsules or extruded
9 microparticles with additional film. coating (enrobing). This technique is most useful for
10 manufacturing extremely “delayed release” profiles (Figure 1.4c).
11
12
13 Burst Release Mechanism
14
Burst release is simply described by a high initial delivery of an entrapped active, before
15
the release reaches a stable profile, thus reducing the system’s effective lifetime and com-
16
plicating the release control. Although burst release may be preferred for flavor high-
17
impact applications, in drugs this mechanism may lead to high toxicity levels and
18
ineffective administration of the active.
19
Burst release can most often take place in reservoir and hydrogel systems, though it can
20
still take place in matrix designs. Reasons for this range from cracks in the protective cap-
21
sule shell to storage effect where the membrane becomes saturated with the active sub-
22
stances or due to very high active loading. When placed in a release medium, the active can
23
quickly diffuse out of the membrane surface causing a burst effect (Huang and Brazel,
24
2001). Low-molecular-weight actives frequently undergo burst release, a result of high
25
osmotic pressure and increased concentration gradient. Other reasons include: processing
26
conditions, surface characteristics of host material, sample geometry, host/drug interac-
27
tions, morphology, and porous structure of dry material.
28
Application of a coating material over a monolithic microparticle can help eliminate
29
burst release, though might change the release profile. Other treatments include washing
30
microparticles to extract surface droplets of actives.
31
32
33
34 First-order
35
Brust
36
release
37 Zero-order
38
39
40
41
42
43
44
45
46S
47N 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 11

Introduction 11

Kinetically, two main release patterns are identified, zero-order and first-order (Figure 1
1.5). Other rates can still occur: 2
3
Zero-order release equation –dA/dt = k 4
First-order release equation –dA/dt = k[C] 5
6
where –dA/dt is the change in active concentration over time, k is the rate constant, and [C] 7
is the active’s concentration. 8
In designing microcapsules with controlled-release systems, it is critical to identify 9
desirable release profile so that adequate materials and technology can be chosen. 10
11
12
References
13
Baker, R.W. and Lonsdale, H.K. 1974. Controlled release: mechanisms and rates. In: Controlled Release of 14
Biologically Active Agents (A.C. Tanquary and R.E. Lacey, eds.), Plenum, New York, pp. 15–71. 15
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.
16
Conde-Petit, B., Escher, F. and Nuessli, J. 2006. Structural features of starch-flavor complexation in food model 17
systems. Trends in Food Science & Technology 17(5): 227–235. 18
Grahm, N.B. and Mao, J. 1996. Controlled drug release using hydrogels based on poly(ethylene glycols): macro- 19
gels and microgels, pp. 52–64. In: Chemical aspects of Drug Delivery, Karsa, D. and Stephenson, R. (Eds). 20
Royal Society of Chemistry.
Hedges, R.A. 1998. Industrial applications of cyclodextrins. Chem. Rev. 98: 2035–2044.
21
Huang, X. and Brazel, C.S. (2001). On the importance and mechanisms of burst release I matrix-controlled drug 22
delivery systems. J. Controlled Release 73: 121–136. 23
Ichikawa, H., Kaneko, S. and Fukumori, Y. 1996. Coating performance of aqueous composite lattices with 24
N-ispropylacrylamide shell and thermosensitive permeation properties of their microcapsule membrane. Chem. 25
Pharm. Bull. 44(2): 383–391.
Larbousse, S., Roos, Y. and Karel, M. 1992. Collapse and crystallization in amorphous matrices with encapsulated
26
compounds. Sci. Aliments 12: 757–769. 27
Martin Del Valle, E.M. 2004. Cyclodextrins and their uses: a review. Process Biochem. 39: 1033–1046. 28
Masters, K. 1979. Spray Drying Handbook, 3rd ed., George Godwinn, London. 29
Parrish, M.A. 1988. Cyclodextrins—A Review. England: Sterling Organics. Newcastle-upon-Tyne NE3 3TT. 30
Parker, R. and Ring, S.G. 1995. Diffusion in maltose-water mixtures at temperatures close to the glass transition.
Carbohydr. Res. 273: 147–155.
31
Rabiskova, M. and Valaskova, J. 1998. The influence of HLB on the encapsulation of oils by complex coacerva- 32
tion. J. Microencapsul. 15(6): 747–751. 33
Sagalowicz, L., Leser, M.E., Watzke, H.J. and Michel, M. 2006. Monoglyceride self-assembly structures as deliv- 34
ery vehicles. Trends in Food Science & Technology 17(5): 204–214. 35
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.
36
Shimada, Y., Roos, Y. and Karel, M. 1991. Oxidation of methyl linoleate encapsulated in amorphous lactose-based 37
food model. J. Agric. Food Chem. 39: 637–641. 38
Speiser, P. 1976. Microencapsulation by coacervation, spray encapsulation and nanoencapsulation. In: Microen- 39
capsulation, Nixon, J.R. (Ed.), pp. 1–11. 40
Szetjli, J. 1998. Introduction and general overview of cyclodextrin chemistry. Chem. Rev. 98: 1743–1753.
41
42
43
44
45
S46
N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:35 PM Page 13

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

2 Improved Solubilization and Bioavailability 1


2
of Nutraceuticals in Nanosized 3
Self-Assembled Liquid Vehicles 4
5
6
7
Nissim Garti, Eli Pinthus, Abraham Aserin, and 8
Aviram Spernath 9
10
11
12
Introduction 13
14
Microemulsions have been known for decades to the scientific community and to experts in 15
the industry. Hundreds of studies have been carried out by experimentalists and many theo- 16
ries have been worked out regarding the self-aggregation of surfactants in aqueous phase as 17
well as in oil phase, to form micellar or reverse micellar (respectively) structures. The 18
micellar phases can be swollen by another liquid phase to form a reservoir of insoluble liq- 19
uid phase entrapped by a tightly packed surfactant layer known as water-in-oil (w/o) or oil- 20
in-water (o/w) microemulsions. 21
Microemulsion, by the most common general definition, is a “structured fluid” (or 22
solution-like mixture) of two immiscible liquid phases in the presence of a surfactant 23
(sometimes with cosurfactant and cosolvent), which spontaneously form a thermodynami- 24
cally stable isotropic solution-like liquid. 25
In spite of the numerous studies and pronounced potential applications in foods, pharma- 26
ceuticals, and cosmetics, only a few practical preparations, in which the solubilized molecules 27
are at very low solubilization levels, are presently available in the market place. It is always 28
an open question as to why these structures did not make their way to final products. 29
The self-assembled nanosized surfactants and oil can solubilize another liquid immisci- 30
ble phase and/or guest molecules (solubilizates). Droplet sizes are in the range of a few up 31
to a hundred nanometers. In theory, in order to form such nanostructures, it is essential to 32
reduce the interfacial tension between the two phases to a value close to zero. In order to do 33
so, surfactants with the proper hydrophilicity must be utilized. In addition, surfactants must 34
have the proper geometry to self-organize in curved structures with the proper critical 35
packing parameters (CPP). 36
Microemulsions are best studied by constructing binary, ternary, or multicomponent 37
phase diagrams, which represent the equilibrium situation of the component mixture or the 38
thermodynamic organization of the components. A typical classical phase diagram is 39
shown in Figure 2.1. 40
Understanding the phase behavior and microstructure of microemulsions is an important 41
fundamental aspect of the utilization of these structured fluids in industrial applications. 42
Today, we have a more profound understanding of the phase behavior and microstructure of 43
microemulsions (Shinoda and Lindman, 1987; Billman and Kaler, 1991; Kahlweit et al., 44
1996; Regev et al., 1996; Solans et al., 1997; Ezrahi et al., 1999). However, industrial appli- 45
cations of microemulsions are rarely simple ternary systems, but more often complicated S46
multicomponent systems. It is not always clear whether, in the complex systems, droplet N47

13
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 14

14 Chapter 2

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
Figure 2.1. Typical phase diagram made with water, emulsifiers, and oil phase. Four types of
25
isotropic regions have been identified. Note that the dilution lines traverse via a two-phase
26 region and full dilution to the far corner of the water phase is not possible.
27
28
29
30 sizes and shapes are similar and remain intact and the role of the different components in
31 stabilizing the interface. Systematic investigations should be carried out to understand the
32 microstructure and the effect of the different components on the system.
33 In recent years, few attempts have been made to formulate and characterize microemul-
34 sions that can be used for food, cosmetic, and pharmaceutical purposes (Dungan, 1997;
35 Gasco, 1997).
36 In this effort, oils acceptable in food industry have replaced normal alkanes. The major-
37 ity of easily made preparations were of oil-continuous phase (w/o). The authors focused on
38 studying the ability of formulating a microemulsion with triglycerides (Alander and Warn-
39 heim, 1989a, b; Malcolmson and Lawrence, 1995; von Corswant et al., 1997; von Corswant
40 and Söderman, 1998; Warisnoicharoen et al., 2000) and perfumes (Hamdan et al., 1995;
41 Tokuoka et al., 1995; Kanei et al., 1999) as the oil component. Some workers (Joubran
42 et al., 1993; Trevino et al., 1998) have studied the phase behavior and microstructure of
43 water-in-triglyceride (w/o) microemulsions based on polyoxyethylene sorbitan hexaoleate.
44 They found that the monophasic area of these systems was strongly dependent on tempera-
45 ture and aqueous phase content. In other studies, o/w microemulsions were used. Lawrence
46S and coworkers (Malcolmson and Lawrence, 1995; Warisnoicharoen et al., 2000) examined
47N the solubilization of a range of triglycerides and ethyl esters in an o/w microemulsion system
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 15

Improved Solubilization and Bioavailability of Nutraceuticals 15

with nonionic surfactants. They concluded that the solubilization capacity depends not only 1
on the nature of the surfactants but also on the nature of the oil. 2
There are very few surfactants that can be used in food formulations. In this respect, 3
polysorbates (Tweens, ethoxylated derivatives of sorbitan esters) and sugar esters are inter- 4
esting families of surfactants. The substitution of the hydroxyl groups on the sorbitan ring 5
with bulky polyoxyethylene groups increases the hydrophilicity of the surfactant. Similarly, 6
monoesterification of sucrose forms hydrophilic emulsifiers. The ability of Tweens to form 7
microemulsions for food applications has been studied by several authors (Constantinides 8
and Scalart, 1997; Trotta et al., 1997; Park and Kim, 1999; Prichanont et al., 2000; Radomska 9
and Dobrucki, 2000). An increased solubility of lipophilic drugs in the microemulsion 10
region was observed and explained by the penetration of these drugs into the interfacial 11
film (Trotta et al., 1997; Park and Kim, 1999; Radomska and Dobrucki, 2000). 12
Even though some food-grade emulsifiers have been mentioned as possible microemul- 13
sion-forming amphiphiles, it was almost impossible to use these systems mainly because 14
the concentrates of oil/surfactant mixtures could not be fully diluted with water or aqueous 15
phases to form o/w microemulsions. Any such dilution line (composition) is always “cross- 16
ing” the two-phase region, resulting in a fast destabilization process and formation of emul- 17
sions or two phases. Such phase separation leads to rapid precipitation of the solubilized 18
matter. Some examples of such discontinued dilution lines illustrate the dilution problem of 19
the classical phase diagrams. In Figure 2.1, these dilution lines are marked as dashed lines. 20
In most studies, the emphasis was on attempts to add just one immiscible liquid such as 21
water (or oil) to the oil (or water)-continuous surfactant phase, that is, to solubilize the oil 22
in the core (inner phase) of the micelles. Practically very few attempts were made to incor- 23
porate additional guest molecules, such as vitamins, aromas, antioxidants, and bioactive 24
molecules, into the solubilized core. Very little has been done to solubilize nutraceuticals 25
within nanosized liquid vehicles in order to provide some pronounced health benefits to 26
humans or to treat chronic diseases. 27
Many structural and compositional limitations, in the presently available food formula- 28
tions, did not permit loading significant amounts of nutraceuticals. It is not an easy task to 29
accomplish, since there is a need for additional technology to be developed. It is essential to 30
introduce new ingredients, new surfactants, and new concepts in microemulsion prepara- 31
tion. Some of the cardinal points to be solved include the following: 32
33
• Progressively and continuously diluting, by aqueous phase or water, without destroying 34
the interface and forming two-phase regions, that is, forming the so-called U-type phase dia- 35
grams that undergo progressive inversion from w/o to o/w microemulsions (Figure 2.2). 36
• Preparing microemulsions that will be based on the use of permitted food-grade emulsi- 37
fiers, oils, cosurfactants, or cosolvents. 38
• Facilitating the entrapment (cosolubilization capacity) of large loads of insoluble guest 39
molecules within the core of the microemulsion or at its interface. 40
• Providing environmental protection of the active addenda (guest molecules) from 41
autooxidation or hydrolytic degradation during shelf storage. 42
• Improving the bioavailability of the entrapped addenda. 43
• Controlling the release from the vehicle to the water-continuous phase or onto human 44
membranes. 45
• Using microemulsions as microreactors to obtain regioselectivity, fast kinetics, and con- S46
trolled and triggered reactions of active molecules once applied on the skin. N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 16

16 Chapter 2

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
Figure 2.2. Typical novel U-type phase diagram composed of selected combinations of
18
cosmetic-grade emulsifiers with progressive full dilution.
19
20
21 A phase diagram with a very large isotropic one-phase region is typical of the novel
22 microemulsions that are made from multicomponents. The isotropic regions represent w/o,
23 bicontinuous mesophase, and o/w microemulsion structures. The phase diagrams are
24 known as U-type. In such compositions, within the isotropic regions of the phase diagram,
25 the oil/surfactant condensed structured mixtures (denoted condensed reverse micelles, L2)
26 can transform to an L1 phase (direct micelles) via a w/o microemulsion, bicontinuous
27 mesophase, and o/w microemulsion regions progressively, without any phase separation.
28 To the best of our knowledge, no reports were available in the literature, prior to the
29 establishment of our formulations as part of the extended new U-type phase diagrams, to
30 comply with these prerequisites of dilutable large isotropic regions (Garti et al., 2001,
31 2003, 2004a, b; Yaghmur et al., 2002a, b, c, 2003a, b, 2004, 2005; Spernath et al., 2002,
32 2003; de Campo et al., 2004). Most of the early studies were conducted on systems with
33 constant water content (>70%), low oil content (ca. 5–10%), and large surfactant excess
34 (high surfactant/oil ratios). We enlarged the scope of the understanding and use of such
35 microemulsions to food and cosmetic preparations. Our studies examined various aspects
36 of solubilization of nutraceuticals, release patterns, and other thermal and environmental
37 conditions. In some of our studies the role of the surfactant was examined. The maximum
38 solubilization load was determined, and efforts were made to estimate the total amounts of
39 active matter that can be entrapped along any dilution line. We were the first to establish the
40 correlation between maximum solubilization capacity and water dilution (Garti et al., 2001,
41 2003, 2004; Spernath et al., 2002, 2003; Yaghmur et al., 2002a, b, c , 2003a, b, 2004, 2005;
42 de Campo et al., 2004).
43 This review summarizes our efforts to develop modified microemulsions as nanosized
44 self-assembled liquid (NSSL) vehicles for the solubilization of nutraceuticals and to
45 improve transmembrane transport for additional health benefits. Attempts were made to
46S achieve solubilization of nonsoluble active ingredients such as aromas and antioxidants
47N into clear beverages that are based on water-continuous phase.
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 17

Improved Solubilization and Bioavailability of Nutraceuticals 17

U-Type Microemulsions, Swollen Micelles, and Progressive 1


and Full Dilution 2
3
Initially we (Garti et al., 2001; Yaghmur et al., 2002a, b) dealt with solubilization of water 4
and oil in the presence of a new set of nonionic ingredients and emulsifiers to form U-type 5
nonionic w/o and o/w food microemulsion systems. It was recognized that certain mole- 6
cules destabilize the liquid crystalline phases and extend the isotropic region to higher sur- 7
factant concentrations. The ability of these additives to provide large monophasic systems 8
(denoted as the AT region in Figure 2.2), in which the total amounts of solubilized oil and 9
water should be as high as possible, was studied. The pseudoternary phase diagrams for 10
R(+)-limonene-based systems with food-grade systems were compared with those based 11
on non-food grade emulsifiers such as Brij 96v, (C18:1(EO)10, Figure 2.2) (Garti et al., 2001; 12
Yaghmur et al., 2002b). These systems offer great potential in practical formulations. We 13
followed the structural evolution and transformation of the microemulsion system from 14
aqueous phase-poor to aqueous phase-rich regions without encountering phase separation. 15
Figure 2.3a demonstrates the size distribution of various droplets along dilution line 73 16
(D73; 70 wt% surfactant and 30 wt% oil phase) from 10 to about 90 wt% water. It can be 17
seen that the droplets in the w/o region are smaller than those at higher water content upon 18
inversion to o/w microemulsions. Figure 2.3b represents a typical structure as seen in the 19
cryo-TEM (transmission electron microscopy) photomicrographs of an o/w microemulsion 20
taken from the rich-in-water region of the U-type diagram (obtained after inversion from an 21
L2 phase into o/w droplets upon dilution with aqueous phase to 90 wt% water). The droplet 22
sizes are ca. 8–10 nm and are mostly monodispersed. It should be noted that most 23
microemulsions, regardless of the type of oil, type of surfactant, and cosolvents, consist 24
of droplets of ca. 5–20 nm in size and do not grow above these sizes at any water or oil 25
contents. 26
Various U-type phase diagrams with different types of hydrophilic surfactants, various 27
cosolvents, and cosurfactants were constructed to form small or large isotropic AT regions. 28
The most desirable phase diagram yielded an isotropic region of AT > 75% from the total 29
area of the phase diagram. The dilution lines connecting the oil/surfactant axis with the 30
water corner were termed Wm lines. Full dilution lines are those that can undergo full and 31
progressive dilution to the far water corner (Wm = 100%). Wm = 50% means that samples 32
can be diluted only up to 50 wt% water and if more water is added the microemulsion will 33
undergo phase separation. An example of Wm = 100% dilution line is line 64 in Figure 2.2, 34
in which a mixture of 60 wt% surfactant phase and 40 wt% oil phase is diluted progres- 35
sively and completely with aqueous phase to the far corner (Wm = 100%) aqueous phase. In 36
dilution line 55 (50 wt% surfactant phase and 50 wt% oil phase), the Wm is of ca. 60% 37
aqueous phase, and further dilution will lead to phase separation. 38
Construction of U-type phase diagrams is essential for formulations of water-dilutable 39
microemulsions. 40
41
42
Solubilization of Nonsoluble Nutraceuticals
43
The growing interest in microemulsions as vehicles for food and cosmetic formulations 44
arises mainly from the advantages of their physicochemical properties. Microemulsions 45
can cosolubilize large amounts of lipophilic and hydrophilic nutraceutical and cosmetoceu- S46
tical additives, together with the inner reservoir. N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 18

18 Chapter 2

1 (a) Normalized to 1 at the maximum


10% AP
2
1 30% AP
3
4 40% AP
5 50% AP
6 60% AP
p(r ) [a.u.]

7 70% AP
8 80% AP
9 90% AP
10
11
12 0
13
14 0 2 4 6 8 10 12 14 16 18
15 r [nm]
16 (b)
17
18
19
20
21
22
23
24
25
26
27
28
29
Figure 2.3. (a) Droplet size distribution of various dilution points along dilution line 73 in
30 phase diagram depicted in Figure 2.2. (b) Photomicrograph of typical o/w droplets derived from
31 a concentrate of w/o after dilution to 90 wt% water content (AP refers to aqueous phase).
32 (Adapted from Garti, with permission from the publisher.)
33
34
35
36 The cosolubilization effect has attracted the attention of scientists and technologists
37 for more than two decades. Oil-in-water microemulsions loaded with active molecules
38 opened new prospective opportunities for enhancing the solubility of hydrophobic vita-
39 mins, antioxidants, and other skin nutrients. This is of particular interest, as it can provide a
40 well-controlled way for incorporating active ingredients and may protect the solubilized
41 components from undesired degradation reactions (Garti et al., 2001; Spernath et al., 2002;
42 Yaghmur et al., 2002a, b, c). Figure 2.4 is a schematic illustration of the loading process of
43 various nutraceuticals onto the o/w microemulsion droplets after inversion.
44 Solubilization of active addenda may, therefore, be defined as spontaneous molecular
45 entrapment of an immiscible substance (or only slightly miscible or soluble) in self-
46S assembled surfactant mixtures to form a thermodynamically stable, isotropic, structured
47N solution, consisting of nanosized liquid structures.
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 19

Improved Solubilization and Bioavailability of Nutraceuticals 19

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
Figure 2.4. A schematic illustration of the loading process of various nutraceuticals onto the 22
o/w microemulsion droplets after inversion. (Adapted from Nutralease and Garti, 2003, with 23
permission from the publisher.) 24
25
26
27
The solubilized active molecules are compounds with nutritional value to human health
28
that, in most cases, are used in food applications. We will mention a few such examples that
29
were studied in our labs, such as lycopene, phytosterols, lutein, tocopherols, CoQ10, and
30
essential oils.
31
32
33
Lycopene
34
Food supplements have become very prominent compounds in recent years, due to 35
increased public awareness of healthy nutrition. The possibility of enhancing the solubility 36
of lipophilic vitamins, essential oils, aromas, flavors, and other nutrients in o/w microemul- 37
sions is of great interest, as it can provide a well-controlled method for the incorporation 38
of active ingredients and may protect the solubilized components from undesired degradation 39
reactions (Dungan, 1997; Holmberg, 1998; Garti et al., 2000a, b). Lycopene (Figure 2.5) is an 40
important carotenoid that imparts a characteristic red color to tomatoes. This lipophilic 41
compound is insoluble in water and in most food-grade oils. For example, lycopene solubil- 42
ity in one of the most efficient edible essential oils, R(+)-limonene, is 700 ppm. Recent 43
studies have indicated the important role of lycopene in reducing risk factors of chronic 44
diseases such as cancer, coronary heart disease, and premature aging (Dungan, 1997; 45
Holmberg, 1998). This, in turn, has led to the idea of studying the effect of lycopene uptake S46
on human health. N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 20

20 Chapter 2

1
2
3
4
5
6
7 Figure 2.5. Molecular structure of lycopene.
8
9
10 Bioavailability of lycopene is affected by several factors:
11
12 • Food matrix containing the lycopene and, as a result, intracellular location of the
13 lycopene, and the intactness of the cellular matrix. Tomatoes converted into tomato paste
14 can enhance the bioavailability of lycopene, as the processing includes mechanical
15 particle size reduction and heat treatment.
16 • Amount and type of dietary fat present in the intestine. The presence of fat affects the for-
17 mation of the micelles that incorporate the free lycopene.
18 • Interactions between carotenoids that may reduce absorption of either one of the
19 carotenoids (Bramley, 2000) owing to competitive absorption between the carotenoids.
20 On the other hand, simultaneous ingestion of various carotenoids may induce antioxidant
21 activity in the intestinal tract, and thus result in increased absorption of the carotenoids
22 (Rao and Agrawal, 1999; Bramley, 2000).
23 • Molecular configuration (cis/trans) of the lycopene molecules. The bioavailability of the
24 cis isomer is higher than the bioavailability of the trans isomer. This may result from the
25 greater solubility of cis isomers in mixed micelles and lower tendency of cis isomers to
26 aggregate (Cooke, 1998; Rao and Agrawal, 1999).
27 • Decrease in particle size (Van het Hof et al., 2000).
28
29 Care must be taken in formulating lycopene as an additive in food systems, since the large
30 number of conjugated bonds in this carotenoid causes instability when exposed to light or
31 oxygen. We explored the ability of U-type microemulsions to solubilize lycopene and have
32 also investigated the influence of solubilized lycopene on the microemulsion microstruc-
33 ture. Phase diagrams have been constructed, lycopene has been solubilized, and several
34 structural methods have been utilized including self-diffusion nuclear magnetic resonance
35 (SD-NMR) spectroscopy. This advanced analytical technology was further developed to
36 determine the microemulsion microstructure at any dilution point.
37 The influence of microemulsion composition on the solubilization of lycopene in a five-
38 component system consisting of R(+)-limonene, cosurfactant, water, cosolvent, and poly-
39 oxyethylene (20) sorbitan mono-fatty esters (Tweens) is presented in Figures 2.6 and 2.7.
40 Solubilization capacity was defined (Spernath et al., 2002, 2003) as the quantity of
41 lycopene solubilized in the microemulsion. Figure 2.7 shows the solubilization capacity of
42 lycopene along water dilution line T64 (at this line the constant ratio of R(+)-limonene/
43 ethanol/Tween 60 is 1/1/3, respectively). Four different regions can be identified along this
44 dilution line. At 0–20 wt% aqueous phase (region Ι), the solubilization capacity of
45 lycopene decreases dramatically, from 500 to 190 ppm (reduction of 62%). This dramatic
46S decrease in the solubilization capacity can be associated with the increase in interactions
47N between the surfactant and water molecules. Water can also strongly bind to the hydroxyl
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 21

Improved Solubilization and Bioavailability of Nutraceuticals 21

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
Figure 2.6. Pseudoternary phase diagram (25ºC) of water/PG/R()-limonene/ethanol/Tween 60 16
system with a constant weight ratio of water/PG (1:1) and a constant weight ratio of R()- 17
limonene/ethanol (1:1). Solubilization of lycopene was studied along dilution line T64. (Adapted 18
from Yaghmur and Garti, 2001, with permission from the publisher.) 19
20
21
22
23
24
25
26
27
28
29
30
31
32
Figure 2.7. Solubilization capacity of lycopene along dilution line T64 as per phase diagram in 33
Figure 2.6. (Adapted from Garti, with permission from the publisher.) 34
35
36
37
groups of the surfactant at the interface. When water is introduced to the core, the micelles 38
swell, and more surfactant and co-surfactant participate at the interface, replacing the 39
lycopene, thus decreasing its solubilization. In region Ι, the reverse micelles swell gradually 40
and become more hydrophobic, causing less free available volume for the solubilized 41
lipophilic lycopene and a reduction in its solubilization capacity. At 20–50 wt% aqueous 42
phase (region II) the solubilization capacity remains almost unchanged (decreases only by 43
an additional 7%). This fairly small decrease in the solubilization capacity could be associ- 44
ated with the fact that the system transforms gradually into a bicontinuous phase and the 45
interfacial area remains almost unchanged when the aqueous phase concentration increases. S46
Surprisingly, in region ΙΙΙ (50–67 wt% aqueous phase) the solubilization capacity increases N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 22

22 Chapter 2

1 from 160 to 450 ppm (an increase of 180%). In region IV the solubilization capacity
2 decreases to 312 ppm (a decrease of 30%).
3 In order to explain the changes in solubilization capacity of lycopene, we characterized
4 the microstructure of microemulsions along dilution line T64 using the SD-NMR tech-
5 nique. Figure 2.8 shows the relative diffusion coefficients of water and R(+)-limonene in
6 empty (containing no solubilizates) microemulsions (Figure 2.8a) and microemulsions
7 solubilizing lycopene (Figure 2.8b), as a function of the aqueous phase concentration
8 (w/w). One can clearly see that the general diffusion coefficient behavior of microemulsion
9 ingredients (R(+)-limonene and water), with or without lycopene, is not very different. The
10 total amount of lycopene does not cause dramatic changes in the diffusion patterns of the
11 ingredients.
12 It can also be seen that, in the two extremes of aqueous phase concentrations (up to
13 20 wt% and above 70–80 wt% aqueous phase), the diffusion coefficients are easily inter-
14 preted, while the regions in between are somewhat more difficult to explain, since gradual
15
16
17
(a)
18 1.000 1.000
19
20
21 0.100 0.000
D W/D0W

D O/D0O
22
23
24 0.010 0.010
25
26
27 0.001 0.001
0 20 40 60 80 100
28
Aqueous phase (wt%)
29
30
31 (b)
1.000 1.000
32
33
34
0.100 0.000
D W/D0W

35
D O/D0O

36
37 0.010 0.010
38
39
40 0.001 0.001
41 0 20 40 60 80 100
42 Aqueous phase (wt%)
43
44 Figure 2.8. Relative diffusion coefficient of water (•) and R()-limonene (▲) in microemulsions
without (a) and with (b) lycopene, as calculated from SD-NMR results at 25ºC. D0w was measured
45 in a solution containing water/PG (1:1), and determined to be 55.510–11 m2 s–1. D0o the pure
46S diffusion coefficient of R()-limonene was determined to be 38.310–11m2 s–1. (Adapted from
47N Garti, with permission from the publisher.)
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 23

Improved Solubilization and Bioavailability of Nutraceuticals 23

changes take place. Regions ΙΙ and ΙΙΙ are difficult to distinguish. However, the structural 1
changes in the presence of lycopene (Figure 2.8b) are more pronounced than those in the 2
absence of lycopene (Figure 2.8a). 3
Microemulsions containing up to 20 wt% aqueous phase, and solubilizing lycopene, 4
have a discrete w/o microstructure, since the relative diffusion coefficients of water and 5
R(+)-limonene differ by more than one order of magnitude. Microemulsions solubilizing 6
lycopene and containing 20–50 wt% aqueous phase have a bicontinuous microstructure, as 7
the diffusion coefficients of water and R(+)-limonene are of the same order of magnitude. 8
Increasing the aqueous phase concentration to above 50 wt% induces the formation of dis- 9
crete o/w microstructures, as the relative diffusion coefficients of water and R(+)-limonene 10
differ by more than one order of magnitude. 11
From the solubilization capacity and SD-NMR results, it is clear that lycopene solubi- 12
lization is structure dependent. 13
The four different regions in the solubilization capacity curve are an indication of the 14
microstructure transition along the dilution line. The first region indicates the formation of 15
w/o (L2) microstructure. The second region indicates the transition from L2 microstructure 16
to a bicontinuous microemulsion. In the third region, a transition from a bicontinuous 17
microemulsion to an o/w (L1) microstructure occurs. In the fourth region a discrete L1 18
microstructure was found. 19
While the general behavior of the diffusion coefficients is the same for microemulsions 20
with or without lycopene, the transition point from one microstructure to another is differ- 21
ent. Lycopene influences the transition from L2 to bicontinuous microstructure and further 22
to L1 microstructure. In empty microemulsions the formation of bicontinuous microstruc- 23
ture occurs when the microemulsion contains 40–60 wt% aqueous phase, whereas in a 24
microemulsion containing lycopene, bicontinuous microstructure starts at low aqueous 25
phase content (20 wt%) and continues up to an aqueous phase content of 50 wt%. It seems 26
that as more water is solubilized in the swollen reverse micelles less free interfacial volume 27
is available for the lycopene. Lycopene appears to disturb both the flexibility of the micelle 28
and the spontaneous curvature. As a result, the interface changes into a flatter curvature 29
(bicontinuous) at an early stage of water concentration, more so in the presence of lycopene 30
than empty micelles. 31
The hydrophilic–lipophilic balance (HLB) of the surfactant influences the quantity of 32
solubilized lycopene in the aqueous surfactant phase. Tween 60, being a hydrophilic surfac- 33
tant with the lowest HLB value (HLB 14.9), solubilizes 10 wt% more lycopene than Tween 34
80 (HLB 15.2). In Tween 40 (polyoxyethylene sorbitan monomyristate)-based microemul- 35
sions, the solubilization capacity drops even further (30%). Replacing Tween 60 with 36
Tween 20, the most hydrophilic surfactant (HLB 16.7), reduces the solubilization capacity 37
of lycopene by 88%. 38
We have also demonstrated that microemulsions stabilized by mixed surfactants enhance 39
the solubilization capacity of lycopene by 32–48%, in comparison to microemulsions stabi- 40
lized by Tween 60 alone (Spernath et al., 2002; Garti et al., 2003, 2007), indicating a syner- 41
gistic effect. Microemulsions stabilized by a mixture of three surfactants—Tween 60, 42
sucrose ester, and ethoxylated monodiglyceride—have the highest solubilization capacity of 43
lycopene—an increase of 48%, in comparison to microemulsion based on Tween 60 alone 44
(Spernath et al., 2002; Garti et al., 2003, 2004a, b). Synergism in surfactant mixtures was 45
attributed to Coulombic, ion-dipole, or hydrogen-bonding interaction (Hou and Shah, 1987; S46
Huibers and Shah, 1997). Therefore, nonionic surfactant mixtures are expected to have a N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 24

24 Chapter 2

1 minimum intermolecular interaction and weak synergistic effects. Nevertheless, Huibers


2 and Shah (1997) demonstrated a strong synergism in nonionic surfactant mixtures, similar
3 to the findings in our study. This behavior remains to be explained. Solubilization capacity is
4 defined as the quantity (mg) of solubilizate entrapped in 100 g microemulsion, and solubi-
5 lization efficiency is the quantity of solubilizate per 100 g of the oil phase or that normalized
6 to oil content solubilization. Solubilization efficacy is the ratio of the quantity of solubilized
7 compound to the quantity of the total amounts of oil and surfactant phase. Microemulsions
8 exhibit very large solubilization capacities and solubilization efficiencies for lycopene.
9 Lycopene was solubilized in a microemulsion up to 10 times its dissolution capacity in
10 R(+)-limonene or in any other edible solvent. The solubilization capacity and efficiency of
11 lycopene are strongly affected by microstructure transitions from w/o to bicontinuous and
12 from bicontinuous to o/w. Solubilization capacity drops significantly with dilution, while
13 the efficiency and efficacy increase as the water content increases, indicating that the inter-
14 face plays a significant role in the solubilization of lycopene.
15
16
17 Phytosterols
18
Elevated serum cholesterol level is a well-known risk factor for coronary heart disease
19
(Hicks and Moreau, 2001). Most strategies for lowering serum cholesterol require dietary
20
restrictions and/or medications. The prospect of lowering cholesterol levels by consuming
21
foods fortified with natural phytonutrients is considered much more attractive.
22
Phytosterols (plant sterols) are steroid alcohols. Their chemical structure resembles
23
human cholesterol, as can be seen in Figure 2.9. Both sterols are made up of a tetracyclic
24
cyclopenta[α]phenanthrene ring system and a long flexible side chain at the C17 carbon
25
atom. The four rings have trans configurations, forming a flat α-system (IUPAC, 1989;
26
Piironen et al., 2000). Moreover, sterols create planar surfaces, at both the top and the
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
Figure 2.9. Molecular structure of cholesterol and some abundant phytosterols (R = H–
45 cholesterol; R = CH2CH3-β-sitosterol; R = CH2CH3 and additional double bond at C22-stigamsterol;
46S R = CH3-campasterol; R = CH3 and additional double bond at C22-brassicasterol. (Adapted from
47N Garti, 2004, with permission from the publisher.)
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 25

Improved Solubilization and Bioavailability of Nutraceuticals 25

bottom of the molecules, since the R-conformation is preferred in the side chain linked to 1
C20 carbon atom of the sterol molecule. This allows for multiple hydrophobic interactions 2
between the rigid sterol nucleus (the polycyclic component) and the membrane matrix 3
(Nes, 1987; Bloch, 1988; Piironen et al., 2000). Only side chains of the various sterols are 4
different. These minor configuration differences result in major differences in their biologi- 5
cal function. 6
Peterson et al. (1951) reported that addition of soy sterols to a cholesterol-enriched diet 7
prevented an increase in the plasma cholesterol level. This effect significantly reduced the 8
incidence of atherosclerotic plaque (Peterson et al., 1951). Since then, numerous clinical 9
investigations have indicated that administration of phytosterols to human subjects reduces 10
the total plasma cholesterol and LDL cholesterol levels (Pelletier et al., 1995; Jones et al., 11
1997). Because of their poor solubility and limited bioavailability, high doses were required 12
to have a noticeable effect. 13
Up to 25 g/day of phytosterol esters were recommended in some reports and up to 14
1.3 g/day of phytosterol esters are to be used as per the FDA recommendation for a 15
decrease of up to 15% of the cholesterol in the blood stream. 16
The exact mechanism by which phytosterols inhibit the uptake of dietary and endoge- 17
nous cholesterol is not completely understood. One theory suggests that cholesterol in the 18
presence of phytosterols precipitates in a nonabsorbable state. A second theory suggests 19
that cholesterol is displaced by phytosterols in the bile salt micelles and phospholipid- 20
containing mixed micelles, thus preventing its absorption (Hicks and Moreau, 2001). 21
Enhanced solubilization of phytosterols in o/w microemulsions has been hypothesized to 22
promote their bioavailability and maximize their absorption in human tissues owing to their 23
small droplet size (in the range of several nanometers). Activity of phytosterols in food for- 24
mulations has not yet been fully studied. Our results (Rozner and Garti, 2006) and that of 25
other investigators (Ostlund, 2002) indicate that phytosterols do not cross human mem- 26
branes, but they significantly retard (or prevent) the penetration of cholesterol and other 27
lipids. We explored the ability of the unique dilutable microemulsions to solubilize phytos- 28
terols and studied the correlation between the solubilization capacity of the phytosterols 29
and the microemulsion microstructure transitions (Spernath, 2003; Garti et al., 2005). 30
Typical solubilization capacity of phytosterols and cholesterol along dilution line T64 are 31
shown in Figure 2.10. The solubilization capacity of phytosterols in concentrated reverse 32
micelle solution–like systems containing surfactant and oil phase (at 6:4 weight ratio, 33
respectively), is 60,000 ppm (6 wt%). As can be seen from Figure 2.10, the solubilization 34
35
36
37
38
39
40
41
42
43
44
45
Figure 2.10. Solubilization capacity (SC) of cholesterol (x) and phytosterols (ο) along dilution S46
line T64 at 25ºC. N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 26

26 Chapter 2

1 capacity of phytosterols decreases with the increase in aqueous phase concentration. In a


2 microemulsion containing 90 wt% aqueous phase, the maximum solubilization capacity is
3 only 2400 ppm, that is, a decrease of 96% in the solubilization capacity of phytosterols.
4 A possible explanation for the dramatic decrease in the solubilization capacity could be
5 related to the nature of the solubilized molecules and to the locus of its solubilization at
6 the interface. In concentrates (without added water), phytosterols are entrapped at the
7 micelle’s interface. As more aqueous phase is added, water-in-oil swollen reverse micelles
8 (w/o microemulsions) are formed, and the hydrophilic OH groups of the phytosterols
9 are oriented toward the aqueous phase, causing the molecules to pack between the surfac-
10 tant hydrophobic chains. This change in the locus of solubilization causes a decrease in
11 solubilization capacity of the interface. Suratkar and Mahapatra (2000) observed a similar
12 change in the locus of solubilization of phenolic compounds in sodium dodecyl sulfate
13 (SDS) micelles.
14 The decrease in solubilization capacity as the aqueous phase concentration increases
15 may be attributed to microstructure transformations. The structural transformation from
16 w/o to o/w microstructure via bicontinuous mesophase forces the phytosterols to solubilize
17 between the hydrophobic amphiphilic chains. This less-preferable location causes a
18 decrease in the solubilization capacity. It seems that the phytosterols have a strong effect on
19 the spontaneous curvature of the micelles. As a result, the interface curvature decreases at
20 lower water concentration. This effect is more pronounced in the presence of phytosterols
21 than in empty micelles or in the presence of lycopene. The effect of phytosterol on choles-
22 terol trans-membrane penetration was extensively studied. Various mechanisms have been
23 suggested for the decrease in the transport of cholesterol in the presence of phytosterols
24 (Trautwein et al., 2003; Hui and Howles, 2005; Rozner and Garti, 2006). Similarly, the
25 competitive adsorption of cholesterol and phytosterols in the microemulsion membrane
26 indicates that reverse microemulsions (w/o) preferentially solubilize more cholesterol over
27 phytosterols. Nevertheless, upon dilution, once inversion to o/w microemulsions occurs,
28 the phytosterols are somewhat better accommodated at the interface and they displace
29 some of the cholesterol molecules from the interface (Figure 2.11).
30
31
Lutein and Lutein Ester
32
33 Evidence that the macular pigment carotenoids—lutein and zeaxanthin—play an important
34 role in the prevention of age-related-macular degeneration, cataract and other blinding dis-
35 orders, is now available (Vandamme, 2002; Bone et al., 2003; Semba and Dagnelic, 2003;
36 Kim et al., 2006). Carotenoids are situated in the macula (macula lutea, yellow spot)
37 between the incoming photons and the photoreceptors and have maximum absorption at
38 445 nm for lutein and 451 nm for zeaxanthin. As a result, lutein and zeaxanthin can func-
39 tion as a blue light filter (400–460 nm). The blue light enters the inner retinal layers,
40 thereby causing the carotenoids to attenuate their intensity. In addition to the protective
41 effect of the macula from blue wavelength damage, these carotenoids can also improve
42 visual acuity and scavenge harmful reactive oxygen species that are formed in the photore-
43 ceptors (Bone et al., 2003; Kim et al., 2006).
44 With aging, some of the eye antioxidant supplies are diminished and antioxidant
45 enzymes are inactivated. This action appears to be related to the accumulation, aggregation,
46S and eventual precipitation in lens opacities of damaged proteins, subsequently leading to
47N numerous eye disorders (Vandamme, 2002; Semba and Dagnelie, 2003).
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 27

Improved Solubilization and Bioavailability of Nutraceuticals 27

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
Figure 2.11. Competitive solubilization of (a, b) cholesterol alone and (c, d) combined
23
phytosterols and cholesterol in bile salt micelles (wt ratio of 1/1) in U-type microemulsions as a 24
function of water dilution. (Adapted from Garti, with permission from the publisher.) 25
26
27
28
To improve the understanding of the potential benefits of carotenoids in general and 29
lutein in particular, it is important to obtain more insight into their bioavailability and the 30
factors that determine their absorption and bioavailability. 31
Lutein, a naturally occurring carotenoid (Figure 2.12), is widely distributed in fruits and 32
vegetables and is particularly concentrated in the Tagetes erecta flower. Epidemiological 33
studies suggest that high lutein intake (6 mg/day) increases serum levels that are associated 34
with a lower risk of cataract and age-related-macular degeneration. Lutein can be extracted 35
either as a free form or as esterified (myristate, palmitate, or stearate) lutein. Both forms are 36
practically insoluble in aqueous systems, resulting in low bioavailability. 37
To improve its bioavailability, lutein was solubilized in U-type microemulsions based on 38
R(+)-limonene. Some of the main findings are (Amar-Yuli et al., 2003, 2004; Garti et al., 39
2003; Amar-Yuli and Garti, 2006): (1) reverse micellar and w/o compositions solubilized 40
both lutein and lutein ester better than o/w microemulsions, while maximum solubilization 41
is obtained within the bicontinuous phase; (2) free lutein is solubilized better than the ester- 42
ified one in the w/o microemulsions, whereas the esterified lutein is better accommodated 43
within the o/w microemulsion; (3) vegetable oils decrease the solubilization of free lutein; 44
(4) glycerol and alcohol enhance the solubilization of both luteins; and (5) solubilization is 45
surfactant-dependent in all mesophase structures, but its strongest effect is in the bicontin- S46
uous phase. N47
Lakkis_Ch_02-013-040 r1.qxd 3/30/07 6:24 PM Page 28

28 Chapter 2

1 (a)
2
3 OH
4
5
6 HO
7
(b)
8
9
10
11
12
13
14
15 Figure 2.12. Chemical structures of (a) free lutein and (b) lutein ester.
16
17
18 On the basis of self-diffusion coefficients of each of the ingredients, a schematic model
19 of the solubilization of lutein in the three possible structures along the dilution line 73
20 (70 wt% surfactant phase and 30 wt% oil phase) was constructed. The schematic location
21 of the lutein at the structures is shown in Figure 2.13.
22
23
Vitamin E
24
25 Microemulsions can also serve as reservoirs for enhanced solubility of lipophilic vitamins
26 or other nutraceuticals within water-based formulations. The pharmaceutical literature is
27 replete with studies of enhanced micellar delivery of vitamins, in particular vitamin E, vita-
28 min K1, and β-carotene (Winn et al., 1989; Traber, 2004).
29 Vitamin E (Figure 2.14), the major lipophilic antioxidant in human body, has invoked a
30 great deal of interest regarding its disease-preventive and health-promoting effects, as well
31 as its unique chemical structure, as a group of amphiphilic homologues exhibiting important
32 interfacial roles in surfactant self-assemblies. Much interest has been devoted to microemul-
33 sions as efficient cosmetic and drug delivery systems, enabling the solubilization of
34 hydrophobic active matter in aqueous media and improving its bioavailability.
35 Therefore, we found it imperative to study the effect of microemulsion composition on
36 the solubilization capacity of different forms of vitamin E and to infer the structural trans-
37 formations from the solubilization data.
38 Our results (Garti et al., 2004a, b) (Figure 2.15) show the following: (1) The solubiliza-
39 tion capacity of α-tocopherols with free-OH head groups in Tween 60-based micro-
40 emulsions drops abruptly at either of the two dilution lines that have been studied at constant
41 surfactant-to-oil ratio, signifying structural transformations in the microemulsion structure.
42 (2) The number of methyl groups on the vitamin’s polar head has an influence on the point at
43 which the solubilization drop occurs, while nonsaturation of the hydrophobic tail of the vita-
44 min enhances its solubilization capacity with no observable impact on the solubilization pat-
45 tern. (3) In contrast to the free-OH vitamin E forms, the acetate form showed continuous
46S decreases in solubilization capacity along the dilution line. (4) The type of oil used in
47N the microemulsion has a strong influence on the solubilization pattern of the vitamin.
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 29

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
Figure 2.13. Schematic model of lutein solubilization.
28
29
30
(a) CH3
31
32
33
HO
CH3 CH3 CH3 34
2R
35
H3C O CH3 36
CH3 4⬘R 8⬘R 37
CH3
38
39
(b) CH3 40
H3C O 41
2R
CH3 CH3 CH3 42
O 43
CH3 O CH3 44
CH3 4⬘R 8⬘R
CH3
45
S46
Figure 2.14. Chemical structures of (a) α-tocopherol and (b) α-tocopherol acetate. N47

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

30 Chapter 2

1
2
3
4
5
6
7
8
9
10
11
12
13 Figure 2.15. Solubilization capacities of free tocopherol (•) and tocopherol acetate (▲) in U-type
14 microemulsions at several dilutions along dilution line 64 (60% surfactant phase and 40 wt% oil
15 phase. (Adapted from Garti, 2002, with permission from the publisher.)
16
17 Triacetin attained a higher solubilization capacity of vitamin E than R(+)-limonene with a
18 certain retardation in the structural transformations along the dilution line. Medium-chain
19 triglycerides (MCT), on the other hand, maintained a constant ratio of TocOH to
20 surfactant with an increasing level of aqueous phase within a certain range, while the solu-
21 bilization capacity of D-α-tocopherol acetate (TocAc) decreased significantly in the same
22 dilution range. (5) Alcohol cosurfactants and propylene glycol (PG) were found to be
23 vitally important for improving the solubilization capacity of TocAc and TocOH. The latter
24 showed a higher boost of solubilization at high levels of alcohols. (6) TocAc was found to
25 prefer higher concentrations of Tween 60 for better solubilization, while TocOH prefers
26 moderate levels. Mixing Tween 60 with diglycerol monooleate (DGM) displayed a pro-
27 nounced enhancement in the solubilization of TocAc, while it caused a significant decrease
28 in that of TocOH. Based on these findings, a commercial vitamin E clear beverage was
29 developed.
30 We have demonstrated that molecules such as essential oils, aromas, isoflavones,
31 β-carotene, and lipoic acid have been similarly solubilized in the NSSL vehicles.
32
33
Oxidative Stability
34
35 In many cases, NSSL vehicles are loaded with nutraceuticals that are very sensitive to oxida-
36 tion. Any preparation containing these formulations should be stable for very long periods of
37 time on the shelf and within the final product. Therefore, protection against environmental
38 oxidative attack is essential. Micelles are very dynamic systems with a very fast exchange of
39 the surfactant molecules between the interface and the continuous phase.
40 Microemulsions are swollen micelles with similar fast exchange. However, systems that
41 are rich in surfactant content form very concentrated phases, where the swollen micelles
42 (the droplets) are tightly packed. Very condensed packed systems with strong inter-droplet
43 interactions are obtained. In these systems the mobility of the surfactants is very restricted.
44 In addition, stability was found to be dependent on the nature of the surfactant; therefore,
45 even more tightly packed, worm–like, and entangled giant micelles can be formed.
46S The stability against oxidation of lycopene, known for its poor oxidative stability once
47N dissolved in solvents, was evaluated. Lycopene, if exposed to air and light, will be much
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 31

Improved Solubilization and Bioavailability of Nutraceuticals 31

100 1

% of lycopene from original


Emulsion
2
NSSL
75 3
4
50
5
6
7
25
8
9
0 10
0 28 52 72 11
Time (days)
12
Figure 2.16. Oxidative stability to air and light of 23 mg lycopene emulsified in 10 g of o/w 13
emulsion versus in the NSSL (modified microemulsion) vehicles. 14
15
16
more stable against autooxidation when solubilized in NSSL vehicles than if loaded onto 17
emulsion droplets, as shown in Figure 2.16. After a few weeks, the emulsified lycopene was 18
totally oxidized, while over 65 wt% of the NSSL lycopene remained stable. Similar results 19
were obtained with other nutraceuticals (private communications). 20
21
Bioavailability 22
23
Some nutraceuticals are known to be practically insoluble in water and, therefore, tablets or 24
capsules that are taken orally tend to precipitate once the active ingredient is diluted with 25
water (in human digestive tracts). As a result, the bioavailability is very limited, and the 26
adsorption from the intestine to the blood serum is poorly controlled. Moreover, tablets and 27
capsules exhibit strong fluctuations and as a result their activity is questionable. Two such 28
examples that are discussed are CoQ10 and lycopene. 29
30
CoQ10 and Improved Bioavailability 31
32
Coenzyme Q10 and related ubiquinones were first discovered in 1955 and were extracted
33
and isolated from the mitochondria. The number of side chain isoprenoid units determines
34
the nomenclature. Coenzyme Q6 is found in bacteria, whereas CoQ10 is found in mam-
35
malian mitochondria. CoQ10 is one part of a complex series of reactions that occur within
36
mitochondria—ultimately linked to the generation of energy within a cell. The chemical
37
structure of a CoQ10 is depicted in Figure 2.17.
38
39
40
41
42
43
44
45
S46
Figure 2.17. Chemical structure of CoQ10. N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 32

32 Chapter 2

1 Virtually every cell in the human body contains coenzyme Q10. The mitochondria, the
2 area of cells where energy is produced, contains most of the human coenzyme Q10. The
3 heart and the liver, due to their high content of mitochondria per cell, contain the greatest
4 quantity of coenzyme Q10. Coenzyme Q10 supplementation has helped some people with
5 congestive heart failure (Salles et al., 2006; Yamamoto, 2006). Ubiquinone, or coenzyme
6 Q10, is an important heart nutrient, used primarily by those who take pills against high cho-
7 lesterol levels. Certain lipid-lowering drugs, such as statins as well as oral agents, which
8 lower blood sugar, cause a decrease in serum levels of coenzyme Q10 and reduce the effects
9 of coenzyme Q10 supplementation (Mortensen et al., 1997; Palomaki et al., 1998; Lankin,
10 2003; Passi et al., 2003; Bettowski, 2005; Cenedella et al., 2005; Hargreaves et al., 2005;
11 Mabuchi et al., 2005; Strey et al., 2005). These drugs inhibit the production of coenzyme
12 Q10 by the liver, and will cause serious complications, unless one supplements coenzyme
13 Q10 back into the diet. A prescription for lipid-lowering statin drugs should always be
14 accompanied with a recommendation to take coenzyme Q10, because if a person is deficient
15 in coenzyme Q10, heart failure is more likely.
16 The second major use of coenzyme Q10 would be in the case of congestive heart failure,
17 where it is particularly effective. Its importance to the human heart is illustrated by the fact
18 that the heart may cease to function when coenzyme Q10 levels fall by 75%. Schematic
19 activity within the mitochondria of CoQ10 is demonstrated in Figure 2.18.
20 Adenosine triphosphate (ATP) is present in every cell of human organs. It serves as a
21 source of energy for many of the body’s biochemical processes and represents the reserve
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S
47N Figure 2.18. Schematic functionality of CoQ10 in mitochondria.
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 33

Improved Solubilization and Bioavailability of Nutraceuticals 33

1
2
3
4
5
6
7
8
9
Figure 2.19. Bioavailability of CoQ10 in humans given a total of 150 mg of active matter in 10
two daily doses in two types of formulations, in best commercial formulation in the market 11
place (entitled 275% more bioavailable, filled bar) versus the CoQ10 solubilized in NSSL vehicles
(white bar).
12
13
14
energy in the muscles. The heart needs a constant supply of ATP, which cannot be produced 15
without coenzyme Q10. Coenzyme Q10 is the catalyst for the creation of ATP. This means 16
that coenzyme Q10 plays a vital role in the inner workings of the human body. Several other 17
chronic diseases are associated with lack of CoQ10 such as Parkinson’s disease (Andrey and 18
Gille, 2003; Batandier et al., 2004; Genova et al., 2004; Sharma et al., 2004; Arroyo and 19
Navas, 2005; Ebadi et al., 2005; Dhanasekaran and Ren, 2005; Moriera et al., 2005). It is 20
also a potent antioxidant since it fights the harmful free radicals generated during normal 21
metabolism. 22
The highest dietary sources of CoQ10 come from fresh sardines and mackerel, the heart, 23
the liver, and beef, lamb, and pork, as well as from eggs. There are plenty of vegetable 24
sources of CoQ10, the richest being spinach, broccoli, peanuts, wheat germ, and whole 25
grains, although the amount is significantly smaller than that found in meat. 26
Coenzyme Q10 is primarily offered in tablet, capsule, or soft gel forms containing a 27
yellow-orange powder. The tablet form, being much less digestible, is not recommended. 28
Adult levels of supplementation are usually 30–90 mg/day, although individuals with spe- 29
cific health conditions may supplement with higher levels, such as 100 mg 3–4 times per 30
day. Most of the research on heart conditions has used 90–150 mg/day. CoQ10 is fat soluble 31
and, like most other fat-soluble compounds, is poorly absorbed from the gastrointestinal 32
tract, especially when taken on an empty stomach. Therefore, it is recommended that 33
CoQ10 be taken with a meal or in a formulation, such as oil phase, that will improve its 34
bioavailability and, hence, absorption. Our studies on humans were conducted at the Tech- 35
nical University of Tokyo by Prof. Yamamoto on eight individuals who were fed for 28 days 36
with CoQ10 from a commercial product known as “275% more bioavailable”: and with our 37
NSSL vehicles incorporated into soft gels. The individual intake was of 150 mg CoQ10 per 38
day (Yamamoto, 2005). 39
The efficacy of the NSSL-based formulations versus the commercial product is demon- 40
strated in Figures 2.19–2.21. It can be clearly concluded that (1) CoQ10 in the NSSL 41
vehicles is more bioavailable than the commercial product in soft gels (claimed to be 275% 42
more bioavailable than other products in tablets); (2) the ratio of total CoQ10 to total choles- 43
terol in the blood stream derived from the NSSL soft gels is higher than from the commer- 44
cial product, indicating that the NSSL vehicles provide extra activity to the CoQ10, which 45
assists in maintaining total cholesterol at lower levels; (3) it is well documented that several S46
nutraceuticals and oil-soluble phytochemicals tend to interfere with the absorption of N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 34

34 Chapter 2

1
2
3
4
5
6
7
8
9 Figure 2.20. Ratio of CoQ10 (TQ) to total cholesterol (TC) in human blood when given 150 mg
10 of CoQ10 in two daily doses in two types of formulations, in best commercial formulation in the
11 market place (entitled 275% more bioavailable, filled bar) versus the CoQ10 solubilized in NSSL
12 vehicles (white bar).
13
14
15
16
17
18
19
20
21
22
23
24 Figure 2.21. Ratio of vitamin E (VE) to total cholesterol in human blood given a total of 150 mg
25 of CoQ10 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
26
vehicles (white bar).
27
28
29
30 vitamins. Therefore, it was expected that the vitamin E levels in the blood stream would
31 decrease with the intake of CoQ10. However, it was found in the human blood tests that vita-
32 min E levels did not decrease in the presence of CoQ10 when CoQ10 was taken in the NSSL
33 vehicles. In fact, it remained at higher levels when compared to its levels derived from the
34 commercial product.
35 On the basis of these and other findings, we have proposed a highly schematic cartooned
36 model (Figure 2.22) of the transport of the nutraceuticals across human membranes. The
37 model shows how the vehicle that is dispersed in the aqueous phase approaches the mem-
38 brane and adheres to it. The CoQ10 is transported across the membrane, while the empty
39 vehicles depart and are excreted from the digestive tract. It should be noted that the surfac-
40 tants do not cross the membrane.
41
42
Water Binding
43
44 The activity of water plays a significant role in any reaction (chemical or enzymatic) that
45 exists in food systems and related products. Microemulsions of w/o can serve as microreac-
46S tors for several such processes, mainly for Maillard reactions (Lutz et al., 2005). Water-in-oil
47N nanodroplets 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 35

Improved Solubilization and Bioavailability of Nutraceuticals 35

1
2
3
4
5
6
7
8
9
10
11
12
13
14
Figure 2.22. Schematic representation of the microemulsion droplet approaching the membrane 15
and releasing the nutraceutical molecules. The surfactant does not cross the membrane. 16
17
18
19
estimate the activity of the water and the binding capacity of the surfactants is of high 20
importance whenever a triggered reaction is required. At certain water levels, the water in 21
the core of the microemulsion will be bound and the activity will be minimal; thus, the reac- 22
tivity of the ingredients (sugars and proteins in Maillard reactions or enzymes in hydrolysis 23
processes) will be low. Upon adding more water and reaching a point where the water 24
becomes free, the reactions will be triggered (Yaghmur et al., 2003a, b). 25
We (Spernath, 2003; Yamomoto, 2006) examined, by a sub-zero differential scanning 26
calorimeter (DSC) technique, the nature of the water in the confined space of a w/o 27
microemulsion, to better understand the role of the entrapped water, in order to control 28
enzymatic reactions carried out in the inner phase (Spernath et al., 2003; Yaghmur et al., 29
2003). We reported (Figure 2.23) that the surfactant/alcohol/PG can strongly bind water in 30
the inner phase, so that it freezes below –10°C and acts, in part, as bound water and, in part, 31
as non-freezable water (Spernath et al., 2003). Even after complete inversion to o/w micro- 32
emulsions, the water in the continuous phase strongly interacts with the cosolvent/surfac- 33
tant and remains partially bound. 34
The water in the core of nonionic microemulsions containing, in addition to the surfac- 35
tants, polyols and alcohol, is strongly bound to the surfactant head group and/or to the 36
polyol groups and freezes at subzero temperatures. The amount of bound water strongly 37
depends on the amounts of the surfactants present in each microdroplet, on the nature of the 38
head groups, and on the amounts of cosolvents (alcohol and PG). 39
On the basis of these findings, Maillard reactions, model reactions of furfural and cys- 40
teine and glucose and isoleucine (Ezrahi et al., 2001; Fanun et al., 2001; Yaghmur et al., 41
2002a, b, 2003, 2005; Lutz et al., 2005), as well as hydrolysis of phosphatidylcholine by 42
phospholipase L2 (PLA2) to lysolecithin (Garti et al., 1997) were studied. It was found that 43
the reactions do not start (lag time) until sufficient water is added to exceed the free water 44
threshold. The reactions are, therefore, very well triggered and controlled by the water 45
activity within the core of the microdroplets. The reaction rates can be delayed or speeded S46
by immobilizing (confining) or freeing the water in the core of the microdroplets. N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 36

36 Chapter 2

1 35
2
3
30
4
Interphasal or free water (wt%)

5
6 25
7
8 20
9
10
11 15
12
13 10
14
15
16 5
17
18 0
19 0 5 10 15 20 25 30 35 40
20 Water content (wt%)
21
Figure 2.23. The amounts (weight percent of free and bound) of interphasal water in
22
microemulsions based on sugar esters along dilution line 64 (60% surfactant and 40% oil
23 phase). (o) Bulk (free) water and (•) interphasal (bound) water. (Adapted from Garti, 1995,
24 with permission from the publisher.)
25
26
27
Conclusions
28
29 Microemulsions have been known for several decades, but their utilization in food systems
30 has been very limited owing to some major structural limitations and the nature of the
31 surfactants and the oils. Another major drawback is that in most cases they were undilutable
32 with water. In recent years, after significant efforts by colloid chemists, experimentalists,
33 and others, some of the key characteristics related to the packing of the surfactant, free
34 energy gain, geometries, and so on, shed light on the basic requirements needed to design
35 U-type phase diagrams. The latter consist of large isotropic regions and have proved
36 capable of making concentrates that can be easily diluted with water and oil phases. In the
37 course of our studies we also learned that:
38
39 • Self-assembled, hydrophilic surfactant in oil phase, in the presence of cosolvents and
40 cosurfactants, can provide high solubilization capacities for entrapment of immiscible
41 phases and active guest molecules. These microstructures can be diluted with excess
42 water to form crystal-clear (transparent) solution-like, isotropic phases, loaded with the
43 active matter.
44 • If the ingredients composing the microemulsions and the cosolvents and cosurfactants
45 are carefully selected, one can form a variety of beverage microemulsions.
46S • Microemulsions of U-type with progressive full dilution with aqueous phase can be
47N formulated.
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 37

Improved Solubilization and Bioavailability of Nutraceuticals 37

• Microemulsions of w/o and bicontinuous structures, as well as o/w microemulsions can 1


solubilize guest molecules at their interface at high solubilization capacities, in some 2
cases up to 100-fold of the solubility of the nutraceuticals in the corresponding solvent! 3
• Molecules such as lycopene, vitamin E, tocopherols and tocopherol acetate, β-carotene, 4
lutein, phytosterols, and CoQ10 can be quantitatively solubilized. 5
• Microemulsions provide some oxidative protection to the nutraceuticals. 6
• Various other guest molecules such as aromas, flavors, and antioxidants can be solubi- 7
lized in the microemulsions. 8
• Water entrapped at the core of a w/o microemulsion can be strongly bound to the surfac- 9
tant head group that will restrict the water activity. Thus, upon adding more water, the 10
reaction by the enzyme or regents can be triggered. 11
12
It seems that we are now ready to start using microemulsions in beverages and other food 13
systems and to incorporate active ingredients within high-quality food for the benefit of 14
human nutrition and health. 15
16
References 17
18
Alander, J.; Warnheim, T. 1989a. Model microemulsions containing vegetable-oils. 1. nonionic surfactant sys-
19
tems. JAOCS 66:1656–1660.
Alander, J.; Warnheim, T. 1989b. Model microemulsions containing vegetable-oils. 2. ionic surfactant systems. 20
JAOCS 66:1661–1665. 21
Amar-Yuli, I.; Garti, N. 2006. Progress in Structural Transformation in Lyotropic Liquid Crystals. Colloids and 22
Surfaces 282: 435–456. 23
Amar-Yuli, I.; Aserin, A.; Garti, N. 2003. Solubilization patterns of lutein and lutein esters in food grade nonionic
24
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 25
lutein esters in food microemulsions. Colloids Surfaces B, 33:143–150. 26
Andrey, V.; Gille, L. 2003. The biomolecule ubiquinone exerts a variety of biological functions. BioFactors 27
18(1–4):23–31. 28
Arroyo, A.; Navas, P. 2005. Specificity of coenzyme Q10 for a balanced function of respiratory chain and endoge-
29
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 30
reactive oxygen species production at the level of the respiratory chain complex I. J. Biol. Chem. 31
279(17):17197–17204. 32
Bettowski, J. 2005. Statins and modulation of oxidative stress. Toxicol. Mech. Meth. 15(2):61–92. 33
Billman, J.F.; Kaler, E.W. 1991. Structure and phase-behavior in 4 component nonionic microemulsions.
34
Langmuir 7:1609–1617.
Bloch, K. 1988. Sterol structure and function. J. Am. Oil Chem. Soc. 65:1763–1766. 35
Bone, R.A.; Landrum, J.T.; Guerra, L.H.; Ruiz, C.A. 2003. Lutein and zeaxanthin dietary supplements raise mac- 36
ular pigment density and serum concentrations of these carotenoids in humans. J. Nutrition 133(4):992–998. 37
Bramley, P.M. 2000. Is lycopene beneficial to human health? Phytochemistry 54:233–236. 38
Cenedella, R.J.; Neely, A.R.; Sexton, P. 2005. Concentration and distribution of ubiquinone (coenzyme Q), the
39
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 40
containing long- versus medium-chain glycerides. Int. J. Pharm. 158:57–68. 41
Cooke, J.P. 1998. Nutraceuticals for cardiovascular health. Am. J. Cardiol. 82:43S–46S. 42
de Campo, L.; Yaghmur, A.; Garti, N.; Leser, M.E.; Glatter, O. 2004. Food-grade microemulsions: structural char- 43
acterization. J. Colloid Interface Sci. 274(1):251–267.
44
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. 45
Dungan, S.R. 1997. “Microemulsions in Foods: Properties and Applications.” In: Industrial Applications of S46
Microemulsions, edited by C. Solans and H. Kunieda, pp. 148–170. Marcel Dekker Inc. N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 38

38 Chapter 2

1 Ebadi, M.; Brown-Borg, H.; El Refaely, H.; Singh, B.B.; Garret, S.; Shavali, S.; Sharma, S.K. 2005. Metallothionein-
2 mediated neuro protection in genetically engineered mouse models of Parkinson’s disease. Molecular Brain
Res. 134(1):67–75.
3 Ezrahi, S.; Aserin, A.; Garti, N. 1999. “Aggregation Behavior in One-Phase (Winsor IV) Microemulsion Sys-
4 tems.” In Handbook of Microemulsion Science and Technology, edited by P. Kumar and K.L. Mittal, pp.
5 185–246. Marcel Dekker Inc.
6 Ezrahi, S.; Aserin, A.; Fanun, M.; Garti, N. 2001. “Subzero Temprature Behavior of Water in Microemulsions.” In
7 Thermal 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-
8 tors for model Maillard reaction. Colloids Surfaces A: Physicochem. Eng. Asp. 194(1–3):175–187.
9 Garti, N.; Aserin, A.; Mytayeva, U. 2007. Submitted for publication.
10 Garti, N.; Lichtenberg, D.; Silberstein, T. 1997. The hydrolysis of phosphatidylcholine A2 in microemulsion as
11 microreactor. Colloids Surfaces A: Physicochem. Eng. Asp. 128(1–3):17–25.
12 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.
13 Garti, N.; Aserin, A.; Fanun, M. 2000b. Non-ionic sucrose esters microemulsions for food applications. Part 1.
14 Water solubilization. Colloids Surfaces A: Physicochem. Eng. Asp. 164(1):27–38.
15 Garti, N; Yaghmur, A; Leser, M.E; Clement,V; Watzke, H.J. 2001. Improved oil solubilization in O/W food-grade
16 microemulsions in the presence of polyols and ethanol. J. Agric. Food Chem. 49:2552–2562.
17 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 and
18 4):397–410.
19 Garti, N.; Yaghmur, A.; Aserin, A.; Spernath, A.; Elfakess, R.; Ezrahi, S. 2004a. Solubilization of active molecules
20 in microemulsions for improved solubilization, and environmental protection. Colloids Surfaces A: Physic-
21 ochem. Eng. Asp. 230(1–3):183–190.
22 Garti, N.; Zakharia, I.; Spernath, A.; Yaghmur, A.; Aserin, A.; Hoffman, R.E.; Jacobs, L. 2004b. Solubilization of
water-insoluble nutraceuticals in nonionic microemulsions for water-based use. Progr. Colloid Polym. Sci.
23 126:184–189.
24 Garti, N.; Spernath, A.; Aserin, A.; Lutz, R. 2005. Nano-sized self-assemblies of nonionic surfactants as solubi-
25 lization reservoirs and microreactors for food systems. Soft Matter 1:206–218.
26 Garti, N.; Aserin, A.; Mytayeva, U. 2007. In preparation.
27 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.
28 Genova, M.L.; Pich, M.M.; Bernacchia, A.; Binachi, C.; Biondi, A.; Bovina, C.; Falasca, A.I.; Formiggini, G.;
29 Castelli, G.P.; Lenaz, G. 2004. The mitochondrial production of reactive oxygen species in relation to aging and
30 pathology. Annals New York Academ. Sci. 1011:86–100.
31 Hamdan, S.; Lizana, R.; Laili, C.R. 1995. Aqueous and nonaqueous microemulsion systems with a palm oil-base
32 emollient. 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 on
33 coenzyme Q10. Possible biochemical/clinical implications. Drug Safety 28(8):659–676.
34 Hicks, K.B.; Moreau, R.A. 2001. Phytosterols and phytostanols: functional food cholesterol busters. Food Tech-
35 nol. 55:63–67.
36 Holmberg, K. 1998. “Quarter Century Progress and New Horizons in Microemulsions.” In Micelles, Microemul-
37 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-
38 lization of water in water/oil microemulsions. Langmuir 3:1086–1096.
39 Hui, D.Y.; Howles, P.N. 2005. Molecular mechanisms of cholesterol absorption and transport in the intestine.
40 Semin. Dev. Biol. 16(2):183–192.
41 Huibers, P.D.T.; Shah, D.O. 1997. Evidence for synergism in nonionic surfactant mixtures: enhancement of solu-
42 bilization in water-in-oil microemulsions. Langmuir 13:5762–5765.
IUPAC, The Nomenclature of Steroids (Recommendations 1989), International Union of Pure and Applied Chem-
43 istry and International Union of Biochemistry and Molecular Biology. Available: http:// www.chem.qmw.ac.uk/
44 iupac/steroid/ [1989].
45 Jones, P.J.H.; MacDougall, D.E.; Ntanios, F.; Vanstone, C.A. 1997. Dietary phytosterols as cholesterol-lowering
46S agents in humans. Can. J. Physiol. Pharmac. 75:217–227.
47N
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 39

Improved Solubilization and Bioavailability of Nutraceuticals 39

Joubran, R.F.; Cornell, D.G.; Parris, N. 1993. Microemulsions of triglyceride and non-ionic surfactant-effect of 1
temperature and aqueous phase composition. Colloids Surf. A 80:153–160. 2
Kahlweit, M.; Busse, G.; Faulhaber, B.; Jen, J. 1996. Shape changes of globules in nonionic microemulsions.
J. Phys. Chem. 100:14991–14994.
3
Kanei, N.; Tamura, Y.; Kunieda, H. 1999. Effect of types of perfume compounds on the hydrophile-lipophile bal- 4
ance temperature. J. Colloid Interface Sci. 218:13–22. 5
Kim, S.R.; Nakanishi, K.; Itagaki, Y.; Sparrow, J.R. 2006. Photooxidation of A2-PE, a photoreceptor outer seg- 6
ment fluorophore, and protection by lutein and zeaxanthin. Experimental Eye Research 82(5):828–839. 7
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-
8
dation during therapy with statins. Molecular Cell. Biochem. 249(1 and 2):129–140. 9
Lutz, R.; Aserin, A.; Garti, N. 2005. Maillard reaction between leucine and glucose in o/w microemulsion media 10
in comparison to aqueous solution. J. Disper. Sci. Tech. 26(5):535–547. 11
Mabuchi, H.; Higashikata, T.; Kawashiri, M.; Katsuda, S.; Mizuno, M.; Nohara, A.; Inazu, A.; Koizumi, J.; 12
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.
13
Malcolmson, C.; Lawrence, M.J. 1995. Three-component non-ionic oil-in-water microemulsions using poly- 14
oxyethylene ether surfactants. Colloids Surfaces B: Biointerfaces 4:97–109. 15
Moriera, P.I.; Santos, M.S.; Sena, C.; Nunes, E.; Seica, R.; Oliveira, C.R. 2005. CoQ10 therapy attenuates 16
amyloid—peptides toxicity in brain mitochondria isolated from aged diabetic rats. Exp. Neurol. 17
196(1):112–119.
Mortensen, S.A.; Leth, A.; Agner, E.; Rohde, M. 1997. Dose-related decreases of serum coenzyme Q10 during
18
treatment with HMG-CoA reductase inhibitors. Molecular Aspects Med. 18(Suppl.):s137–s144. 19
Nes, W.R. 1987. “Multiple Roles for Plant Sterols.” In The Metabolism, Structure and Function of Plant Lipids, 20
edited by P.K. Stumpf, B.J. Mudd and W.R. Nes, pp. 3–9. Plenum Press. 21
Palomaki, A.; Malminiemi, K.; Solakivi, T.; Malminiemi, O. 1998. Ubiquinone supplementation during lovastatin 22
treatment: 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 parenteral
23
delivery. Int. J. Pharm. 181:173–179. 24
Passi, S.; Stancato, A.; Aleo, E.; Dmitrieva, A.; Littarru, G.P. 2003. Statins lower plasma and lymphocyte 25
ubiquinol/ubiquinone withput affecting and PUFA. Biofactors 18(1–4):113–124. 26
Pelletier, X.; Belbraouet, S.; Mirabel, D.; Mordret, F.; Perrin, J.L.; Pages, X.; Debry, G. 1995. A diet moderately 27
enriched in phytosterols lowers plasma cholesterol concentrations in normocholesterolemic humans. Ann. Nutr.
Metab. 39:291–295.
28
Peterson, D.W.; Nichols, C.W.; Schneour E.W. 1951. Some relationships among dietary sterols, plasma and liver 29
cholesterol levels and atherosclerosis in chicks. Proc. Soc. Exp. Biol. Med. 78:1143–1147. 30
Piironen, V.; Lindsay, D.G.; Miettinen, T.A.; Toivo, J.; Lampi, A.M. 2000. Review—plant sterols: biosynthesis, 31
biological function and their importance to human nutrition. J. Sci. Food Agric. 80:939–966. 32
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.
33
Asp. 166:177–186. 34
Radomska, A.; Dobrucki, R. 2000. The use of some ingredients for microemulsion preparation containing retinol 35
and its esters. Int. J. Pharm. 196:131–134. 36
Rao, A.V.; Agarwal, S. 1999. Role of lycopene as antioxidant carotenoid in the prevention of chronic diseases: a 37
review. 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 the
38
microstructure of a four-component nonionic microemulsion by cryo-TEM, NMR, SAXS, and SANS. Lang- 39
muir 12:668–674. 40
Rozner, S.; Garti, N. 2006. The activity and absorption relationship of cholesterol and phytosterols. Colloids and 41
Surfaces A: Physicochem. Eng. Aspects 282:435–456. 42
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-
43
chondrial diabetes treated with Coenzyme Q10. Diabet. Res. Clin. Pract. 72(1):100–103. 44
Semba, R.D.; Dagnelie, G. 2003. Are lutein and zeaxanthin conditionally essential nutrients for eye health? Med. 45
Hypotheses 61(4):465–472. S46
N47
Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 40

40 Chapter 2

1 Sharma, S.; Kheradpezhou, M.; Shavali, S.; El Refaely, H.; Eken, J.; Hagen, C.; Ebadi, M. 2004. Neuroprotective
2 actions of coenzyme Q10 in Parkinson’s disease. Meth. Enzymol. 382(Quinones and Quinone Enzymes,
Part B):488–509.
3 Shinoda, K.; Lindman, B. 1987. Organized surfactant systems: microemulsions. Langmuir 3:135–149.
4 Solans, C.; Pons, R.; Kunieda, H. 1997. “Overview of Basic Aspects of Microemulsions.” In Industrial Applica-
5 tions of Microemulsions, edited by C. Solans and H. Kunieda, pp. 1–17. Marcel Dekker Inc.
6 Spernath, A.; Yaghmur, A.; Aserin, A.; Hoffman, R.E.; Garti, N. 2002. Food grade microemulsions based on non-
7 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
8 microstructure transitions in Winsor IV food-grade microemulsions studied by self-diffusion NMR. J. Agric.
9 Food Chem. 51(8):2359–2364.
10 Strey, C.H.; Young, J.M.; Molyneux, S.L.; George, P.M.; Florkowski, C.M.; Scott, R.S.; Frampton, C.M. 2005.
11 Endothelium-ameliorating effects of statin therapy and coenzyme Q10 reductions in chronic heart failure. Ath-
12 erosclerosis 179:201–206.
Suratkar, V.; Mahapatra, S. 2000. Solubilization site of organic perfume molecules in sodium dodecyl sulfate
13 micelles: new insights from proton NMR studies. J. Colloid Interface Sci. 225:32–38.
14 Tokuoka, Y.; Uchiyama, H.; Abe, M.; Christian, S.D. 1995. Solubilization of some synthetic perfumes by anionic-
15 nonionic mixed surfactant systems 1. Langmuir 11:725–729.
16 Traber, M.G. 2004. The ABCs of vitamin E and β-carotene absorption. Am. J. Clin. Nutr. 80(1):3–4.
17 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.
18 Trevino, S.F.; Joubran, R.; Parris, N.; Berk, N.F. 1998. Structure of a triglyceride microemulsion: a small-angle
19 neutron scattering study. J. Phys. Chem. B 102:953–960.
20 Trotta, M.; Morel, S.; Gasco, M.R. 1997. Effect of oil phase composition on the skin permeation of felodipine
21 from o/w microemulsions. Pharmazie 52:50–53.
22 Vandamme, T.F. 2002. Microemulsions as ocular drug delivery systems: recent developments and future chal-
lenges. Progr. Retinal Eye Res. 21(1):15–34.
23 Van het Hof, K.H.; West, C.E.; Weststrate, J.A.; Hautvast, J.G.A.J. 2000. Dietary factors that affect the bioavail-
24 ability of carotenoids. J. Nutr. 130:503–506.
25 von Corswant, C.; Söderman, O. 1998. Effect of adding isopropyl myristate to microemulsions based on soybean
26 phosphatidylcholine and triglyceride. Langmuir 14:3506–3511.
27 von Corswant, C.; Engström, S.; Söderman, O. 1997. Microemulsions based on soybean phophatidylcholine and
triglyceride phase behavior and microstructure. Langmuir 13:5061–5070.
28 Warisnoicharoen, W.; Lansley, A.B.; Lawrence, M.J. 2000. Nonionic oil-in-water microemulsions: the effect of oil
29 type on phase behavior. Int. J. Pharm. 198:7–27.
30 Winn, M.J.; White, P.M.; Scott, A.K.; Pratt, S.K.; Park, B.K. 1989. The bioavailability of a mixed micellar prepa-
31 ration of vitamin K1, and its procoagulant effect in anticoagulated rabbits. J. Pharm. and Pharmacol.
32 41(4):257–260.
Yaghmur, A.; Aserin, A.; Garti N. 2002a. Furfural-cysteine model reaction in food-grade nonionic o/w
33 microemulsions for selective flavor formation. J. Agric. Food Chem. 50:2878–2883.
34 Yaghmur, A.; Aserin, A.; Garti N. 2002b. Phase behavior of microemulsions based on food-grade nonionic surfac-
35 tants: effect of polyols and short-chain alcohols. Colloids Surfaces A 209:71–81.
36 Yaghmur, A.; Aserin, A.; Tiunova, I.; Garti, N. 2002c. Structural behavior of nonionic surfactants in the presence
37 of 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-water
38 microemulsions by PGSE-NMR, conductivity, and viscosity. Langmuir 19(4):1063–1068.
39 Yaghmur, A.; Fanun, M.; Aserin, A.; Garti, N. 2003b. “Food Grade Microemulsions Based on Nonionic Emulsi-
40 fiers as Microreactors for Selective Flavor Formation by Maillard Reaction.” In Self-Assembly, edited by B.H.
41 Robinson, pp. 144–151, IOS Press.
42 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.
43 Yaghmur, A.; Aserin, A.; Abbas, A.; Garti, N. 2005. Reactivity of furfural-cysteine model reaction in food grade
44 five-component nonionic microemulsions. Colloids and Surfaces A 253(1–3):223–234.
45 Yamamoto, Y. 2005. Private communication report.
46S Yamamoto, Y. 2006. Coenzyme Q10, free radicals, and heart disease. Oxidative Stress and Disease 21(Molecular
47N Interventions in Lifestyle-Related Diseases):37–46.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 41

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

3 Emulsions as Delivery Systems in Foods 1


2
3
4
Ingrid A.M. Appelqvist, Matt Golding, 5
Rob Vreeker, and Nicolaas Jan Zuidam 6
7
8
9
10
11
12
Introduction 13
14
Many industries use emulsion technology as a delivery vehicle for either aqueous- or oil- 15
based actives (or both). Examples include paint, pharmaceutical and bitumen industries. In 16
all cases, there are two considerations that must be taken into account when formulating an 17
emulsion for controlled delivery. First, the emulsion system must be (storage) stable right 18
up to the point of application. Secondly, upon its application the emulsion should behave in 19
a consistent manner so that it achieves the desired delivery. In many (but by no means all) 20
cases this equates to the “making and breaking” of emulsions for stability and subsequent 21
delivery. 22
Emulsion systems are, of course, an integral part of food manufacturing. Emulsion tech- 23
nology in the context of foods is not in itself novel—examples include milk, dairy cream, 24
and mayonnaise. The latter can be traced back to the 17th century. However, the use of 25
emulsions as delivery vehicles represents a rapidly developing area for the application of 26
emulsions within the food industry. 27
Similar to other industries, same essential formulation and processing considerations 28
apply. First, the emulsion should be stable up to the point of application—in other words: 29
shelf stable. This is true for all food emulsions, although there may be significant variation 30
in the length of time that the product is required to be stable. Generally, this is limited by the 31
microbiological stability of the particular product: pasteurized emulsions, such as milk or 32
cream, may have a two-week shelf life. In contrast, sterilized emulsions such as crème 33
liqueurs may be stable for over a year. However, in all cases it is important that during the 34
lifetime of the product the emulsion does not show signs of instability or phase separation. 35
Secondly, the emulsion should be designed so that it performs in a defined manner upon 36
application. 37
In food products, there are effectively two main points of application: consumption and 38
digestion. From a consumer perspective, the first point of application might be construed 39
as being the most important. The whole sensory experience of food is dominated by 40
its behavior in the mouth. Emulsions play an important role here, both in terms of flavor 41
delivery and release and in terms of textural behavior and response in the mouth. Mouth 42
is a remarkably sensitive tool at differentiating between organoleptic sensations— 43
most of us are able to differentiate between skimmed, semi-skimmed and whole milk. 44
Consequently, even small changes to emulsion composition and in-mouth behavior 45
can have a significant impact on whether a particular product is perceived in a positive or S46
negative way. N47

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

42 Chapter 3

1 In recent years, consumer demand for highly nutritional food products has increased and
2 can be interpreted as:
3
4 1. removal of so-called “bad” ingredients, such as sugar, fat (saturated or trans) and salt;
5 2. enhancement of “good” ingredients, such as fiber, protein or fruit and vegetable content
6 and;
7 3. direct fortification with actives, such as vitamins, minerals, ω-3 oils.
8
9 In all cases it is important that not only there is no compromise in quality but also any
10 claimed fortification should have good bioavailability during digestion. Consequently, in
11 addition to in-mouth behavior, emulsion systems are becoming increasingly utilized in food
12 products as a means of achieving controlled delivery in the gastrointestinal (GI) tract as well.
13 This chapter highlights recent developments in the application of food emulsions as
14 delivery vehicles from the consideration of both mouth and gut as areas for targeted delivery.
15 We aim to demonstrate the technical challenges and solutions for delivering both oil- and
16 water-soluble actives, providing examples from flavor delivery in mouth to delivery of active
17 compounds and sterols under gastric conditions. We also aim to show how nature can pro-
18 vide solutions for the application of emulsions as delivery systems, as well as looking at
19 future developments and opportunities in this richly diverse field.
20
21
Stabilization and Destabilization of Emulsion Systems
22
23 Emulsion Stabilization
24
Processed foods are often complex multiphase systems. In the cases where both water and
25
oil are present, emulsification is of course necessary to prevent separation of these two
26
incompatible phases. Emulsion design within the food industry is not a trivial issue. The
27
diversity of manufactured food and beverages means that the relative balance of water and
28
oil phases can vary widely depending on product type, and both oil-in-water (o/w) and
29
water-in-oil (w/o) type emulsions have found a wide variety of applications. Some examples
30
of food emulsions along with concentrations of water and oil are given in Table 3.1.
31
Food emulsions are created and stabilized through a combination of process and formu-
32
lation design. Homogenization facilitates droplet break-up to create the dispersed phase,
33
whilst food ingredients displaying appropriate amphipathic properties are able to adsorb
34
onto the newly formed droplet interfaces during homogenization to provide electrostatic
35
36
37 Table 3.1. Examples of typical food emulsions and their relative concentration of fat
38 Food Emulsion type Fat/oil content (wt%)
39
Milk o/w 0–4
40
Ice creama o/w 0–10
41 Cream o/w 20–50
42 Light mayonnaise o/w 20–50
43 Mayonnaise o/w 65–75
44 Butter w/o 80
Margarine w/o 80
45
46S aIce cream can be considered as a four-phase colloid, comprising dispersed phases of ice, air, and fat in a
47N concentrated continuous phase.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 43

Emulsions as Delivery Systems in Foods 43

(formation of a charged interface) or steric (formation of a viscoelastic interface) stabiliza- 1


tion against immediate coalescence. 2
It can be noted that industrial manufacturing of food emulsions generally employs only 3
a limited number of homogenization technologies, depending on product type. Colloid 4
or Ross mills are commonly used in the manufacture of mayonnaise or similar products 5
with high oil content. High-pressure homogenizers are used in the manufacture of such 6
products as ice cream, (homogenized) milk, and other beverages as well as many other soft 7
solid products of low-to-intermediate fat content. Water-in-oil emulsions, such as mar- 8
garines, are most commonly prepared on votator lines. Other aspects of processing, too, 9
play an important role in the formation of food emulsions, such as pre-homogenization and 10
thermal treatment (pasteurization, sterilization); however, these will not be discussed as 11
part of this chapter. More information on the processing aspects of food emulsions can be 12
found in the literature (Paquin, 1999; Schultz et al., 2004; Perrier-Cornet et al., 2005; 13
Lambrich and Schubert, 2005). The specific role and choice of food ingredients in the sta- 14
bilization (and controlled destabilization) of emulsions will be discussed in the section 15
“Release Triggers for Emulsions.” 16
The most important rule of food emulsion production is that the emulsion should ini- 17
tially be stable. Emulsions are kinetically rather than thermodynamically stable two-phase 18
systems and, ultimately, both oil and water phases will separate. To understand how to opti- 19
mize emulsion stability, it is necessary to understand the mechanisms by which emulsions 20
are destabilized. There are four main mechanisms whereby emulsion phase separation may 21
be accelerated. These are summarized accordingly. 22
23
Creaming/Sedimentation 24
25
For most food emulsions, the oil phase has a lower density than the aqueous phase and can
26
thus separate out due to gravity. For o/w-type emulsions, creaming specifically refers to the
27
motion of emulsion droplets under gravity to form a concentrated creamy layer at the top of
28
the emulsion. Whether or not there is a change in droplet size in this highly concentrated
29
region depends on the stability of the droplets against coalescence. Creaming of poorly sta-
30
bilized emulsions may result in complete breaking of the emulsion layer, resulting in phase
31
separation. For well-stabilized emulsion droplets even an extensively creamed layer can be
32
fully re-dispersed. For w/o-type emulsions, the movement of droplets under gravity is
33
referred to as sedimentation.
34
The rate of creaming for an individual noninteractive spherical droplet, s, can be
defined for highly dilute emulsions through Stokes’ Law: 35
36

S 
(
2 r 2 ρ0  ρ g) 37
38
9η0 39
where g is the acceleration due to gravity, r is the radius of the droplet,  is the density of 40
the dispersed phase, 0 is the density of the continuous phase and 0 is the Newtonian shear 41
viscosity of the continuous phase. From this equation it can be seen that the rate of cream- 42
ing can be reduced by: 43
44
• Reducing droplet size—homogenization of milk typically reduces droplet size from 45
ca. 4 μm in diameter to <1 μm in diameter resulting in a considerable improvement in S46
creaming stability of the milk. N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 44

44 Chapter 3

1 • Increasing the viscosity of the continuous phase.


2 • Density matching the continuous and dispersed phases.
3
4 From a quantitative perspective, the Stokes’ equation applies only to highly dilute, nonin-
5 teracting emulsions and is therefore not applicable to concentrated, polydisperse or aggre-
6 gated emulsion systems. However, the parameters governing creaming rate, as defined by
7 the Stokes’ equation, can of course be applied nonquantitatively to improve creaming
8 stability.
9 The simplest way to measure creaming is by direct observation. However, this relies on
10 the formation of a well-defined layer between cream-rich and cream-depleted regions.
11 Where there is a more diffuse concentration gradient, it may be impossible to directly or
12 accurately detect when creaming is occurring. There are several commercially available
13 techniques that can be used to provide a more accurate analysis of emulsion creaming.
14 These include ultrasound, magnetic resonance imaging and conductivity. The advantage of
15 these techniques is that they are noninvasive and can provide a reasonable approximation of
16 changes to dispersed phase volume across a sample (Dickinson et al., 1994; Dickinson,
17 1996).
18
19 Flocculation
20
21 Flocculation is a general term referring to the various mechanisms for aggregation or
22 association of droplets whereby the interfacial layer of the droplets remains intact
23 (Dickinson, 1998). Generally, for dilute emulsions, flocculation results in enhanced cream-
24 ing, since the flocs structures rise more quickly under gravity relative to individual
25 droplets. However, in more concentrated emulsions, it is possible that flocculation can lead
26 to the formation of a percolating network, which can be controlled to manipulate both sta-
27 bility and rheological properties of the emulsion (Chanamai et al., 2000; Dalgleish, 2006).
28 Flocculation of emulsion droplets takes place when the pair-interaction free energy
29 becomes appreciably negative at a particular separation. This can be achieved through
30 a number of different mechanisms of varying interaction potential. These are briefly
31 summarized.
32
33 Depletion flocculation
34 This is encountered in emulsion systems containing suitable depletants such as non-
35 interacting polysaccharides (e.g. xanthan) or micellar species (e.g. SDS or sodium
36 caseinate) and recently reported to take place in bimodal emulsions, where there is an
37 adequate difference between distributions (Dickinson and Golding, 1997a; Moschakis
38 et al., 2005; Djerdjev et al., 2006). Depletion takes place due to the entropic exclusion
39 of the depletant as droplets approach each other. Due to the exclusion of the depletant,
40 an osmotic pressure gradient forms between droplets resulting in a net attraction thereby
41 leading to flocculation. The strength of the interaction potential is proportional to the sizes
42 of both droplets and depletant as well as the relative concentration of the depletant.
43 Depletion flocculation is termed weak or reversible flocculation, since flocs can be broken
44 up by simple shaking. However, even though the interaction is weak, depletion can result
45 in extensive aggregation of droplets leading to rapid creaming and separation in the case
46S of dilute emulsions, or greatly increased viscosity in the case of more concentrated
47N emulsions.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 45

Emulsions as Delivery Systems in Foods 45

Bridging flocculation 1
Bridging flocculation is a general term to describe the association of emulsion droplets 2
through interfacial interaction. The strength of the interaction depends greatly on the spe- 3
cific nature of the bridging mechanism, which are discussed below. 4
Electrostatic bridging: Droplets stabilized by charged interfacial layers can be bridged 5
through the addition of a species containing counterions. This can be achieved through the 6
addition of an appropriate salt or of a charged biopolymer with the capability of multiple 7
binding sites. Salt bridging requires a minimum divalent ion to enable bridges to form 8
between droplets. An example is the calcium-ion bridging of a casein-stabilized emulsion 9
at neutral pH. In this case, the divalent calcium is able to bind to negatively charged amino 10
acids on the peptide chain of the protein adsorbed onto the interface. This can result in 11
interfacial cross-linking between droplets or cross-linking between droplets and free pro- 12
tein in the continuous phase (Dickinson and Golding, 1998; Ye and Singh, 2001). 13
Alternatively, a biopolymer with opposite charge to the interfacial layer can also behave 14
as an electrostatic bridging link between droplets (Bratskaya et al., 2006). Taking an 15
adsorbed casein interface as an example, at neutral pH, where the net charge at the interface 16
is negative, and a cationic biopolymer will be required to form electrostatic cross-links. 17
(Note: There are very few cationic biopolymers available within the foods industry. 18
Examples include acid/high pI gelatin and chitosan.) However, at pH below the pI of the 19
protein, where the net surface charge is cationic, electrostatic cross-links can be formed 20
using an anionic biopolymer (such as pectin or carrageenan) in the aqueous phase. Electro- 21
static bridging flocculation provides a stronger inter-droplet interaction relative to deple- 22
tion, although high shear can be used to break up aggregated structures. 23
Incomplete surface coverage bridging: This takes place for emulsions stabilized by 24
higher molecular weight emulsifiers, such as proteins. For emulsion droplets to possess 25
good stability, an adequate surface coverage of the droplet surface is required. If no suffi- 26
cient emulsifying material is present to provide good coverage, it is possible that biopoly- 27
mer molecules will become adsorbed to more than one droplet surface, leading to bridging 28
flocculation (Dickinson and Golding, 1997b). A similar effect can be observed when emul- 29
sions are homogenized at very high pressures. Under these circumstances, protein can 30
become adsorbed to more than one droplet surface during homogenization, leading to the 31
formation of an inter-connected protein network. 32
Covalent bridging: Droplets stabilized by biopolymer interfaces can also be cross-linked 33
through covalent bridging mechanisms (Dickinson, 1997; Romoscanu and Mezzenga, 34
2005). There are several routes by which covalent cross-linking can be induced. For 35
example, covalent cross-linking can be thermally induced for protein-stabilized emulsions 36
whereby the adsorbed layer contains accessible disulphide peptides capable of intermolecu- 37
lar cross-linking. The use of high static pressure treatments can also act in a similar manner 38
to induce disulphide cross-linking for appropriate globular protein-stabilized emulsions 39
(Galazka et al., 2000). 40
Enzymatic covalent cross-linking: This is another mechanism by which droplet bridging 41
may take place. One example is the use of the microbial enzyme transglutaminase, which 42
catalyses covalent cross-linking between lysine and glutamine amino acids. Consequently, 43
adsorbed protein interfacial layers which display good availability and accessibility of 44
these two amino acid residues (such as the casein proteins) may become covalently 45
cross-linked. For covalent cross-linking to take place, droplets must be in close proximity. S46
Consequently, for dilute emulsions cross-linking may not occur (or interaction with N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 46

46 Chapter 3

1 cross-linking species present in the continuous phase may take place). At higher dispersed
2 phase volumes, where droplet–droplet separation is sufficiently low for cross-linking
3 to happen, bridging mechanisms tend to result in the formation of a droplet network
4 (Dickinson and Yamamoto, 1996). In this respect, bridging flocculation is used more
5 as a structuring pathway to create emulsion-based gel systems rather than as a cause of
6 instability, per se.
7
8
Coalescence
9
10 Coalescence of emulsion droplets occurs when there is film rupture at the interfaces of
11 adjoining droplets. This leads to the irreversible conjoining of the droplets into a single
12 larger entity. Catastrophic coalescence, such as may take place in a homogenized emulsion
13 in the absence of emulsifier or for poorly stabilized emulsions, can rapidly lead to breaking
14 of the emulsion and partitioning into separate (free) oil and aqueous phases. Where more
15 limited coalescence takes place (such as through controlled shear), the emulsion can then
16 become unstable due to gravity forces enhancing the creaming rate of the larger droplets.
17 For film rupture to take place there must be sufficient film thinning between droplets.
18 Film thinning depends on the relative hydrodynamics within the film, and is dependent on a
19 number of factors, such as the rheological properties of the continuous phase, the concen-
20 tration of the dispersed phase and the effective stabilization of the droplets and their ability
21 to maintain appreciable inter-droplet distance.
22 Interfacial rupture depends on the mechanical properties of the film and the influence of
23 shear and temperature. Emulsion droplets can be stabilized through adsorption of a vis-
24 coelastic and/or charged interface, which is often the case with adsorbed protein, or alter-
25 native biopolymer layers. Increasing interfacial viscoelasticity can provide effective droplet
26 stability against coalescence, even at high applied shear forces. Crystalline interfaces can
27 also provide surface rigidity and effective stabilization against coalescence (Dickinson
28 et al., 1988; Simovic and Prestidge, 2004; Giermanska-Kahn et al., 2005; Tcholakova et al.,
29 2005).
30 Emulsions stabilized with small molecule surfactants do not possess viscoelastic or
31 charge-stabilized (in the case of nonionic emulsifiers) interfaces. For such systems, stability
32 against coalescence is provided through the Marangoni effect, in which surfactant
33 streaming at the point of film thinning leads to an osmotic pressure differential between the
34 film and the surrounding solvent. Consequently, water is drawn into the film gap, thereby
35 preventing further thinning from taking place.
36 An intermediate state of coalescence, termed partial coalescence, is often utilized in the
37 food industry to deliberately induce fat structuring in a number of products, such as ice
38 cream, whipping cream and spreads. In such cases, the interface of the emulsion is
39 designed through formulation, such that it will rupture under appropriate conditions,
40 thereby resulting in coalescence of the emulsion. However, for emulsion systems where the
41 dispersed phase contains a prerequisite level of solid fat, the presence of the solid fat can
42 prevent formation of a single larger entity. Instead, fat droplets form agglomerated struc-
43 tures, sharing a common interface, but in which a degree of the original droplet integrity is
44 maintained. The formation of such agglomerated structures is used to improve the quality
45 of aerated products such as whipped cream and ice cream, by improving the stability of
46S foam structures and reducing drainage (Vanapalli and Coupland, 2001; Coupland, 2002;
47N Hotrum et al., 2005).
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 47

Emulsions as Delivery Systems in Foods 47

Ostwald Ripening 1
2
Ostwald ripening is essentially the growth of larger emulsion droplets at the expense of 3
smaller ones. It may happen in polydisperse emulsions, and takes place due to the fact that 4
the solubility of the oil phase increases with decreasing droplet size. For small droplets this 5
increase in solubility may allow the oil to dissolve and diffuse through the aqueous phase, 6
condensing into larger droplets where solubility is lower. It is difficult to see how the mech- 7
anism could be exploited for encapsulation and controlled release; however, for the inter- 8
ested reader additional information can be found in the literature (Taylor, 1998; Hoang 9
et al., 2004; Meinders and van Vliet, 2004; Mun and McClements, 2006). 10
This section on “Emulsion stabilization” is aimed to highlight the various mechanisms 11
by which emulsions can be destabilized. These are represented in Figure 3.1. Before going 12
on to explore potential routes for formulating food emulsions, it is important to remind our- 13
selves that use of emulsions for controlled delivery and release depends primarily on two 14
things: first, the necessity to stabilize the emulsion against separation prior to delivery; and, 15
secondly, to control destabilization of the emulsion using one or more of the above instabil- 16
ity mechanisms to achieve the required release and delivery under the appropriate physio- 17
logical conditions. 18
19
Formulation Design for Food Emulsions 20
One of the more obvious constraints on the formulation of food emulsions is that all com- 21
ponents should be edible, food-grade and approved for use by international legislation. 22
23
24
Two approaching 25
colloidal particles
26
27
28
29
Interaction between particles Attractive forces highly dominate 30
van der Waals, electrostatic, steric, depletion 31
32
Coagulation
33
Hard spheres 34
35
36
Partial 37
coalescence 38
Solid/liquid 39
Repulsive forces dominate 40
Stable
41
42
Coalescence 43
Emulsions 44
Attractive forces dominate
Flocculation 45
S46
Figure 3.1. Schematic representation of mechanisms for droplet stabilisation and instability. N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 48

48 Chapter 3

1 Whilst this places certain restrictions on the scope of formulation, it has led to considerable
2 creativity with regard to the application of these ingredients for food structuring. The for-
3 mulation of a food emulsion needs to be considered from three perspectives: the design of
4 the aqueous phase, the nature of the dispersed phase and the composition of the interface.
5 The choice of formulation also depends on whether the emulsion is intended to be o/w or
6 w/o (or on occasion w/o/w or o/w/o). For an o/w-type emulsion, options for formulation
7 can be summarized as below.
8
9
10 Design of the Aqueous Phase
11 There are two important aspects to consider when formulating the continuous phase of an
12 o/w food emulsion. The first is microbiological stability, which can be tailored according to
13 the anticipated shelf life of the product. This has less relevance to encapsulation and con-
14 trolled release, although it is important to note that for products designed with a long closed
15 shelf life in mind (>3 months), there should be no change in the emulsion structure, to
16 ensure that the release properties remain consistent over the lifetime of the product. Micro-
17 biological stability can be improved through both thermal treatment (pasteurization/sterili-
18 zation) and aseptic packaging. In addition, emulsions prepared at low pH (<4), high sugar
19 or alcohol content, with low water activity, or containing preservatives will all have
20 improved microbiological stability.
21 The second point of consideration is that manipulation of the continuous phase can have
22 a significant impact on the rheological properties of the emulsion. This can have important
23 consequences on delivery, both in terms of oral response behavior of the emulsion and for
24 the subsequent behavior in the GI tract. Continuous phase properties are very much depen-
25 dent on product type. For example, beverage emulsions are generally low in viscosity, and
26 while it is necessary that the dispersed phase remains stable, there is little requirement for
27 the addition of thickeners or stabilizers. In contrast, a reduced fat mayonnaise requires the
28 addition of aqueous thickeners to the continuous phase in order to compensate for reduced
29 viscosity through the removal of fat.
30 Whilst the addition of biopolymers, such as starch and guar gum, can be used to control
31 aqueous phase rheology independently of the dispersed phase, biopolymer addition can
32 also result in additional rheological manipulation through structuring of the fat phase. This
33 may take place through such effects as depletion flocculation, bridging flocculation or
34 phase separation, and are discussed in the section “Emulsion Stabilization.” A summary of
35 a number of biopolymers available for aqueous phase structuring is given in Table 3.2. This
36 list is not comprehensive and ignores any synergistic effects that may exist for combina-
37 tions of biopolymers.
38 As can be seen from Table 3.2, a number of functional effects can be achieved through
39 aqueous phase structuring. Depending on the nature of structuring, and the potential
40 for aqueous phase biopolymers to interact with the dispersed phase, it is possible to control
41 the release properties of the emulsion, both in terms of emulsion structure failure in mouth
42 (textural response and taste/flavor release) and in terms of emulsion separation under
43 gastric conditions. Examples of how continuous phase behavior can influence release
44 properties will be given in more detail in the sections “Delivery of Water-Soluble
45 Food Actives via Emulsions” and “Delivery of Hydrophobic Food Actives via O/W
46S Emulsions.”
47N
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 49

Emulsions as Delivery Systems in Foods 49

Table 3.2. Examples of aqueous phase structuring ingredients 1


2
Interaction with
Biopolymer type Biopolymer name Effect dispersed phase 3
4
Polysaccharide Guar Thickener, stabilizer None
5
Pectin (low/high methoxy) Stabilizer, thickener, gelling Electrostatic
bridginga and 6
depletion 7
Xanthan Thickener, yield stress Depletion 8
Carrageenan Thickener, stabilizer, gelling Bridginga 9
Locust bean gum Thickener, gelling (cryogelation) None
10
Starch Starch (modified) Thickening, gelation (heat set, cold set) None
Protein Casein (SMP) Thickening, gelation (acid) Bridginga 11
Casein (SMP, caseinate) Gelation (enzyme) Bridginga, 12
depletion 13
Whey Gelation (thermal, covalent) Bridginga 14
Soy Gelation (thermal, covalent) Bridginga
15
Gelatin Gelation (thermal, coil-helix transition) Bridginga
Egg (ovalbumin) Gelation (thermal, covalent) Bridginga 16
17
a Under appropriate interfacial conditions. 18
19
Choice of Lipid Phase 20
21
Many fats and oils are available for use in food emulsions. Whilst the main role of fat in a
22
food product is sensoric, from a product design perspective the use of a particular fat/oil or
23
blending of different lipids can have a significant impact on food emulsion properties.
24
A summary outlining examples of fat types and their composition is given in Table 3.3.
25
Generally a food fat is any triglyceride composition which is solid at room temperature,
26
whilst oil is liquid. For many food emulsions, where the emulsion has been designed to pro-
27
vide structure, solid or hardened fats are often used. Typical examples include ice cream
28
and whipping cream, where the solid fat droplets are able to stabilize air bubbles in the
29
product, as well as generating structure through partial coalescence. This form of fat struc-
30
turing improves product quality through improved stabilization of the air phase and a
31
slower rate of melt. Saturated fats are also utilized extensively in the spreads and margarine
32
industry, where required crystallization is an essential aspect of continuous phase structur-
33
ing and stabilization for w/o-type emulsions. For both these examples it would be impo-
34
ssible to use unsaturated fats to achieve this degree of structuring. In contrast, a product
35
such as mayonnaise uses liquid oils, since it would be impossible to achieve the correct
36
sensory properties with a hardened fat.
37
In addition to structuring, another functional use of the oil phase for an o/w emulsion is
38
as a carrier for lipophilic actives such as flavors, nutrients (e.g. oil-soluble vitamins) and
39
colors. Design of the emulsion system is important for how these actives are stabilized in
40
product and how they are released on consumption. The technical challenges associated
41
with the delivery of lipophilic actives will be discussed in more detail, with examples in the
42
sections “Delivery of Hydrophobic Food Actives via O/W Emulsions” and “Delivery of
43
Dietary Fats as O/W Emulsions and Their Protection against Oxidation”. It should also be
44
noted that w/o and water-in-oil-in-water (w/o/w) type emulsions can be utilized in a similar
45
manner for the controlled delivery and release of water-soluble actives such as vitamins and
S46
enzymes.
N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 50

50 Chapter 3

1 Table 3.3. Examples of common food fats and oils and their melting points (MP) and fat
2 composition (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 of
3
unsaturation
4
5 Other Other
(satu- (unsatu-
6 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)
7
8 Butter fat 32.2 2.5 11.1 29 9.2 2.4 4.8 4.6 26.7 3.6 – 8.5
Lard oil 30.5 – 1.3 28.3 11.9 – – – 74–76 – – –
9 Castor oil −18 0.6 0.6 0.6 0.6 – – – 7.4 3.1 – 87
10 Cocoa 34.1 – – 24.4 35.4 – – – 38.1 2.1 – –
11 butter
12 Coconut oil 25.1 45.4 18 10.5 2.3 0.4 14.6 0.4 7.5 Trace – –
13 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 – –
14 Olive oil −6 – Trace 6.9 2.3 0.1 – – 84.4 4.6 – –
15 Palm oil 35 – 1.4 40.1 5.5 – – – 42.7 10.3 – –
16 Palm kernel 24.1 46.9 14.1 8.8 1.3 – 9.7 – 18.5 0.7 – –
17 Peanut 3 1.92 2 1.4 1.9 2.4 – – 17.8 – 17.5 –
18 Rapeseed −10 – – 1 – – – – 32 15 1 50
Safflower – 1.1 1.1 1.2 1.1 1.2 – – 18.6 70.1 3.4 –
19 Sesame oil −6 – – 9.1 4.3 0.8 – – 45.4 40.4 – –
20 Soybean −16 0.2 0.1 9.8 2.4 0.9 – 0.4 28.9 50.7 – –
21 Sunflower −17 – – 5.6 2.2 0.9 – – 25.1 66.2 – –
22 seed
23
24
25
26 Finally, it should be noted that the choice of fat/oil is also important from a nutritional
27 perspective. Generally, highly saturated fats can have a negative impact on health. Oils with
28 high polyunsaturated triglyceride content are considered preferential in terms of dietary
29 intake, and that oil-containing ω-3 fatty acids are often quoted within nutritional literature
30 as imparting specific health benefits when consumed regularly. However, replacing all satu-
31 rated fats with polyunsaturated oils is not a trivial exercise. As has been mentioned, the
32 degree of saturation can have an impact on the structuring behavior of the emulsion. In
33 addition, highly unsaturated oils are prone to oxidation, which can lead to the development
34 of adverse odors and flavors. Inhibition of oxidative processes within emulsion systems
35 remains one of the key challenges in the move towards a healthier diet, and will be dis-
36 cussed in more detail in the section “Delivery of Dietary Fats as O/W Emulsions and Their
37 Protection against Oxidation.”
38
Interfacial Formulation and Design
39
40 Arguably the most important aspect of emulsion preparation is the composition of the inter-
41 face. Of course, the number of ingredients available for formulation of food emulsions is
42 limited to what can be considered edible and food-grade, and may vary between countries
43 depending on legislation. However, food emulsifiers can broadly be characterized into two
44 categories: low molecular weight species based on fatty acids and high molecular biopoly-
45 mers with amphipathic properties. Examples of food emulsifiers are given in Table 3.4.
46S For all food products, choice of emulsifier is a key to achieving the desired textural and
47N sensory properties. The design of the interface will also play a controlling role in the
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 51

Emulsions as Delivery Systems in Foods 51

Table 3.4. Summary of some food emulsifiers available for stabilization of oil-in-water and 1
water-in-oil food emulsions, including application 2
Name Type Functionality Application 3
4
High-molecular- Skimmed milk Dairy protein O/W emulsion stability Ice cream, yoghurt
5
weight powder at neutral pH, emulsion
biopolymer structuring (bridging) 6
at acid pH 7
Sodium caseinate Dairy protein O/W emulsion Cream liqueurs, 8
stability at neutral pH, meat emulsions 9
emulsion structuring
10
(bridging) at acid pH
Whey powder Dairy protein O/W emulsion stability Neutral pH 11
at neutral pH, emulsion- beverages, infant 12
based gels under formulations 13
thermal processing 14
Gelatin Bovine/Porcine/ Encapsulation, emulsion- Nutritional
15
Fish Protein based gels supplements
Gum arabic Polysaccharide O/W emulsion stability Acid beverages 16
galactan protein at low pH 17
OSA starch Modified starch O/W emulsion stabilizer Acid beverages 18
Low-molecular- Lecithin Phospholipid O/W and w/o stability Spreads, 19
weight depending on mayonnaise 20
emulsifier modification 21
Monoglycerides Glycerol-esterified W/O emulsion Spreads, ice cream,
fatty acids and stabilization and o/w cream liqueurs
22
triglycerides destabilization 23
Sodium stearoyl Lactic acid- O/W emulsion Salad dressings, 24
lactylate esterified stabilization creamers 25
monoglyceride 26
Datem Diacetyl tartaric O/W emulsion Powder mixes,
acid-esterified stabilization gravy
27
monoglyceride 28
Tween Polysorbate- O/W emulsion Desserts, ice cream, 29
esterified stabilization, dressings 30
triglyceride destabilization 31
PGPR Polyglycerol- W/O emulsion Spreads and other
esterified stability oil continuous
32
monoglycerides products 33
34
35
stabilization and breakdown of emulsion structure. High molecular weight amphipathic 36
biopolymers, in particular milk proteins, are very effective at stabilizing o/w-type emulsions. 37
Here, adsorption of casein and whey protein onto the oil–water interface provides effective 38
steric and electrostatic stabilization against coalescence and flocculation. Most dairy-based 39
food emulsions, such as milk, dairy and nondairy creams, ice cream and yoghurt are based on 40
milk protein stabilization of the emulsion. The advantages of using proteins are the nutritional 41
and clean-label aspects associated with proteins. In addition, caseins (essential milk protein 42
fraction) are able to maintain functionality as a result of thermal processing. The disadvantage 43
of proteins is that the peptide interface can be sensitive to changes in pH and ionic composi- 44
tion. For example, acidification of the emulsion towards the isoelectric point of the protein or 45
addition of calcium ions can result in flocculation of the emulsion. In some instances this S46
results in undesirable separation of the emulsion. However, aggregation of dairy emulsions N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 52

52 Chapter 3

1 through pH and calcium is also deliberately used for specific product design, such as in the
2 manufacture of yoghurt or cheese.
3 Where pH sensitivity is an issue for emulsion stability, alternative emulsifiers are avail-
4 able. In particular, for beverage emulsions, where the dispersed phase is dilute and requires
5 effective stabilization, gum arabic or modified starch can be used. In both cases, the struc-
6 ture of the biopolymer allows for effective interfacial stabilization, even at low pH.
7 Low molecular weight emulsifiers do not have the clean-label perception of proteins.
8 However, they are often able to bring specific functional benefits not achieved by proteins
9 alone. Most low molecular weight emulsifiers are based on triglycerides or fatty acids that
10 have undergone an esterification process to produce amphiphilic molecules, whereby the
11 fatty acid chain provides the hydrophobic tail group and the esterified species (such as gly-
12 cerol) provides the more hydrophilic headgroup. Depending on the chain length, degree of
13 (un)saturation of the triglyceride, and the type of headgroup, the amphiphilic properties of
14 emulsifiers can be controlled to a certain degree.
15 This classification of emulsifiers according to their amphiphilic properties is known as
16 the hydrophilic–lipophilic balance (HLB). HLB is a numerical scale for emulsifiers. Emulsi-
17 fiers which are more hydrophobic in nature have low HLB values. Increasing hydrophilicity
18 increases HLB score. For example, polyglycerol polyricinoleate (PGPR), which is a
19 hydrophobic emulsifier used for the stabilization of w/o-type emulsions, has an HLB value
20 of 2. In contrast, polysorbate emulsifiers, which are commonly used for the stabilization of
21 o/w emulsions, have HLB values of around 16.
22 The choice of an emulsifier for a product is very much dependent on the required func-
23 tionality imparted by the emulsifier. Emulsifiers are mainly used for emulsion stabilization
24 as well as its controlled destabilization in food products. Other uses include functional
25 applications such as fat crystal modification, aeration, wetting, and the formation of stable
26 mesophases. The other main application of emulsifiers, often but not always in combination
27 with emulsion systems, is as delivery vehicles for active components. The use of interfacial
28 design for both biopolymer- and emulsifier-stabilized interfaces will be discussed in more
29 detail in subsequent sections.
30
31
Release Triggers for Emulsions
32
33 To be able to design food emulsions for encapsulation and delivery, it is necessary to iden-
34 tify where and how delivery will take place.
35 For delivery of actives that are intended to impart a sensory benefit of the product, delivery
36 takes place on consumption. Behavior of the active in-mouth is therefore of considerable
37 importance. Mouth is a remarkable receptor for sensory perception. Our preference or
38 dislike for certain foods and drinks is driven by three key attributes:
39
40 1. Taste and aroma perception.
41 2. Texture perception: design of food microstructure is responsible for the initial textural
42 perception of a foodstuff.
43 3. Our like or dislike of a food may derive not only from the initial texture of the product
44 but also from the way in which it breaks down in the mouth. Consequently, when
45 designing food structure, it is necessary to take into account the manner in which the
46S product microstructure breaks down on eating. This is often an issue for fat replacement
47N in foods. It may be possible to replace the fat in a product with aqueous fillers such as
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 53

Emulsions as Delivery Systems in Foods 53

starch, such that the initial rheological properties of the two products are the same. 1
However, the breakdown of the two different structures during mastication may result in 2
the two products being texturally perceived entirely differently. The breakdown of 3
microstructure in the mouth will also influence both taste perception and flavor percep- 4
tion. The breakdown of food structures in the mouth is dependent on the applied shear 5
forces during mastication, dilution of the structure with saliva and the fact that in-mouth 6
temperature is generally higher than ambient temperatures ex-vivo. In addition, amylase 7
present in saliva will rapidly start the digestion of starch in the mouth, which may 8
impact on texture. Physical changes in microstructure on eating, such as melting point 9
transitions (fat, gelatin), phase inversions (w/o to o/w emulsions), break-up of aggre- 10
gated emulsion structures, can not only influence textural response, but also be used to 11
control the release of actives, such as tastants or flavors, in the mouth. 12
13
Alternatively, delivery of an active for nutritional benefit will need to take place after con- 14
sumption. Consequently, the release properties of the emulsion/active under gastric condi- 15
tions will be paramount. In the GI tract, emulsions face a low pH (around 2.0) and pepsin in 16
the stomach and lipases and bile salts in the beginning of the small intestine. Lipophilic 17
actives may then be absorbed by the human body in the intestine via dietary micelles. Fur- 18
thermore, since nutritional actives often possess negative taste attributes (e.g., bitterness or 19
astringency), there may also be the additional requirement that release of the active in- 20
mouth should be minimized as much as possible and that the active should be encapsulated 21
in such a way that it is undetectable on consumption. 22
Clearly, in designing emulsions with particular delivery and release properties, the phys- 23
iological conditions of the delivery site will need to be taken into consideration. For in- 24
mouth and gastric conditions the delivery environment will be quite different. 25
26
27
Delivery of Water-Soluble Food Actives via Emulsions 28
Water-in-Oil Emulsions for Controlling Water-Soluble Actives 29
30
Most spreads containing 40% or more fat are fat-continuous products with the aqueous 31
phase being dispersed as w/o emulsions. One of the key requirements for a good spread is 32
phase inversion during breakdown in the mouth (see also “Release Triggers for Emul- 33
sions”), thereby releasing, for example, salt. Margarines that do not do that are perceived as 34
waxy and with very little taste. 35
In principle, stable w/o emulsions that do not invert in the mouth should be able to trap, 36
for example, bitter hydrophilic molecules (such as proteins) in the water phase without 37
being perceived. 38
39
Effect of O/W Emulsions on Taste Release and Perception 40
41
In foods containing both water and lipids, the lipid phase may also take part in the sensory 42
perception by influencing the distribution of food components in the oil and aqueous 43
phases or at the oil—water interface (Yamamoto and Nakabayashi, 1999). Similarly, fats 44
can modify the perception of other sapid food components by influencing their partitioning 45
between the food matrix, saliva, taste receptors (for taste), or headspace (for aroma) within S46
the oral cavity (Forss, 1973). Traditionally the majority of flavor studies in emulsion-based N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 54

54 Chapter 3

1 foods have focused on aroma release and little has been reported on the effect of fat on taste
2 perception. Shamil and co-workers (1991) found that the reduction of fat in cheese led to an
3 increase in bitterness and astringency and a reduction in saltiness. Increased bitterness and
4 astringency were assumed to arise from the hydrophobic character of these ingredients and
5 the resultant increase in their concentration in the aqueous phase when the fat level was
6 reduced. Conversely, the decrease in salt intensity when the fat level was reduced was
7 proposed to be due to the reduced concentration of salt in the aqueous phase. Wendin
8 (Wendin, et al., 1999) also reported that a decrease in fat content led to a decrease in sour
9 taste due to the reduced concentration of the acid in the aqueous phase. Earlier studies have
10 shown a correlation between oil mouth coatings and taste perception (Lynch et al., 1993).
11 Valentova and Pokorny (1998) also found that the intensity of sweet, bitter, and astringent
12 compounds was reduced by the prior consumption of oil, but acidic and salty tastes were
13 not affected. Lynch and co-workers (1993) found that all the taste modalities were affected,
14 albeit by a small amount, and that coconut oil had a more suppressive effect than sunflower
15 oil and proposed that fat mouth coatings physically interfere with tastant access to the taste
16 receptors.
17 Yamamoto and Nakabayashi (1999) concluded from their work on the effects of increas-
18 ing oil-phase volume on salt perception in o/w emulsions that taste intensity will be influ-
19 enced by a combination of an increased concentration of salt in the aqueous phase and a
20 suppressed contact of salt with taste receptors.
21 Overall, however, the results cited in the literature on the effects of oil on tastants have
22 been inconclusive but suggest that oils and emulsions can influence taste perception. The
23 taste-modifying effects of fats may be mediated through:
24
25 1. Changes in the partitioning of flavor (taste) compounds between the food (e.g., changes
26 in the aqueous phase concentration as the fat level is altered), saliva and the taste recep-
27 tor cell membranes and pores.
28 2. Physical interference with diffusion processes affecting access or binding of taste mole-
29 cules to receptors through a mouth coating action by the oil.
30 3. Changes in the rate of regeneration of interfacial surfaces required for the release of
31 sapid compounds into the surrounding media.
32
33 In order to test these ideas, Malone et al. (2003) investigated the relationship between taste
34 perception and microstructure and specifically the effect of fat content in o/w emulsions on
35 salt perception. Their studies involved a sensory analysis of salt perception on isoviscous
36 o/w emulsions with fat contents ranging from 0 to 95%. The results showed that as the fat
37 content was increased for products kept at constant salt concentration, the perceived salti-
38 ness increased due to the associated increase in the salt concentration in the aqueous phase
39 (Figure 3.2).
40 However, when the salt concentration on aqueous phase was kept constant the saltiness
41 decreased with increasing fat content (Figure 3.2), suggesting that perceived saltiness is
42 nonlinearly dependent on a combination of salt concentration and aqueous phase volume.
43 Based on their results a psychophysical model was developed that related taste intensity to
44 salt concentration and the phase volume of fat:
45
46S
I  k [C ]np (φaq )n [1  exp(mφaq )]
47N
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 55

Emulsions as Delivery Systems in Foods 55

1
2
3
4
5
6
7
8
9
10
11
12
13
14
Figure 3.2. Effect of oil phase volume in o/w emulsions on salty taste perception for 15
constant salt concentration on product or on aqueous phase.
16
17
where I is the taste intensity, k is a constant, n is a power-law number, Cp is the concentra-
18
tion of tastant on product, aq is the aqueous phase volume and m is the amount of tastant in
19
the aqueous phase.
20
Indeed, through this insight, water-in-oil-in-water duplex emulsions have been designed
21
to control taste perception by effectively controlling the extent to which the aqueous phase
22
is able to interact with the oral surfaces (further described in the section “Control of Taste
23
Using W/O/W Emulsions”).
24
25
Double Emulsions for Controlling Water-Soluble Actives 26
Double emulsions (also known as “duplex emulsions”) have potentially promising applica- 27
tions in the food industry, primarily for sustained release of active components via a con- 28
trolled transport mechanism (Matsumoto and Kang, 1989; Garti, 1996, 1997a, 1997b, 29
1998; Garti and Aserin, 1996a, 1996b; Garti and Bisperink, 1998; Garti and Benichou, 30
2001), for taste masking (Malone et al., 2003) and for encapsulating sensitive ingredients 31
such as flavors and active components (Kim and Lee, 1999; Yoshida et al., 1999; Edris and 32
Bergnståhl, 2001; Benichou et al., 2004; Onuki et al., 2004; Shima et al., 2005) in both the 33
water and oil phases (van der Graaf et al., 2005). Two main types of double emulsion can be 34
distinguished: w/o/w emulsions, in which a w/o emulsion is dispersed as droplets in an 35
aqueous phase, and oil-in-water-in-oil (o/w/o) emulsions, in which an o/w emulsion is dis- 36
persed in an oil phase. This latter emulsion is less common primarily due to having few 37
hydrophobic emulsifiers that are food-grade to stabilize water droplets in continuous oil 38
phase (Pays et al., 2002). 39
The advantage of double emulsion technology is in their double compartment structure, 40
in which they act as reservoirs, encapsulating a range of active components. Actives that 41
are water-soluble but insoluble in the oil phase can be entrapped in w/o/w emulsions, and 42
actives that are oil-soluble but insoluble in the water phase can be entrapped in o/w/o emul- 43
sions. Actives that are both soluble in oil and water cannot be “encapsulated.” The encapsu- 44
lated actives may be released subsequently under variable conditions. This will be the main 45
topic covered in this section, although the production and the issues of instability will also S46
be addressed briefly. N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 56

56 Chapter 3

1 Production of W/O/W Emulsion


2
Membrane emulsification is one of the key technologies for producing stable double emul-
3
sions due to the prevailing mild shear stress conditions (van der Graaf et al., 2005). Usually
4
double emulsions are prepared in a two-step emulsification process using two surfactants:
5
a hydrophobic surfactant to stabilize the internal w/o emulsion and a hydrophilic one for
6
the external interface of the oil drops with the aqueous environment. In conventional emul-
7
sification processes high shear stresses are needed to reduce the internal droplet size and
8
droplet size distribution of the emulsion, but this also causes internal streaming in the
9
droplets, which causes more collisions and therefore coalescence of the internal water
10
phase with that of the outer aqueous phase (Muguet et al., 1999; Klahn et al., 2002).
11
Membrane emulsification (Nakashima et al., 1991) is a relatively new method for pro-
12
ducing double emulsions but has added benefits in its low energy consumption, better con-
13
trol of the droplet size and distribution and mildness of processing. In general two methods
14
are used: cross-flow membrane emulsification and pre-mix membrane emulsification
15
(Suzuki et al., 1998). In the latter case a pre-mix is forced through a membrane, which fur-
16
ther breaks up the droplets. In the cross-flow method the to-be-dispersed phase is pressed
17
through a microporous membrane while the continuous phase flows along the membrane
18
surface. Once this primary emulsion is produced, the second emulsification step to produce
19
the o/w/o emulsion can also be carried out using membrane emulsification, which helps
20
prevent rupture of the double emulsion and inversion into a single o/w emulsion. Mem-
21
brane emulsification for the production of single emulsions has been reviewed by a number
22
of authors (Joscelyne and Tragardh, 2000; Charcosset et al., 2004) and for microstructured
23
emulsion systems by Lambrich and Vladisavljevic (2004). Gijsbertsen-Abrahamse et al.
24
(2004) reviewed the current status of membrane emulsification and van der Graaf et al.
25
(2005) have recently reviewed membrane emulsification techniques for the production of
26
w/o/w emulsions and also highlighted new developments in nanoengineered and micro-
27
engineered membranes, such as microsieves to improve the flux of emulsions through the
28
membrane (van Rijn, 2004).
29
30
31 Instability of W/O/W Emulsions
32
33 The main problem with double emulsions though is that they tend to be unstable since they
34 contain more interface and therefore are more thermodynamically unstable than single
35 emulsions. Much literature has been published on this subject since around the mid-1980s
36 (Garti et al., 1994; Garti and Aserin, 1996a, 1996b). Florence and Whitehill (1981)
37 described four possible mechanisms for instability of w/o/w emulsions:
38
39 1. Coalescence (see “Coalescence”) of the internal aqueous droplets
40 2. Coalescence of the oil droplets
41 3. Rupture of the oil film separating the internal and external aqueous phases
42 4. Migration of water (including the water-soluble ingredients) between the internal and
43 external water phases through the oil layer.
44
45 This migration of the water phase discussed above could be via reverse micellar transport; by
46S diffusion across the oil layer, where it is at its thinnest; and transport via hydrated surfactant
47N (for further details see “Transport and Release Mechanisms of Water-Soluble Components”).
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 57

Emulsions as Delivery Systems in Foods 57

An enormous amount of formulations for double emulsions is known in the literature 1


with various types of oil, different fractions of phases and different sorts of surfactants in 2
varying concentrations (van der Graaf et al., 2005). 3
Several methods have been documented in the literature for improving stabilization and 4
slowing solute release (Davis et al., 1985). The methods can be categorized into three main 5
areas: 6
7
1. Stabilization of the internal interface of the inner emulsion 8
2. Selection of appropriate oil phase and components that could structure it and 9
3. Stabilization of the outer interface. 10
11
A range of low molecular weight emulsifiers, oils, co-solvents and co-emulsifiers have 12
been tried (Benichou et al., 2004). Materials that have been investigated also include 13
biopolymers, synthetic graft and comb co-polymers and polymerized emulsifiers that 14
impart steric or mechanical stabilization. Monomeric nonionic emulsifiers (hydrophobic 15
and hydrophilic) have some limitations in terms of emulsion stability and therefore other 16
molecules such as naturally occurring macromolecular materials (e.g., gums and proteins) 17
are of interest (Garti, 1997a, 1997b). Macromolecules such as proteins offer excellent sta- 18
bilization effects through electrostatic repulsion in combination with steric contributions. 19
Proteins, polysaccharides and their blends are natural surface-active biopolymers. Under 20
appropriate conditions these may complex through electrostatic interaction, and the newly 21
formed macromolecular amphiphile can anchor onto oil–water interfaces more strongly 22
(Benichou et al., 2002). Complexation between proteins and polysaccharides at the emul- 23
sion droplet surface can improve steric stabilization. Droplet size can be smaller if the 24
polysaccharide is present during homogenization, and so rate of creaming may be reduced 25
so long as there is no bridging flocculation (Benichou et al., 2002). Combinations of 26
surfactants in the outer water phase have also shown a beneficial effect on stability and 27
these multianchoring flexible amphiphilic surfactants are effective emulsifiers since they 28
can improve the steric stabilization by forming thick multilayered coating on the emulsion 29
droplets and also by providing protection against coalescence by making them resistant to 30
shear (Garti, 1998). 31
32
33
Transport and Release Mechanisms of Water-Soluble Components
34
Two major release mechanisms involved in the release of a water-soluble active such as 35
common salt (NaCl), which is entrapped in the aqueous core of w/o/w double emulsions, 36
have been suggested (Pays et al., 2002). The first one describes the rupture of the thin liquid 37
film separating the internal droplets and the double emulsion surfaces. The second is when 38
the entrapped species diffuses or permeates through the oil membrane. These two mecha- 39
nisms can be controlled by varying the composition and amounts of surfactant in the sys- 40
tem. In w/o/w emulsions stabilized with sodium dodecyl sulfate below its critical micelle 41
concentration (cmc), the release of salt occurs by diffusion across the oil membrane. Water 42
transport also needs to be taken into account and a number of mechanisms have been 43
suggested (Wen and Papadopoulos, 2000, 2001): through the surfactant thin lamellae, by 44
reverse micelles and via hydrated surfactant. Ficheux et al. (1998) identified two types of 45
thermodynamic instabilities that allow the (uncontrolled) release of entrapped actives. The S46
first mechanism involves the coalescence of the inner and outer aqueous phase, through the N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 58

58 Chapter 3

1 rupture of the thin nonaqueous film between them, so that inversion to a single emulsion
2 occurs. This mechanism is best suited for the release of water-soluble components, and the
3 release kinetics can be controlled by the hydrophilic surfactant concentration. Depending
4 on the concentration of surfactant, double emulsions may be destabilized with a time scale
5 ranging from a few months to a few minutes. The second mechanism involves the coales-
6 cence between the smaller droplets inside the oil phase leading to an increase in the internal
7 droplet size and a reduction in number. Interestingly, even if the osmotic pressure is bal-
8 anced between the internal and external phase, electrolytes can still be transported out
9 through the reverse micellar mechanism, which is controlled by the viscosity of the oil
10 phase and the nature of the oil membrane (Garti and Bisperink, 1998). The release rate
11 from double emulsions in general tends to follow first-order kinetics (Garti et al., 1994;
12 Sela et al., 1995; Jage-Lezer et al., 1997) with the release rate being controlled primarily by
13 an increase in the diffusion of the active through the oil phase by the selection of appropri-
14 ate secondary hydrophilic emulsifiers.
15 Delivery of hydrophilic actives in the GI tract has also been studied with w/o/w emul-
16 sions (see “General Applications of W/O/W Emulsions”). The oil layer is supposed to pro-
17 tect the active from inactivation by the digestive process in the GI tract (see also “Release
18 Triggers for Emulsions”). However, osmotic pressures in the GI tract are often not con-
19 trolled and this might limit the use of duplex emulsions.
20
21
General Applications of W/O/W Emulsions
22
23 The potential applications of double emulsion technology are enormous, with primary
24 focus being in the food, cosmetics, medical and pharmaceutical industries. Potential appli-
25 cations have been demonstrated in improved biological availability (Elson et al., 1970;
26 Brodin et al., 1978), delivery of drugs (Pandit et al., 1987), and adsorption of toxic com-
27 pounds (Lata et al., 1987). Garti (1997b) reviewed the progress made up until then with
28 respect to food applications of double emulsions. Since then many potential applications
29 for double emulsions have been well documented and some have been patented (Thill-
30 Francis, 1993; Gaonkar, 1994; Takahashi et al., 1994). In most cases double emulsions are
31 aimed for slow and sustained release or controlled release of active matter from an internal
32 reservoir into the continuous phase. Double emulsions have also been used to improve
33 dissolution and solubilization of insoluble materials. Application of double emulsions in
34 the protection of sensitive and active molecules from oxidation (Gallarate et al., 1999;
35 Kim and Lee, 1999; Yoshida et al., 1999; Edris and Bergnståhl, 2001) has also been investi-
36 gated, and double emulsions used to mask acid taste was described by Malone et al. (2003)
37 (see below).
38
39
Control of Taste Using W/O/W Emulsions
40
41 It has already been mentioned in “Effect of O/W Emulsions on Taste Release and Percep-
42 tion” that the results by Malone et al. (2003) on the effect of oil content on taste perception
43 indicated that the perceived intensity of a tastant is dependent on the oil-phase volume oil
44 so for any given system the taste intensity can be manipulated by making w1/o/w2 duplex
45 emulsions to control the apparent oil. Upon consumption the external w2 phase will be
46S perceived but the internal w1 phase will be shielded from the taste receptors during the time
47N scales of eating.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 59

Emulsions as Delivery Systems in Foods 59

In order to test this hypothesis, a series of w1/o/w2 double emulsions were made having 1
fixed oil contents of 30% w/w and internal w1 phase volumes ranging from 0 to 50% w/w. 2
The concentration of the solutes, in particular the citric acid, was at the same levels in both 3
the w1 and w2 phases in order to remove the osmotic and concentration gradients that nor- 4
mally destabilize the emulsions during storage and to maintain the acid functionality in 5
both phases. Their results showed that as more aqueous phase was incorporated into the oil 6
phase the titratable acidity decreased (Malone et al., 2003). The acid in the internal w1 7
phase was not released from the duplex emulsion over the duration of the experiment 8
(which had typical timescales in minutes). Sensory evaluation showed that the perceived 9
acidity decreased, clearly demonstrating the dependence of taste perception on the volume 10
of aqueous phase coming into direct contact with the mouth (i.e., w2). 11
These results demonstrated that it is possible to manipulate the taste intensity by con- 12
trolling the external w2 phase volume that contacts the taste receptors without resorting to 13
traditional encapsulation approaches. This approach is different in that it provides a means 14
of controlling active release whilst allowing for the thermodynamic stable distribution of 15
the active between the constituent phases of the product. In emulsion-based foods, duplex 16
emulsions also provide the benefit of controlled taste whilst remaining within acceptable 17
textural limits for the product concerned. 18
19
20
Delivery of Hydrophobic Food Actives via O/W Emulsions 21
22
Lipophilic Health Ingredients in O/W Emulsions
23
Oil-in-water emulsions can be utilized to provide consumers with lipophilic health ingredi- 24
ents, such as dietary fat, antioxidants (such as the arytenoids -carotene and lycopene), 25
vitamins (e.g. vitamin E), or sterols. They dissolve in the oil of the o/w emulsion, which 26
may stabilize them during storage and increase their bioavailability. The use of dietary 27
fat and how to protect these against oxidation are discussed in more detail in “Delivery of 28
Dietary Fats as O/W Emulsions and Their Protection against Oxidation.” This later 29
section may also illustrate routes to stabilize lipophilic health ingredients against oxidation 30
(if necessary). 31
Lycopene may have various health benefits, such as antioxidation, induction of cell com- 32
munication and growth control, and lower risk of cancer (Ribeiro et al., 2003 and refer- 33
ences therein). Ribeiro et al. (2003) found that the chemical stability of lycopene was 34
particularly high in orange juice (pH 3.7), in contrast to skimmed milk (pH 6.6) or emul- 35
sions in water (pH 5.7). The authors stated that this result might be due to the antioxidant in 36
the orange juice (ascorbic acid, or vitamin C), presence of iron in the milk, or influence of 37
pH on oxidation. The use of
-tocopherol or of nitrogen strongly inhibited the oxidation in 38
all the three different food systems studied. The use of different emulsifiers (Tween 20, 39
Lamegin ZE609, or lecithin) had little influence on the stability of lycopene. The bioavail- 40
ability of lycopene dissolved in oil is high, in contrast to its crystal form (Ribeiro et al., 41
2003 and references therein). Therefore, o/w lycopene emulsions could be a base to 42
develop functional foods. 43
Plant sterols or phytosterols can reduce serum cholesterol by inhibiting intestinal cho- 44
lesterol absorption (Trautwein et al., 2003). Different mechanisms—such as competing 45
with cholesterol for absorption into the dietary micelles (the vehicles for the transport S46
of lipophilic compounds in the intestine) or for cholesterol transporters, co-crystallization N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 60

60 Chapter 3

1 with cholesterol to form insoluble crystals and interference with the hydrolysis process by
2 lipases and cholesterol esterases—are believed to play a role in this process. About 3 g/day
3 of phytosterols are needed to have a significant effect on cholesterol lowering in the serum.
4 The phytosterols are insoluble in water and poorly soluble in oil. Engel and Schubert
5 (2005) produced a high loading of phytosterols in emulsions by using triglyceride and the
6 emulsifiers lecithin or monoglyceride as crystallization inhibitors. Esterifying phytosterols
7 with fatty acids increases their solubility in oil dramatically and this allows easy incorpora-
8 tion of plant sterol fatty acid esters into food products (both in fat-based products, such as
9 margarine and spreads, and in o/w emulsion-based products, such as yoghurt or milk
10 (Trautwein et al., 2003; Noakes et al., 2005 and references therein).
11
12
Aroma Release from O/W Emulsions
13
14 The concentration of flavor (aroma) reaching the olfactory receptors will be influenced by
15 the structure and composition of the food and by the physiological environment and masti-
16 cation behavior that impacts on the rate of aroma released from the foodstuff (Harrison and
17 Hills, 1997a, 1997b; Harrison, 1998; Malone et al., 2000, 2003). Specifically, the flavor-
18 release kinetics may depend on the flavor concentration, the microstructure and tempera-
19 ture of the food, the occurrence of reversible/irreversible binding, structure breakdown
20 during mastication, mixing with saliva, and most importantly (for lipophilic flavors) the
21 concentration of the fat (Delahunty and Piggott, 1995; Overbosch et al., 1991; Malone
22 et al., 2000, 2003; Le Guen and Vreeker, 2003).
23 Fat is recognised as playing a critical role in influencing many flavor attributes such as
24 flavor quality, flavor release, flavor stability and the masking of off-flavor (McGorrin and
25 Leland, 1994). Previous studies have demonstrated that when the fat content of a product is
26 reduced the release of lipophilic flavors is altered resulting in changes to the intensity and
27 release profiles, which alter the overall flavor balance and acceptability of the product
28 (Overbosch et al., 1991; McGorrin and Leland, 1994; Malone et al., 2000; Doyen et al.,
29 2001). These changes are particularly apparent when the fat content is reduced to below
30 5%, where both the intensity and temporal profile are significantly altered (Malone et al.,
31 2000). These differences arise primarily due to the reduction in absorption of the lipophilic
32 flavors in accordance with simple partition theory.
33 Among all food constituents, lipids (and thus also emulsions) affect aroma release most
34 notably, as they not only lower the aroma partial pressure or the air-product partition coeffi-
35 cient of most of the flavor compounds but also change the time scale of release with vary-
36 ing concentrations (de Roos, 1997; Guichard, 2002; Rabe et al., 2003; McClements, 2005).
37 The higher the lipid content and the lipophilicity of the components (i.e., oil–water parti-
38 tion coefficient, Pow), the stronger the decrease in aroma release (with the exception of
39 highly polar compounds possessing log Pow values 0, such as vanillin or acetic acid, see
40 Leland, 1997). However, this effect is stronger under static conditions than under in vivo
41 and artificial throat conditions (Weel et al., 2004b). Generally, increasing fat concentrations
42 result in decreasing flavor release. The affinity of aroma to a lipid phase depends on its
43 chemical composition, degree of saturation, chain length and sequence of fatty acids incor-
44 porated in a triacylglycerol. Hydrophobicity of the aroma compound is the determining
45 factor for the distribution of aroma in the oil and water phases. Using homologues series of
46S hydrocarbons, aldehydes, ketones and alcohols, Jo and Ahn (1999) found that aroma
47N release decreased linearly with the fat content of the emulsion. The effect was less
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 61

Emulsions as Delivery Systems in Foods 61

pronounced for ketones and greater for hydrocarbons, which can be explained by their di- 1
fferent solubilities in oil. However, hydrogen bonds between aroma and lipids are an addi- 2
tional parameter (as has been demonstrated for, e.g., 1-octen-3-ol and linoleic acid by 3
Le Thanh et al., 1998). Small changes in oil content can have significant effect on the aroma 4
partial pressure or the air-product partition coefficient of lipophilic aroma compounds, in 5
contrast to hydrophilic ones (Guichard, 2002). As a consequence, the overall aroma percep- 6
tion is changed often contributing to an imbalance to the nature of the overall flavor. 7
Studies of the influence of the nature and surface area of an oil–water interface on 8
the volatility of aroma in biphasic systems show that there is no general rule for under- 9
standing the effects of interfaces on the aroma release (Druaux and Voilley, 1997, and refer- 10
ences therein). If the volatile compounds (such as dimethylsulfide) accumulate at the 11
interface, the aroma concentration in the headspace of an emulsified system is significantly 12
reduced compared to a two-phase, nonemulsified system. In the case of sunflower-oil/water 13
emulsions that were stabilized with a sugar ester as the emulsifier, diacetyl displayed a 14
higher volatility in w/o emulsions than in o/w emulsions. The presence of proteins at the 15
oil–water interface of emulsions may induce retention for the compounds with high bind- 16
ing constants (Guichard, 2002). For example, ethyl hexanoate was significantly better 17
released from emulsions containing
-lactalbumin (protein with lower affinity for aroma 18
compounds) than from those with -lactoglobulin. The presence of -lactoglobulin at the 19
oil–water interface increases the resistance to the transfer of hydrophobic aroma com- 20
pounds from oil to water and thus induces a decrease in aroma release (and perception). On 21
the other hand, emulsifier concentrations above the cmc of Tween-80 did not influence the 22
release (Rabe et al., 2003). The results with ionic emulsifiers might be different, due to 23
ionic interactions. 24
In general, droplet size of emulsions normally found in foods (5–100 m) will not affect 25
the aroma release of a food product (Rabe et al., 2003; Weel et al., 2004b; McClements, 26
2005), although divergent results have been reported in the literature about the effect on 27
droplet size on aroma release in vivo (Guichard, 2002; Rabe et al., 2003; Weel et al., 28
2004a). Smaller droplets may lead to faster mass transfer due to increased interfacial area 29
and shorter diffusion distance through the oil droplets. However, the exchange of aromas 30
between the two phases is generally assumed to be extremely rapid and it is the water–air 31
interface that is the rate-limiting step for soft solids containing emulsion droplets in the 32
normal size range (1–100 m). Moreover, recent data showed that only small amounts of 33
volatiles are dynamically released from water within 30 s. Therefore, the re-equilibration 34
process between the lipid and the aqueous phase should not be rate-limiting for the initial 35
release process, as the concentration gradients to be adjusted are very flat. 36
Surveys of food preferences among consumers indicate that the most important 37
attributes of foods are aroma (flavor), appearance, and taste with flavor being the primary 38
basis upon which food is selected and reselected. One aspect of flavor delivery that has had 39
little attention is the control of the temporal flavor release profile (i.e., shape of the flavor 40
delivery curve). An increasingly important market requirement is that a new flavor for a 41
particular product should make a specific perceptual impression, for example a powerful 42
initial impact or a novel sensation during eating. Since the rate and duration at which a fla- 43
vor is delivered influence the perception, there is a rational that by controlling the temporal 44
release profile the perception of that flavor can be manipulated. Factors that are important 45
for flavor delivery in the mouth include the composition and microstructure of the product, S46
dilution and mixing with saliva, changes in temperature, flavor concentration and the N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 62

62 Chapter 3

1 occurrence of reversible/irreversible flavor binding (Overbosch et al., 1991; de Roos and


2 Wolswinkel, 1994; Taylor, 1996; see also “Release Triggers for Emulsions”). Furthermore,
3 mass transfer of flavor in the mouth is affected by the gas and saliva flow rates, the degree
4 of agitation and the temperature—all affected by the food structure and composition
5 (Overbosch et al., 1991; Harrison and Hills, 1997a; Harrison, 1998; van Ruth et al., 2000).
6 Aroma release in the mouth is a nonequilibrium process. Consequently, the maximum
7 headspace concentration of a static system will hardly be reached in the mouth. A number of
8 mathematical and mechanistic physicochemical models have been developed that describe
9 flavor release from solid and semi-solid food matrices during eating (de Roos and Wol-
10 swinkel, 1994; Hills and Harrison, 1995; Harrison and Hills, 1996; Harrison et al., 1998;
11 Wright et al., 2003, Wright and Hills, 2003). These theories have been based on the fact that
12 during mastication, the kinetics of flavor release is primarily dependent on the generation of
13 new surfaces (Harrison, 2000) and that the rate-limiting step is the mass transfer of flavor
14 volatiles across the solid–saliva and saliva–gas interfaces for solid (Hills and Harrison, 1995;
15 Harrison and Hills, 1996; Harrison et al., 1998; Wright and Hills, 2003; Wright et al., 2003)
16 and semi-solid foods (Harrison et al., 1997b; Bakker et al., 1998). Much of the impetus for
17 this work has been to model the link between the perception of flavor intensity and the foods
18 composition, microstructure and breakdown during eating (Wright and Hills, 2003; Wright
19 et al., 2003). Two important factors for controlling flavor delivery have been identified. These
20 are (i) the rate-limiting step for soft-solid materials is the mass transfer of volatiles across the
21 solid–liquid and liquid–gas interfaces (Harrison et al., 1998) and (ii) these rates are propor-
22 tional to the mass transfer coefficient and the interfacial surface area (Hills and Harrison,
23 1995). This implies that by controlling the interfacial surface area by creating new surfaces
24 (through particle breakdown) the temporal flavor profile could be altered.
25 Harrison and Hills developed a mathematical model for the release of flavor volatiles
26 from solid foods based on the two-film stagnant film theory (Hills and Harrison, 1995;
27 Harrison and Hills, 1997a; Harrison et. al., 1998). This took into account the saliva flow,
28 decrease in particle size, increase in new surfaces (surface area) and mixing with saliva
29 during mastication. The expression they used based on Euler’s approximation was
30
31 ⎡ c (t ) ⎤
32 M = hD Asf ⎢ cf − s ⎥ t
33 ⎢⎣ Ksf ⎥⎦
34
35 where M is the total mass of volatile diffusing across the interface, hD is the mass transfer
36 coefficient of a volatile, Asf is the surface area of the saliva–food interface, Ksf is the
37 saliva–food partition coefficient, and cf and cs denote the concentration of flavor in the food
38 and saliva, respectively.
39 In principle, the same physicochemical model could be used to model the flavor released
40 from particles that break down, provided that the model takes into account the different
41 release mechanisms that are involved, each of which is rate determining at a different time.
42 Based on considerations of mass transport between the food surface, the surrounding
43 saliva layer and the gas phase of the breath, these treatments have drawn attention to the
44 importance of describing the changing food surface area and saliva content during the
45 course of mastication.
46S One of the biggest challenges and as yet not really solvable with current model con-
47N straints is developing models that can predict the flavor-release behavior when the kinetics
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 63

Emulsions as Delivery Systems in Foods 63

and mechanism of food structure breakdown are changing and sometimes in dramatic 1
ways. Currently, most models are empirical and generally based on a number of assump- 2
tions that have not been fully tested. For example, these models have so far needed to 3
assume idealized spherical shapes for the food particles, which can be related to surface 4
area measurements. The goal would be to mathematically predict the effect of food struc- 5
ture and composition, material behavior and breakdown during eating on the time-intensity 6
flavor release profile, based on mathematical models requiring fewer approximations. 7
The increasing demand for low calorie foods has engendered much interest in the devel- 8
opment of low-fat and sugar-free foods with the required consumer satisfaction. The difficulty 9
in reduced-fat food lies in the multiple functions that fat plays, as it influences all aspects of 10
food perception including appearance, texture, mouthfeel, and flavor. Fat is a source of fla- 11
vor and also influences flavor character, flavor release, off-flavor, and taste perception. 12
Lowering of fat levels is known to reduce the binding of lipophilic flavors to the food 13
matrix thereby influencing the flavor balance (Overbosch et al., 1991; Shamil et al., 1991; 14
Hatchwell, 1996; Malone et al., 2000; Doyen et al., 2001). Reduction in fat levels not only 15
affects the intensity of the flavor perception but also influences the temporal profile. In 16
high-fat products the initial impact of the flavor is gradual providing a well-balanced flavor 17
profile whereas, in fat-free foods the flavor tends to be intense and transient manifesting 18
itself as an “unbalanced” flavor with an “uneven” release profile. 19
20
21
Structured Emulsions in Hydrogels for Controlled Release of Aromas
22
The microstructure and composition of food affects flavor release since aroma compounds 23
may be adsorbed and absorbed by food components (Kinsella, 1990; Bakker, 1995) or 24
influenced by the material and rheological properties of the food, which affect its break- 25
down during eating (Baines and Morris, 1987; Malkki et al., 1990; Guinard and Marty, 26
1995; Wilson and Brown, 1997). The relative importance of each of these mechanisms 27
varies with the properties of the aroma compounds and the physicochemical properties of 28
the food. Binding phenomena, however, generally involve interactions that are specific to 29
the flavor and composition of the food and are not a versatile or practical means of control- 30
ling flavor release in low-fat foods. Likewise, the rheological and material properties have 31
been demonstrated to influence flavor release but large textural changes are required in 32
order to have a noticeable effect; in addition, the scale of these changes is beyond the tex- 33
tural tolerances that would be acceptable for many products. 34
From the consideration of the partitioning of the flavor between the oil and aqueous 35
phases at equilibrium, it can be shown that a considerable proportion of lipophilic flavors 36
are present in the oil phase, even at relatively low oil phase volumes (0.5–5%). Hence, one 37
strategy to control the release of lipophilic flavors would be to inhibit the rate of mass trans- 38
fer of flavor molecules between the oil phase and the continuous aqueous phase. To do this 39
a new approach was taken in our laboratory in which oil was encapsulated within gel 40
particles (Malone and Appelqvist, 2003). 41
In standard o/w emulsions, the rate of inter-phase transport of small solutes from the oil 42
to the water phase occurs on a millisecond timescale (Wedzicha and Couet, 1995). There- 43
fore, during eating, it would appear that the aqueous phase of low-fat o/w emulsions is rap- 44
idly stripped of its flavor, creating a strong driving force for rapid mass transfer of aroma 45
from the oil phase to the aqueous phase. This rapid replenishment of the aqueous phase is S46
the principal reason for the increase in maximum flavor intensity (FImax) and the rate of N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 64

64 Chapter 3

1 release into the headspace. One of the challenges in developing low-fat products is the
2 design of microstructures that will reduce the rate of release of lipophilic aroma. Malone
3 and co-workers (Malone et al., 2003) have demonstrated that one approach to reducing
4 aroma release in low-fat systems is via incorporating the oil droplets into biopolymer gel
5 particles, termed microstructured emulsions. In such microstructures the oil droplets are
6 enveloped in a gel phase, creating a static diffusion layer around the oil droplets. This
7 increases the path-length through which the aroma must diffuse before coming under the
8 influence of the advective conditions existing within the bulk of the product during eating.
9 The result of these structures was to inhibit the rate at which the lipophilic aromas replenish
10 the continuous phase and reduce the rate of aroma release into the headspace.
11 The principle behind encapsulating oil within a gelled particle was to increase the effec-
12 tive path-length for diffusion into the aqueous continuous environment to reduce the rate of
13 lipophilic flavor release. A model for describing the flavor release from gel particles was
14 developed by Lian et al. (2004). The model relates release rates to the composition
15 (oil/water phase volume) and particle size and takes into account the resistance to mass
16 transfer in both the particle and the bulk liquid phase. It should be noted that this approach
17 differs from conventional encapsulation techniques in that it is the oil and not the aroma
18 that is encapsulated. The aroma is allowed to reach thermodynamic equilibrium in the
19 product, and it is the concentration of lipophilic aroma in the oil phase of the gel particles
20 that forms the basis on which the controlled release is achieved. This is an important dis-
21 tinction because this approach does not attempt to resist the thermodynamic distribution of
22 aroma between the oil and water phases.
23 Release can be triggered by matrix melting in the mouth (e.g. gelatin), enzymatic break-
24 down (amylase hydrolysis of starch) and compression-induced fracture (chewing). Aroma
25 release can take place upon breaking the initial equilibrium of aroma between the oil and
26 water phases. As aroma is stripped from the aqueous phase the system attempts to re-
27 establish the o/w equilibrium by diffusion of aroma from the oil but the additional diffu-
28 sional pathway formed by the surrounding gel increases the half-life (t1/2), which, for
29 diffusion into an infinite sink, can be approximated by
30
31
32 t1/2 =
(
0 . 693r 2 1 + Kow φo )
33  D
2

34
35 where r is the radius of the particle, Kow is the oil–water partition coefficient of the flavor
36 compounds, o is the phase volume of oil in the particle, and D is the diffusion coefficient
37 of the aroma compounds in the particle. From this equation, the key parameters that influ-
38 ence the rate of aroma release are the radius r, the oil content of the particle, and Kow of the
39 aroma species.
40 It has been possible to make microstructured emulsions from a range of biopolymer gels
41 including Ca-alginate, gellan, gelatin, gelatin/gum arabic, agar, and starch. This provides
42 the option to design particles that demonstrate controlled breakdown under physiological
43 conditions during eating (Malone et al., 2003). Hence, by careful selection and design of
44 the gel particle it should be possible to manipulate the shape of the aroma-release profile by
45 the particle breakdown pattern.
46S Other mechanisms that might contribute to changes in flavor-release profiles include
47N brittle/elastic fracture involving syneresis of gel particles. For highly structured foods such
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 65

Emulsions as Delivery Systems in Foods 65

as crackers, flavor release can be triggered by plasticization of the matrix by saliva. 1


Changes in pH, ionic strength, and salt type (Kuhn and Foegeding, 1991) within the mouth 2
environment could also be used to trigger physical disruption of particles to release flavor 3
molecules in a controlled manner. 4
There are a large number of factors such as biopolymer type, concentration, salt and so 5
on that could be adjusted to design structured particles that break down and release under 6
physiological conditions. By doing this, it is possible to manipulate the flavor release pat- 7
terns, so that novel flavor profiles are obtained that may be appealing to the consumer. The 8
ability to predict the effect of varying composition and structure on flavor-release profiles 9
will be of great value to the food industry, particularly when trying to design new sensa- 10
tions. Combining microstructure design and mathematical modeling, it would be possible 11
to formulate foods for a desired flavor profile, taking into account both the composition of 12
food and individual or group differences in mastication behavior. 13
14
15
Delivery of Dietary Fats as O/W Emulsions and Their Protection
16
against Oxidation
17
Dietary fats fall in three main groups: saturated, mono-unsaturated and polyunsaturated. Olive 18
oil is the best-known example of dietary oil that contains predominantly mono-unsaturated fatty 19
acids. Polyunsaturated fatty acids are further divided into two subgroups called ω-6 and ω-3 20
fatty acids. Here, the term ω-6 means that the first double bond in the carbon backbone of the 21
fatty acid occurs in the sixth carbon–carbon bond (counted from the terminal carbon atom 22
opposite the acid group). Similarly, ω-3 fatty acids have their first double bond in the third 23
carbon–carbon bond. Examples of ω-3 fatty acids are
-linolenic acid (ALA), eicosapentaenoic 24
acid (EPA), and docosahexaenoic acid (DHA). ALA is an essential fatty acid (i.e., it is not syn- 25
thesized in the human body and must be obtained from food) and can be found in vegetable 26
sources such as the seeds of flax or wall nut; EPA and DHA are abundantly present in fish oils 27
and other marine oils and are assumed to play an important role in the prevention of cardiovas- 28
cular diseases and several other disorders (Nestel, 2000). Furthermore, DHA has been proposed 29
to play an important role in neural and visual developments of infants (Conner, 2000). Because 30
of their beneficial health properties, ω-3 fatty acids have great potential as functional food ingre- 31
dient (Jacobsen, 2004). Unfortunately, the use of ω-3 fatty acids in foods is limited due to their 32
susceptibility to oxidation. Oxidation is a major cause of quality deterioration in foods contain- 33
ing significant amounts of ω-3 fatty acids and gives rise to changes in, for example, flavor (ran- 34
cidity) and nutritional value. Considerable effort has been made to elucidate the mechanisms of 35
lipid oxidation in bulk oils and emulsions. 36
Lipid oxidation can occur via three mechanisms: autoxidation, photo-oxidation, or 37
enzyme action. Emulsified foods usually do not contain enzymes that catalyze oxidation and 38
therefore the latter mechanism is probably less relevant. Photo-oxidation occurs in the pres- 39
ence of light (visible or ultraviolet) and photosensitizing pigments (such as chlorophyll or 40
riboflavin). Photo-oxidation can be a major cause of quality deterioration and is efficiently 41
controlled by storing foods in the dark. Autoxidation is perhaps the most common mecha- 42
nism in foods. It proceeds via a complex series of free-radical reactions with initiation, pro- 43
pagation, and termination steps (Karel, 1992). Transition metal ions (such as iron or copper) 44
are known to be important catalysts for autoxidation. In theory, these metals are capable of 45
directly breaking down unsaturated lipids (RH) into radicals. This reaction, however, is not S46
believed to be important in initiating lipid oxidation. More likely, oxidation is initiated by N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 66

66 Chapter 3

1 metal-catalyzed decomposition of hydroperoxides (ROOH) (lipid hydroperoxides are found


2 in small quantities in all food oils). For the initiation reaction, it is necessary that metal ions
3 and hydroperoxides are in close proximity. In emulsion systems, hydroperoxides tend to
4 accumulate at the surface of the oil droplets because of their relatively polar nature
5 (McClements and Decker, 2000) and therefore are able to interact with metal ions or other
6 pro-oxidants in the aqueous phase. This interaction leads to decomposition of the hydroper-
7 oxides and formation of highly reactive peroxyl (ROO•) or alkoxyl (RO•) radicals. Once
8 these free radicals have been formed at the droplet surface they will interact with polyunsat-
9 urated lipids in their vicinity. This triggers a complex series of oxidation reactions (the
10 reader is referred to Karel, 1992, for more details). Flavor changes typical for oxidized oil
11 products (rancidity, fishy off-flavors, etc.) result from the formation of secondary oxidation
12 products (such as aldehydes, ketones, alkanes, etc.) (Belitz et al., 2001).
13 Various approaches can be taken to retard lipid autoxidation in food products. Removal of
14 oxygen by packing under vacuum or nitrogen can be effective in certain cases. The use of
15 high quality oils with low levels of hydroperoxides and ingredients with low levels of metal-
16 ion contamination is also important. A well-known strategy of controlling lipid oxidation is
17 by the addition of antioxidants (Frankel, 1996; McClements and Decker, 2000). Antioxi-
18 dants are classified depending on their mechanism of action as either primary or secondary
19 antioxidants. Primary antioxidants are compounds that react with free radicals and which are
20 capable of interrupting lipid oxidation chain reactions. Tocopherols and plant polyphenols
21 are important examples of natural primary antioxidants. Secondary antioxidants can retard
22 lipid oxidation through a number of different mechanisms such as metal chelation, oxygen
23 scavenging, or by replenishing hydrogen to primary antioxidants. Examples of metal chela-
24 tors are ethylenediaminetetraacetic acid (EDTA) and citric acid; various food proteins and
25 polysaccharides are also known for their excellent metal-chelating properties.
26 Recently, the impact of microstructure and interfacial characteristics on the oxidative
27 stability of emulsions has been highlighted (see McClements and Decker, 2000, for an
28 excellent review). Various studies have shown that controlling the type and concentration of
29 emulsifiers at the droplet interface can influence the rate of oxidation. One of the physico-
30 chemical factors that appear to be important is the electrical charge of the interfacial layer.
31 An electrically charged surface will attract oppositely charged ions (counter ions) in the
32 surrounding aqueous phase. A negatively charged surface (anionic emulsifier) will thus
33 attract positively charged metal ions and bring them into close proximity of the lipid sub-
34 strate. This is expected to reduce the oxidative stability of the emulsion. On the other hand,
35 a positively charged emulsifier will repel metal ions from the surface and may thus help
36 to stabilize the emulsion against oxidation. The importance of surface charge was demo-
37 nstrated by Mei et al. (1997) for corn oil-in-water emulsions produced using three different
38 emulsifiers. Oxidation rates (in the initial stage of the process) were found to be largest for
39 an emulsion stabilized by sodium dodecyl sulfate (a negatively charged emulsifier); emul-
40 sions stabilized by Brij 35 (uncharged) or dodecyltrimethylammonium bromide (positively
41 charged) appeared to oxidize at a lower rate (at pH 6.5). The authors suggested that the use
42 of positively charged emulsifiers could be an effective means of retarding iron-catalyzed
43 lipid oxidation. As low molecular weight cationic emulsifiers are not commonly used in
44 foods, it was suggested to use protein stabilized emulsions at a pH below the isoelectric
45 point (pI) of the protein. Under these conditions proteins can form a positively charged
46S interfacial membrane around the oil droplet that will repel metal ions. Most food proteins
47N have isoelectric points in the pH range 4.5–5.5 and thus positively charged emulsion
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 67

Emulsions as Delivery Systems in Foods 67

droplets can only be prepared at relatively low pH (i.e., lower than usually desirable in food 1
emulsions). Gelatin (produced by acid hydrolysis of collagen) is an exception as this pro- 2
tein has a relatively high isoelectric point (pI  7–8). Acid-treated gelatin can thus be used 3
to prepare o/w emulsions with positively charged droplets over a wider range of pH values 4
than is possible with other food proteins (Surh et al., 2006). 5
The ability of positively charged proteins to retard lipid oxidation was studied by Hu 6
et al. (2003a). The authors studied the oxidative stability of salmon oil-in-water emulsions 7
stabilized by different whey proteins (viz.
-lactalbumin, -lactoglobulin, sweet whey, and 8
whey protein isolate). Oxidative stability was greatest at pH values below the isoelectric 9
point of the proteins, which was explained from electrostatic repulsion of metal ions away 10
from the positively charged emulsion droplet surface. The authors noted, however, that the 11
ability of whey proteins to alter oxidation rates is not solely due to charge effects, because 12
the positive charge ( potential) of the emulsion droplets (at pH 3.0) decreased in the order 13
-lactoglobulin >
-lactalbumin > whey protein isolate > sweet whey, whereas the oxida- 14
tive stability decreased in the order -lactoglobulin  sweet whey >
-lactalbumin  whey 15
protein isolate. This suggests that other factors also influence the ability of adsorbed pro- 16
teins to retard lipid oxidation. In a subsequent study the authors compared oxidation rates 17
of corn oil-in-water emulsions stabilized by casein, whey protein isolate, and soy protein 18
isolate (Hu et al., 2003b). The oxidative stability (at pH 3.0) decreased in the order casein > 19
whey protein isolates  soy protein isolate. It was concluded that the magnitude of the posi- 20
tive droplet charge again is not the only factor responsible for differences in oxidative stab- 21
ility and that other membrane properties probably also play a role. One of the factors that 22
might be involved is the thickness of the interfacial membrane: a thick layer at the emulsion 23
droplet interface is assumed to hinder interactions (i.e., acts as a physical barrier) between 24
water-soluble pro-oxidants and lipids inside the emulsion droplets (Silvestre et al., 2000). 25
Caseins form a relatively thick layer on the emulsion droplet interface (as compared to, 26
e.g., whey protein isolate), which might contribute to the lower oxidation rate observed in 27
casein-stabilized emulsions. 28
Another factor of importance is the metal–ion chelation properties of proteins. Villiere 29
et al. (2005) compared the oxidative stability of sunflower oil-in-water emulsions stabilized 30
by bovine serum albumin and sodium caseinate. At pH 6.5, emulsions stabilized by sodium 31
caseinate were found to oxidize faster than emulsions stabilized by bovine serum albumin. 32
The faster oxidation was attributed to the better chelating properties of sodium caseinate 33
(as compared to bovine serum albumin) and to electrostatic interactions that favor position- 34
ing of metal ions at the interface. The authors suggest that proteins with good metal chela- 35
tion properties, such as sodium caseinate, should not be used as emulsifiers in systems 36
containing oxidation sensitive lipids, but preferably should be added to the aqueous phase 37
as a natural antioxidant after the emulsification process. This does not hold for emulsions in 38
which metal ions are deactivated and kept away from the interface by the addition of 39
EDTA; in the presence of EDTA, emulsions stabilized by sodium caseinate appeared to be 40
more stable than emulsions stabilized by bovine serum albumin, which was attributed to 41
free-radical-scavenging properties of sodium caseinate. 42
In protein-stabilized emulsions, usually only a fraction of the proteins adsorbs at the oil 43
droplet interface, whereas the remaining proteins are located in the continuous water phase. 44
If the proteins in the water phase are able to chelate metals ions, they can remove the ions 45
away from the oil droplet and inhibit oxidation. The impact of various continuous phase S46
proteins (viz., soy protein isolate, casein and whey protein isolate) on the oxidative stability N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 68

68 Chapter 3

1 of menhaden oil-in-water emulsions was studied by Faraji et al. (2004). In their experi-
2 ments, continuous phase proteins were removed in a number of “washing” steps and the
3 oxidative stability of washed emulsions was compared to those of nonwashed emulsions.
4 Unwashed emulsions (at pH 7.0) were more oxidatively stable than washed emulsions indi-
5 cating that continuous phase proteins are indeed antioxidative and could be used as an
6 effective means of protecting ω-3 fatty acids. Under the conditions used, soy protein isolate
7 was found to have the greatest antioxidant activity of all proteins tested, that is, larger than
8 casein, which was found to have the largest chelation capacity. The authors suggested that
9 in case of soy, antioxidant activity most likely results from a combination of metal-ion
10 chelation and free-radical scavenging. The latter may be due to the presence of specific
11 amino acids with antioxidant activity (such as free sulfhydryl groups) or antioxidants (e.g.,
12 isoflavones) associated with the soy protein.
13 Klinkesorn et al. (2005) studied the effect of multilayer membranes on the oxidative stab-
14 ility of tuna oil-in-water emulsions. Multilayer membranes were produced by sequential
15 deposition of oppositely charged emulsifiers. First, an emulsion was made by dispersing oil
16 in a solution of an anionic emulsifier (lecithin) and then this emulsion was mixed with a
17 solution of a positively charged polysaccharide (chitosan). This “layer-by-layer deposition
18 technique” could be used to produce cationic and relatively thick emulsion droplet inter-
19 faces. The oxidative stability of emulsion droplets coated by a lecithin-chitosan multilayer
20 was found to be higher than that of emulsion droplets coated with lecithin only. The
21 improved stability is likely due to the cationic nature of the droplets that causes repulsion of
22 the prooxidative metals and possibly also from a thicker interfacial region that reduces
23 interactions between lipids and water-soluble prooxidants. According to the authors, pro-
24 duction of emulsion droplets with a multilayer lecithin-chitosan coating might be an excel-
25 lent technology for protecting labile oils.
26 The previous examples have highlighted the importance of prooxidant location. How-
27 ever, the location of chain-breaking antioxidants can also play a critical role in stabilizing
28 emulsions (Frankel, 1996; McClements and Decker, 2000; Chaiyasit et al., 2005). Chain-
29 breaking antioxidants are expected to be most effective at retarding lipid oxidation when
30 they are located in the oil–water interfacial region, where oxidation reactions are initiated.
31 Hydrophilic antioxidants, in general, are less effective than lipophilic antioxidants in o/w
32 emulsions. This is because a significant portion of the hydrophilic antioxidant will partition
33 into the aqueous phase, where it is considered to be inactive (Schwarz et al., 2000). The
34 effectiveness of chain-breaking antioxidants in general increases as their polarity
35 decreases, because they are then more likely to be localized in the lipid phase or near the
36 lipid surface (Huang et al., 1996a, 1996b, 1997).
37 The importance of the electrical charge of chain-breaking antioxidants (relative to the
38 charge of emulsion droplets) was demonstrated by Mei et al. (1997). The authors measured
39 oxidation rates for salmon oil-in-water emulsions stabilized by anionic surfactants (sodium
40 dodecyl sulfate) or uncharged surfactants (Brij 35) containing negatively charged,
41 uncharged, or positively charged phenolic antioxidants. In emulsions stabilized by sodium
42 dodecyl sulfate (at pH 7), the negatively charged antioxidants were found to be less effect-
43 ive than the positively or uncharged antioxidants, which suggests that the negatively
44 charged antioxidants are electrostatically repelled from the surface of the emulsion
45 droplets. In emulsions stabilized by Brij 35, the uncharged phenolic antioxidants were
46S found to be most effective, which was thought to result from the low solubility of
47N uncharged phenolic antioxidants (as compared to the charged phenolics) and a tendency to
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 69

Emulsions as Delivery Systems in Foods 69

accumulate at the oil–water interface. Physical properties, such as polarity and partitioning 1
between different phases, are thus important criteria in selecting a proper antioxidant sys- 2
tem. However, as mentioned by Huang et al. (1997), other criteria such as relative oxidative 3
stability and hydrogen-donating ability in different phases should also be considered in the 4
selection of antioxidants. 5
The literature on oxidation in real food products (e.g., fish-oil enriched mayonnaise, 6
margarine, or milk drinks) is still relatively limited (Jacobsen, 2004). Most studies so far 7
have concentrated on model emulsion systems. The knowledge gained from model studies 8
is expected to lead to new product opportunities. In particular, the possibility of designing 9
interfacial properties (“interfacial engineering”) will enable food scientists to engineer 10
foods with improved oxidative stability. 11
12
13
Future Trends
14
Current efforts are focusing on naturalness, convenience, and perfection. The use of “natu- 15
ral emulsions” and the production of monodispersed emulsions are discussed here. The use 16
of nanoemulsions will be discussed in Chapter 2 in this book. 17
18
19
Nature-Made Emulsions
20
Nature-made emulsions can be used when purified or reconstituted. The idea here is to 21
entrap active components in these pre-formed emulsions. Potentially all plant, animal, and 22
microbial cells can be used and as with all release devices selection will be dependent on 23
the ability of the system to deliver the required release characteristics against a particular 24
application. Three types of preformed capsule systems will be briefly discussed here, oil or 25
lipid bodies, yeast cells, and plant cells. Their use may enhance the “natural” image of a 26
food product, in addition to other functional advantages. 27
28
29
Oil or Lipid Bodies
30
Seed oil bodies (Figure 3.3) are lipid storage organelles of 0.5–2 m in diameter and com- 31
prise a triacylglycerol matrix shielded by a monolayer of phospholipids and proteins. These 32
proteins include abundant structural proteins, oleosins (a structural protein), and at least two 33
minor proteins caleosin (a calcium-binding protein) and steroleosin (an NADP-dependent 34
sterol-binding protein) (Chen et al., 2004). Native oil bodies—modified and reconstructed— 35
can be a useful structure for a range of applications especially as a carrier for hydrophobic 36
molecules. 37
The layer of oleosin coating imparts stability to the oil body by protecting the phospho- 38
lipid monolayer both from attack by the phosphorlipases present in the cell and by giving 39
the oil body a negatively charged surface, which prevents the oil bodies from aggregating 40
and stops coalescence if the structures touch (Tzen and Huang, 1992). In fact oil bodies are 41
remarkably stable both in and out of the cell due to steric hindrance and electronegative 42
repulsion provided by the oleosins on the surface of the oil bodies (Tzen et al., 1992). 43
Oleosins are insoluble in aqueous media, have a pI of 5.7–6.6 and make up 8–20% of the 44
total seed protein (Murphy, 1993; Huang, 1996). 45
It is thought that the oil body size is determined by the ratio of oil to oleosin during oil S46
body formation (Murphy, 1999), which means that it could be possible to control the size of N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 70

70 Chapter 3

1
(a) (b)
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
Figure 3.3. Confocal scanning light microscopic images of an intact pine tree seed cell (left) in
17
the presence of Nile Blue. The dotted line represents the cell wall. Purified oil bodies could be
18 isolated from these cells (right). The light grey spheres in both images depict the oil core of the
19 oil bodies. The white colour represents the protein containing cell structures (hardly visible in
20 the right picture). These pictures have been kindly provided by our colleagues C.M. Beindorff
21 and E. Drost of Unilever R&D Vlaardingen, The Netherlands.
22
23
24 oil body by controlling the rate that oleosin is produced. The nature of the oil within the oil
25 body can also be important both for determining the types of actives that can be encapsu-
26 lated and for the specific application in foods and pharmaceuticals.
27 During normal extraction of oil from plant materials the oil bodies are normally
28 destroyed due to the high shear processes of crushing and milling followed by degumming
29 and further refining (Gunstone et al., 1994). In the last ten years a number of companies
30 (e.g., Sembiosys) have developed methods to extract oil bodies from seeds or plants with-
31 out destroying them and in good yield. A number of papers and patents have been published
32 concerning the specific use of oil bodies for therapeutic and nutraceutical purposes by
33 attaching active peptides to the termini of the oleosin protein and using the oil body as a
34 carrier of the active component concerned (Boothe et al., 1997; Deckers et al., 1998, 1999).
35 This type of research has also stimulated many workers in the field to look at a number of
36 ways in which oil bodies can be modified to make them more functional. This has included
37 improving the payload of lipophilic material by extracting all of the oil from the oil body to
38 leave an empty ghost (Tzen and Huang, 1992; Tzen et al., 1998), which can be later filled
39 with a combination of different oils and actives. These regenerated oil bodies possess the
40 same physiochemical properties as the original oil bodies but now possess higher payloads
41 of active.
42 Oil bodies have also been modified to target specific sites for the delivery of an active by
43 modifying the oleosin proteins, which due to their high level of functional groups make
44 them very susceptible to alterations. Much is already known about the genetics of different
45 plant species, and genetically modified oil bodies have already been produced in which, for
46S example, -glucuronidase enzyme has been fused to an oil body and shown to be active
47N
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 71

Emulsions as Delivery Systems in Foods 71

(Abenes et al., 1997). Other forms of modification to the oil bodies have been via chemical 1
modification (cross-linked with glutaraldehyde or genipin) to enhance their stability (Peng 2
et al., 2003) and self-assembling targeting systems, in which oil bodies can be targeted 3
effectively to their site of action via multivalent antigen-binding proteins (Frenken et al., 4
1999) since antibodies are easily raised to oleosin (Cummins and Murphy, 1992; Wu 5
et al., 1997). 6
Since the constituents of native oil bodies and their proportions are well known, it has 7
been possible to produce stable artificial oil bodies technically reconstituted from their 8
three main components: triglycerols, phospholipids, and oleosin protein (Tzen and Huang, 9
1992; Tzen et al., 1998; Tai et al., 2002). Artificial oil bodies were successfully reconsti- 10
tuted with various compositions of these components and compared to native oil bodies for 11
size and stability. Increasing the size of the oil body led to a decrease in the thermostability 12
and structural stability of the reconstituted oil bodies. 13
Native oil bodies, modified and reconstructed, can be a useful structure for a range of 14
applications especially as a carrier for hydrophobic molecules such as flavors, vitamins, 15
nutraceutical actives (e.g., antioxidants) and pharmaceutical drugs (e.g., steroids), and cos- 16
metic lipids (e.g., healthy fatty acids) (Peng et al., 2003). Other applications are as a vehicle 17
for the production of recombinant proteins (van Rooijen and Moloney, 1995), as a biocap- 18
sule for encapsulation of lactic acid bacteria in dairy products (Hou et al., 2003) and the use 19
of artificial oil bodies reconstituted with olive oil and phospholipid in the presence of cale- 20
osin to elevate the bioavailability of hydrophobic drug cyclosporin A via oral administra- 21
tion (Chen et al., 2005). 22
23
24
Yeast Cells
25
Yeast cells have been explored recently by a number of workers for their potential as con- 26
trolled delivery devices for flavor release (Bishop et al., 1998; Normand et al., 2005) and to 27
improve the bioavailability of poorly soluble drugs in the GI tract (Nelson et al., 2006). 28
Indeed, yeast cells have been investigated as early as the 1970s when Laboratoires 29
Sérozym, France (Laboratoires Sérozym, 1973) and Swift and Co., USA (Shark, 1977) 30
patented a technique using specially prepared yeast cells containing >40% loading of lipid. 31
They described the encapsulation of dyes, drugs, and flavors in viable and nonviable 32
microorganisms including fungi and protozoa. The mechanism of the encapsulation 33
process in yeast cells relies on the relative affinity of would-be encapsulated material for 34
the internal lipid phase of the yeast cell. Flavor components which display ideal solution 35
with this lipid phase will be encapsulated to the greatest degree. 36
It has been suggested that the internal lipid phase is primarily made of phospholipid 37
bilayer membranes unlike a classic micelle structure. Actives which are extensively 38
nonpolar (such as -carotene) might be expected to exist in the interior of the micelle 39
(Wedzicha, 1988); however, their molecular size would involve geometric changes to 40
the micelle and therefore very high molecular weight hydrocarbons may be excluded from 41
the cell. 42
Rebalancing flavors, the use of co-encapsulates to alter the properties of the internal 43
lipid phase to compensate for disproportionate uptake, and other cell modifications such as 44
extraction of the cell wall using detergents (Chow and Palecek, 2004) to improve perme- 45
ability have helped extend the allocation range of the yeast cells as preformed capsules. S46
N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 72

72 Chapter 3

1 Indeed, yeast cell wall composition and thickness can be modified using different cell
2 strains for enzyme expression or by mutating genes involved in cell wall biosynthesis or
3 degradation (Chow and Palecek, 2004).
4 Under dry conditions (e.g., water activity below 0.7), release rates are considerably
5 low due to limited mass transfer. Flavour release can be resumed upon rehydration
6 (Normand et al., 2005). Normand et al. (2005) have used limonene as a model marker for
7 hydrophobic flavors and discussed the flavor-release mechanism with regard to the cell wall
8 structure and its behavior toward water uptake and also desorption during the drying of the
9 yeast cells. The basis of the driving force for flavor release from hydrated yeast cells
10 appears in good agreement with the theory describing monolithic solution release, a theory
11 derived by Crank (1956) and applied to spherical controlled-release devices by Baker and
12 Lonsdale (Baker and Lonsdale, 1974; Baker, 1987) demonstrating a biphasic release
13 pattern. Importantly, the resistance to transfer of flavor materials within the hydrated yeast
14 cell is not rate-determining, and the kinetics of release are dictated by the aqueous phase
15 solubilities.
16
17
Plant Cells
18
19 A plant cell in nature is surrounded by a cell wall and therefore not prone to allowing
20 macromolecules from outside to accumulate within the cell (Rosenbluh et al., 2004).
21 Indeed, cells are protected from the surrounding environment by plasma membrane, which
22 is impenetrable for most hydrophilic and hydrophobic materials. However, it would appear
23 that a process resembling cell endocytosis, which occurs in animals, can also occur in plant
24 cells (Robinson et al., 1998; Daelemans et al., 2002) although much less is known about the
25 detailed mechanism. It has been shown that the addition of macromolecules that have been
26 biotinylated such as hemoglobin, BSA or IgG to cultured soybean cells resulted in their
27 intracellular accumulation (Horn et al., 1990, 1992) and that this process was temperature
28 dependent indicating a requirement for metabolic energy.
29 There are, however, certain low molecular weight proteins that appear able to cross the
30 plasma membrane at least for mammalian cells without the involvement of the endocytic
31 pathway (Lindgren et al., 2000) and have been termed “cell-penetrating protein/peptides”
32 (CCP). These types of molecules such as purified core histones (Rosenbluh et al., 2004) are
33 also capable of crossing plasma membranes of plant cells and acting as CCPs in plant cells.
34 These molecules can be used to mediate the internalization of larger molecules such as
35 oligonucleotides, peptides, proteins, and nanoparticles following their conjugation to the
36 CCP (Fawell et al., 1994; Pooga et al., 1998; Astriab-Fisher et al., 2002). In plant cells it has
37 been confirmed using confocal laser-scanning microscopy that histone-BSA conjugates
38 have penetrated into protoplasts of petunia plants via direct translocation through the
39 plasma membrane (Rosenbluh et al., 2004). This type of technology therefore gives an
40 approach that could be used to introduce and deliver a whole range of actives and macro-
41 molecules into plant cells. Although in the biotechnology area, the internalization of CPPs
42 and the attached molecules by plant cells may open up a new method for transfection in
43 plant cells (Mae et al., 2005), this method could also be used to load plant cells with active
44 molecules such as flavors, vitamins, and so on to be used as controlled delivery devices.
45 Due to the plasma membrane and cell wall structures, plant cells make excellent preformed
46S capsules that can contain a range of macromolecules in a very natural system, which can be
47N used in a range of foods.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 73

Emulsions as Delivery Systems in Foods 73

Monodispersed Emulsions 1
2
Several technologies have been developed to produce highly uniform emulsion droplets 3
(see Link et al., 2004, and references therein). Technologies to reduce polydispersity of 4
already formed emulsions include repeated fractionation and shearing immiscible fluids 5
between uniformly separated plates (Mabille et al., 2003). Alternatively, single-drop tech- 6
nologies are available, such as flow through a micromachined comb, hydrodynamic flow 7
focusing through a small orifice, and drop break off in co-flowing streams (Figure 3.4). 8
Using microchannel technology, more-complex droplet structures have been prepared: 9
w/o/w emulsions (Okushima et al., 2004; Sugiura et al., 2004), gelled beads with a variety 10
of shapes (Seo et al., 2005; Dendukuri et al., 2006), Janus particles where the two halves 11
present different properties (Nisisako et al., 2004), and a variety of encapsulates. Currently, 12
these single-drop technologies are limited in production rate (in the order of l–ml per 13
hour). Highly parallel production at a small scale by microfluidic technology may reduce 14
this limitation in the future. 15
Monodispersed emulsions may have a more defined behavior and release pattern of 16
entrapped actives than polydispersed ones. This can be very important in pharmaceutics 17
and when the emulsions are used as a template to make new materials for, for example, 18
electronics. Currently, it is not clear whether or not this would constitute a real advantage in 19
food systems. Using these technologies may allow forming a better picture of the rheologi- 20
cal and organoleptic behavior of monodispersed emulsions by experimentally testing their 21
properties. 22
23
24
25
26
27
28
(a) Microchannel (100 μm width) 29
30
Oil flow Tip
31
32
Water flow 33
34
Oil flow 35
36
(b)
37
38
39
40
41
42
43
44
Figure 3.4. Emulsion production via microfluidic technology. Here a so-called psi-junction is
used. Other geometries are possible as well. (a) shows the schematic overview and (b) is a 45
microscopic “real” picture that has been kindly provided by Conchi Pulido de Torres, Unilever S46
R&D Colworth, UK. N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 74

74 Chapter 3

1 References
2
Abenes, M., Holbrook, L., and Moloney, M. 1997. Transient expression and oil body targeting of an Arabidopsis
3 oleosin-GUS reporter fusion protein in a range of oilseed embryos, Plant Cell Rep. 17(1), 1–7.
4 Astriab-Fisher, A., Sergueev, D., Fisher M., Shaw, B.R., and Juliano, R.L. 2002. Conjugates of antisense oligonu-
5 cleotides with the Tat and antennapedia cell-penetrating peptides: effects on cellular uptake, binding to target
6 sequences, 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 above
7
and below C*, Food Hydrocolloids 1, 197–205.
8 Baker, R.W. 1987. Controlled release of biologically active agents. Wiley-Interscience, New York.
9 Baker, R.W., and Lonsdale, H.K. 1974. Controlled release of biologically active agents. A.C. Tanquary and R.E.
10 Lacey (Eds) Plenum Press, New York, 15–22.
11 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.
12
Bakker, J., Boudaud, N., and Harrison, M. 1998. Dynamic release of diacetyl from liquid gelatin in the headspace,
13 J. Agri. Food Chem. 46, 2714–2720.
14 Belitz, H.D., Grosch, W., and Schieberle, P. 2001. Lehrbuch der lebensmittelchemie. Springer, Heidelberg.
15 Benichou, A., Aserin, A., and Garti, N. 2002. Protein–polysaccharide interactions for stabilization of food emul-
16 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
17
entrapment and slow release of active matters, Adv. Colloid Interface Sci. 108–109, 29–41.
18 Bishop, J.R.P., Nelson, G., and Lamb, J. 1998. Microencapsulation in yeast cells, J. Microencapsul. 15(6),
19 761–773.
20 Boothe, J.G., Saponia, J.A., and Parmenter, D.L. 1997. Molecular farming in plants: Oilseeds as vehicles for the
21 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-
22
sions by hydrophobically modified chitosan derivatives, Colloids Surfaces A-Physicochem. Eng. Aspects
23 275(1–3), 168–176.
24 Brodin, A.F., Kavaliunas, D.R., and Frank, S.G. 1978. Prolonged drug release from multiple emulsions, Acta
25 Pharmacol. 45, 135–136.
26 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.
27
Food Chem. 53, 4982–4988.
28 Chanamai, R., Herrmann, N., and McClements, D.J. 2000. Probing floc structure by ultrasonic spectroscopy, vis-
29 cometry, and creaming measurements, Langmuir 16(14), 5884–5891.
30 Charcosset, C., Limayem, I., and Fessi, H. 2004. The membrane emulsification process—a review, J. Chem. Tech-
31 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 oil
32
bodies with triacylglycerol, phospholipid, and caleosin, J. Agric. Food Chem. 52, 3982–3987.
33 Chen, M.C.M., Wang, J.L., and Tzen, J.T.C. 2005. Elevating bioavailability of cyclosporine A via encapsulation in
34 artificial oil bodies stabilised by caleosin, Biotechnol. Prog. 21, 1297–1301.
35 Chow, C.K, and Palecek, S.P. 2004. Enzyme encapsulation in permeabilised Sacchraromyces cerevisiae cells.
36 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.
37
Coupland, J.N. 2002. Crystallization in emulsions, Current Opinion Colloid Interface Sci. 7(5–6), 445–450.
38 Crank, J. 1956. The mathematics of diffusion. Oxford University Press, London.
39 Cummins, I., and Murphy, D.J. 1992. cDNA sequence of a sunflower oleosin and transcript tissue specificity, Plant
40 Mol. Biol. 19(5), 873–876.
41 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.
42
Acad. Sci. USA. 99, 14440–14445.
43 Dalgleish, D.G., 2006. Food emulsions—their structures and structure-forming properties, Food Hydrocolloids
44 20(4), 415–422.
45 Davis, S.S., Hadgraft, J., and Palin, K.J. 1985. “Medical and pharmaceutical applications of emulsions”, in
46S Becher, P. (Ed.) Encyclopedia of emulsion technology, Volume 2, Applications. New York: Marcel Dekker.
159–238.
47N
de Roos, K.B. 1997. How lipids influence food flavor, Food Technol. 51(1), 60–62.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 75

Emulsions as Delivery Systems in Foods 75

de Roos, K.B., and Wolswinkel, K. 1994. “Non-equilibrium partition model for predicting flavour release in the 1
mouth”, in Maarse, H., van der Heij, D.G. (Eds) Trends in flavour science. Elsevier, Amsterdam. 15–32. 2
Deckers, H., Moloney, M., and Baum, A. 1999. The case of recombinant production of pharmaceutical proteins in
plants, Annu. Rep. Med. Chem. 34, 237–245.
3
Deckers, H.M., van Rooijen, G., Booth, J., Goll, J., Moloney, M., and Mahmoud, S.S. 1998. Uses of oil bodies, 4
WO9853698. 5
Delahunty, C.M., and Piggott, J.R. 1995. Current methods to evaluate contribution and interactions of components 6
to flavour of solid foods using hard cheese as an example, Int. J. Food Sci. Tech. 30, 555–570. 7
Dendukuri, D., Pregibon, D.C., Collins, J., Hatton, T.A., and Doyle, P.S. 2006. Continuous-flow lithography for
high-throughput microparticle synthesis, Nature Materials 5(5), 365–369.
8
Dickinson, E. 1996. Biopolymer interactions in emulsion systems: Influences on creaming, flocculation, and rhe- 9
ology, ACS Symp. Series 650, 197–207. 10
Dickinson, E. 1997. Enzymatic crosslinking as a tool for food colloid rheology control and interfacial stabiliza- 11
tion, Trends Food Sci. Technol. 8(10), 334–339. 12
Dickinson, E. 1998. Structure, stability and rheology of flocculated emulsions, Curr. Opinion Colloid Interface
Sci. 3(6), 633–638.
13
Dickinson, E., and Golding, M. 1997a. Depletion flocculation of emulsions containing unadsorbed sodium 14
caseinate, Food Hydrocolloids 11(1), 13–18. 15
Dickinson, E., and Golding, M. 1997b. Rheology of sodium caseinate stabilized oil-in-water emulsions, J. Colloid 16
Interface Sci. 191(1), 166–176. 17
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.
18
Dickinson, E., and Yamamoto, Y., 1996. Rheology of milk protein gels and protein-stabilized emulsion gels cross- 19
linked with transglutaminase, J. Agric. Food Chem. 44(6), 1371–1377. 20
Dickinson, E., Ma, J.G., and Povey, M.J.W. 1994. Creaming of concentrated oil-in-water emulsions containing 21
xanthan, Food Hydrocolloids 8(5), 481–497. 22
Dickinson, E., Murray, B.S., and Stainsby, G. 1988. Coalescence stability of emulsion-sized droplets at a planar
oil–water interface and the relationship to protein film surface rheology, J. Chem. Soc. Faraday Trans. I 84,
23
871–883. 24
Djerdjev, A.M., Hunter, R.J., and Beattie, J.K., 2006. Self-depletion flocculation of tetralin oil-in-water emulsions, 25
Langmuir 22(1), 84–87. 26
Doyen, K., Carey, M., Linforth, R.S.T., Marin, M., and Taylor, A.J. 2001. Volatile release from an emulsion: Head- 27
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,
28
Trends Food Sci. Tech. 8, 364–368. 29
Edris, A., and Bergnståhl, B. 2001. Encapsulation of orange oil in a spray dried double emulsion, Nahrung Food 30
45(2), 133–137. 31
Elson, L.A. Mitcheley, B.C.V., Collings, A.J., and Schneider, R. 1970. Chemotherapeutic effect of a water-oil- 32
water emulsion of methotrexate on the mouse L1210 leukemia, Revue Europeene détudes Cliniques et
Biologiques 15, 87–90.
33
Engel, R., and Shubert, H. 2005. Formulation of phytosterols in emulsions for increased dose response in func- 34
tional foods, Innovative Food Sci. Emerg. Technol. 6, 233–237. 35
Faraji, H., McClements, D.J., and Decker, E.A. 2004. Role of continuous phase protein on the oxidative stability 36
of fish oil-in-water emulsions, J. Agric. Food Chem. 52, 4558–4564. 37
Fawell, S., Seery, J., Daikh, Y., Moore, C., Chen, L.L., Pepinsky, B., and Barsoum, J. 1994. Tat-mediated delivery
of heterologous proteins into cells, Proc. Natl. Acad. Sci. USA. 91, 664–668.
38
Ficheux, M.F., Bonakdar, L., Leal-Calderon, F., and Bibette, J. 1998. Some stability criteria for double emulsions, 39
Langmuir 14, 2702–2706. 40
Florence, A.T., and Whitehill, D. 1981. Some features of breakdown in w/o/w multiple emulsions, J. Colloid Inter- 41
face Sci. 79(1), 243–256. 42
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.
43
Frenken, L.G.J., Howell, S., Ledeboer, A.M., and van der Logt, C.P.E. 1999. Multivalent antigen-binding proteins, 44
WO9923221 (Galazka et al., 2000). 45
Gallarate, M., Carlotti, M.E, Trotta, M., and Bovo, S. 1999. On the stability of ascorbic acid in emulsified systems S46
for topical and cosmetic use, Int. J. Pharm. 188, 233–241. N47
Gaonkar, A.G. 1994. Method for preparing a multiple emulsion, US 5322704.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 76

76 Chapter 3

1 Garti, N. 1996. “Delivery of microparticulated liquid systems in food”, in Barenholz, Y., and Lasic, D.D. (Eds)
2 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 Biointerfaces
3 123, 233–246.
4 Garti, N. 1997b. Progress in stabilization and transport phenomena of double emulsions in food applications, Food
5 Sci. Technol. 30, 222.
6 Garti, N. 1998. In Seiller, M., and Grossiord, J.L. (Eds) Multiple emulsions: Structure, properties and applica-
7 tions. Editions de santé, Paris.
Garti, N., and Aserin, A. 1996a. “Double emulsions stabilized by macromolecular surfactants” in Chattopadhyay,
8 A.K., and Mittal, K.L. (Eds) Surfactants in solution. Surfactant Science Series 64. Marcel Dekker, New York.
9 297–332.
10 Garti, N., and Aserin, A. 1996b. Double emulsions stabilized by macromolecular surfactants, Adv. Colloid Inter-
11 face Sci. 65, 37–69.
12 Garti, N., and Benichou, A. 2001. “Double emulsions for controlled release applications: Progress and trends”, in
Sjoblom, J. (Ed.) Encyclopedic handbook of emulsion technology. Marcel Dekker, New York. 377–408.
13 Garti, N., and Bisperink, C. 1998. Double emulsions: Progress and applications, Curr. Opin. Colloid Interface Sci.
14 3, 657–667.
15 Garti, N., Aserin, A., and Cohen, Y. 1994. Mechanistic considerations on the release of electrolytes from multiple
16 emulsions stabilised by BSA and non-ionic surfactants, J. Control. Release 29, 41–51.
17 Giermanska-Kahn, J., Laine, V., Arditty, S., Schmitt, V., and Leal-Calderon, F. 2005. Particle-stabilized emulsions
comprised of solid droplets, Langmuir 21(10), 4316–4323.
18 Gijsbertsen-Abrahamse, A.J., van der Padt, A., and Boom, R.M. 2004. Status of cross-flow membrane emulsifica-
19 tion and outlook for industrial application, J. Membr. Sci. 230, 149–159.
20 Guichard, E. 2002. Interactions between flavor compounds and food ingredients and their influence on flavor per-
21 ception, Food Rev. Int. 18(1), 49–70.
22 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.
23 Gunstone, F.D., Harwood, J.L., and Padley, F.B. 1994. The lipid handbook. Chapman and Hall, London.
24 Harrison, M., 1998. Effect of breathing and saliva flow on flavor release from liquid food, J. Agric. Food Chem. 46,
25 2727–2735.
26 Harrison, M. 2000. Flavour Release, Roberts, D.D., and Taylor, A.J. (Eds) American Chemical Society.
27 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.
28 Harrison, M., and Hills, B.P. 1997a. Effects of air flow-rate on flavour release from liquid emulsions in the mouth,
29 Int. J. Food Sci. Technol. 32, 1–9.
30 Harrison, M., and Hills, B.P. 1997b. Mathematical model of flavour release from liquids containing aroma-bind-
31 ing macromolecules, J. Agric. Food Chem. 45, 1883–1890.
32 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.
33 Harrison, M., Hills, B.P., Bakker, J., and Clothier, T. 1997. Mathematical models of flavor release from liquid
34 emulsions, J. Food Sci. 62, 653.
35 Hatchwell, C. 1996. “Implications of fats on flavours”, in McGorrin, R.J., and Leland, J.V. (Eds) Flavor–food
36 interactions. ACS symposium series 633, Washington DC.
37 Hills, B.P., and Harrison, M. 1995. Two-film theory of flavour release from solids, Int. J. Food Sci. Technol. 30,
425–436.
38 Hoang, T.K.N., Deriemaeker, L., La, V.B., and Finsy, R. 2004. Monitoring the simultaneous Ostwald ripening and
39 solubilization of emulsions, Langmuir 20(21), 8966–8969.
40 Horn, M., Heinstein, P., and Low, P. 1990. Biotin-mediated delivery of exogenous macromolecules into soybean
41 cells, Plant Physiol. 93, 1492–1496.
42 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.
43 Hotrum, N.E., Stuart, M.A.C., van Vliet, T., Avino, S.F., and van Aken, G.A. 2005. Elucidating the relationship
44 between the spreading coefficient, surface-mediated partial coalescence and the whipping time of artificial
45 cream, Colloids and Surfaces A-Physicochem and Eng. Aspects 260(1–3), 71–78.
46S 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-
47N bacillus delbrueckii ssp. bulgaricus in artificial sesame oil emulsions, J. Dairy Sci. 86, 424–428.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 77

Emulsions as Delivery Systems in Foods 77

Hu, M., McClements, D.J., and Decker, E.A. 2003a. Impact of whey protein emulsifiers on the oxidative stability 1
of salmon oil-in-water emulsions, J. Agric. Food Chem. 51, 1435–1439. 2
Hu, M., McClements, D.J., and Decker, E.A. 2003b. Lipid oxidation in corn oil-in-water emulsions stabilized by
casein, whey protein isolate and soy protein isolate, J. Agric. Food Chem. 51, 1696–1700.
3
Huang, A.H.C. 1996. Oleosins and oil bodies in seeds and other organs. Plant Physiol. 110, 1055–1061. In “Water 4
model systems”, J. Agric. Food Chem. 45, 1991–1994. 5
Huang, S.W., Hopia, A., Schwarz, K., Frankel, E.N., and German, J.B. 1996a. Antioxidant activity of α-tocopherol 6
and Trolox in different lipid substrates: Bulk oils vs oil-in-water emulsions, J. Agric. Food Chem. 44, 444–452. 7
Huang, S.W., Frankel, E.N., Schwartz, K., Aeschbach, R., and German, J.B. 1996b. Antioxidant activity of
carnosic acid and methyl carnosoate in bulk oil and in oil-in-water emulsion, J. Agric. Food Chem. 45,
8
2951–2956. 9
Jacobsen, C. 2004. “Developing polyunsaturated fatty acids as functional ingredients”, in Arnoldi, A. (Ed.) Func- 10
tional foods, cardiovascular disease and diabetes. Woodhouse Publishing, Utah. 11
Jage-Lezer, N., Terrisse, I., Bruneau, F., Tokgoz, S, Ferreirra, L., Clausse, D., Seiller, M., and Grossiord, J.L. 1997. 12
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.
13
Jo, C., and Ahn, D.U. 1999. Fat reduces volatiles production in oil emulsion system analyzed by purge and trap 14
dynamic headspace/gas chromatography, J. Food Sci. 64, 641–643. 15
Joscelyne, S.M., and Tragardh, G. 2000. Membrane emulsification—a literature review, J. Membr. Sci. 169, 107–117. 16
Karel, M. 1992. “Kinetics of lipid oxidation”, in Schwartzberg, H.G., and Hartel, R.W. (Eds) Physical chemistry 17
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-
18
ties and in vitro skin permeability, J. Ind. Eng. Chem. 5(4), 306–313. 19
Kinsella, J.E. 1990. Flavour perception and binding, Int. News Fat Oils Relat. Matter 1(3), 215–216. 20
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- 21
ing phase inversion of an emulsion, Colloid surf A–Physicochem Eng. Aspects 210, 167. 22
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-
23
ogy, J. Agric. Food Chem. 53, 4561–4566. 24
Kuhn, P.R., and Foegeding, E.A. 1991. Mineral salt effects on whey protein gelation, J. Agric. Food Chem. 39(6), 25
1013–1016. 26
Laboratoires Sérozym. 1973. Procédé pour faire pénétrer, absorber et/ou fixer par des microorganismes des sub- 27
stances diverses, French Patent no. 72.12622.
Lambrich, U., and Vladisavljevic, G.T. 2004. Emulgieren mit microstrukturierten Systemen, Chem. Ing. Tech.
28
76(4), 376. 29
Lambrich, U., and Schubert, H. 2005. Emulsification using microporous systems, J. Memb. Sci. 257(1–2), 76–84. 30
Lata, M., Nasa, S.L., and Murphy, R.S. 1987. Microencapsulation of salbutamol sulphate using multiple emulsion 31
technique, Eastern pharmacist 30, 137–138. 32
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.
33
Le Guen, S., and Vreeker, R. 2003. “Interactions between flavour compounds and milk proteins under static and 34
dynamic conditions”, in Le Quéré, J.L., and Etievant, P.X. (Eds) Flavour research at the dawn of the twenty-first 35
century. Lavoisier, Cachan, France. 36
Leland, J.V. 1997. Flavor interactions: The greater whole, Food Technol. 51(1), 75–80. 37
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.
38
Lindgren, M., Hallbrink, M., Prochiantz, A., and Langel, U. 2000. Cell-penetrating peptides, Trends Pharmacol. 39
Sci. 21, 99–103. 40
Link, D.R., Anna, S.L., Weitz, D.A., and Stone, H.A. 2004. Geometrically mediated breakup of drops in microflu- 41
idic devices, Phys. Rev. Lett. 92(5), 054503. 42
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.
43
Mabille, C., Leal-Calderon, L., Bibette, J., and Schmitt, V. 2003. Monodisperse fragmentation in emulsions: 44
Mechanisms and kinetics, Europhysics Letters 61(5), 708–714. 45
Mae, M., Myrberg, H., Jiang, Y., Paves, H., Valkna, A., and Langel, U. 2005. Internalisation of cell-penetrating S46
peptides into tobacco protoplasts, Biochimica et Biophysica Biomembranes 1669, 101–107. N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 78

78 Chapter 3

1 Malkki, Y., Heinio, R.L, and Autio, K. 1990. Influence of oat gum, guar gum and carboxymethyl cellulose on the
2 perception 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 lipophilic
3 volatiles during eating, J. Control Release 90(2), 227–241.
4 Malone, M.E., Appelqvist, I.A.M., and Norton, I.T. 2003. Oral behaviour of food hydrocolloids and emulsions—
5 Part 2: Taste and aroma release, Food Hydrocolloids 17, 775–784.
6 Malone, M.E., Appelqvist, I.A.M., Goff, T.C., Homan, J.E., and Wilkins, J.P.G. 2000. “A novel approach to the
7 selective control of lipophilic flavour release in low fat foods”, in Roberts, D.D., and Taylor, A.J. (Eds) Flavour
Release. ACS Symposium Series 763, American Chemical Society, USA, 212–227.
8 Matsumoto, S., and Kang, W.W. 1989. Formation and applications of multiple emulsions, J. Disper. Sci. Technol,
9 10, 455–482.
10 McClements, D.J. 2005 (2nd edn). Food emulsions: Principles, practices, and techniques. CRC Press, Boca
11 Raton, Florida.
12 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.
13 McGorrin, R.J., and Leland, J.V. 1994. Flavour-food interactions. ACS Symposium Series 633, American Chemi-
14 cal Society, USA.
15 Mei, L.Y., McClements, D.J., Wu, J.N., and Decker, E.A. 1997. Iron-catalyzed lipid oxidation in emulsion as
16 affected by surfactant, pH and NaCl, Food Chem. 61, 307–312.
17 Meinders, M.B.J., and van Vliet, T. 2004. The role of interfacial rheological properties on Ostwald ripening in
emulsions, Adv. Colloid Interface Sci. 108, 119–126.
18 Moschakis, T., Murray, B.S., and Dickinson, E. 2005. Microstructural evolution of viscoelastic emulsions sta-
19 bilised by sodium caseinate and xanthan gum, J. Colloid Interface Sci. 284(2), 714–728.
20 Muguet, V., Seiller, M., Barratt, G., Clausse, D., Marty J.P., and Grossoird, J.L. 1999. W/O/W multiple emulsions
21 submitted to a linear shear flow: Correlation between fragmentation and release, J. Colloid Interface Sci. 218,
22 335–337.
Mun, S.H., and McClements, D.J. 2006. Influence of interfacial characteristics on Ostwald ripening in hydrocar-
23 bon oil-in-water emulsions, Langmuir 22(4), 1551–1554.
24 Murphy, D.J. 1993. Structure, function and biogenesis of storage lipid bodies and oleosins in plants, Prog. Lipid
25 Res. 32(3), 247–280.
26 Murphy, D.J. 1999 (2nd Edn). “Plant lipids. Their metabolism, function and utilization”, in Lea, P.J., and Leegood,
27 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 and
28 other tissues, J. Plant Physiol. 158, 471–478.
29 Nakashima, T., Shimizu, M., and Kukizaki, M. 1991. Membrane emulsification by microporous glass, Key Eng.
30 Mater. 61/61, 513.
31 Nelson, G., Duckham, S.C., and Crothers, M.E.D. 2006. Microencapsulation in yeast cells and applications in
32 drug delivery. Polymeric drug delivery, Volume 1 Particulate drug carriers. ACS Symposium Series 923,
268–281.
33 Nestel, P.J. 2000. Fish oil and cardiovascular disease: Lipids and arterial function, Am. J. Clin. Nutr. 71, 228–231.
34 Nisisako, T., Torii, T., and Higuchi, T. 2004. Novel microreactors for functional polymer beads, Chem. Eng. J. 101,
35 23–29.
36 Noakes, M., Clifton, P.M., Doornbos, A.M.E., Trautwein, E.A. 2005. Plant sterol ester–enriched milk and
37 yoghurt effectively reduce serum cholesterol in modestly hypercholesterolemic subjects, Eur. J. Nutr. 44,
214–222.
38 Normand, V., Dardelle, G., Bouquerand, P.E., Nicolas, L., and Johnston, D.J. 2005. Flavour encapsulation in
39 yeasts: Limonene used as a model system for characterisation of the release mechanism, J. Agric. Food Chem.
40 53(19), 7532–7543.
41 Okushima, S., Nisisako, T., Torii, T., and Higuchi, T. 2004. Controlled production of monodisperse double emul-
42 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-
43 sion for intestinal insulin delivery, J. Control. Release 97, 91–99.
44 Overbosch, P., Agterof, W.G.M., and Haring, P.G.M. 1991. Flavour release in the mouth, Food Rev. Int. 7,
45 137–184.
46S Pandit, J.K., Mishra, B., and Chand, B. 1987. Drug release from multiple w/o/w emulsions, Indian J. Pharm. Sci.
47N 49, 103–105.
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 79

Emulsions as Delivery Systems in Foods 79

Paquin, P. 1999. Technological properties of high pressure homogenizers: The effect of fat globules, milk proteins, 1
and polysaccharides, Int. Dairy J. 9(3–6), 329–335. 2
Pays, K., Giermanska-Kahn, J., Pouligny, B., Bibette, J., and Leal-Caldron, F. 2002. Double emulsions: How does
release occur, J. Control. Release, 79(1–3), 193–205.
3
Peng, C.C., Lin, I.P., Lin, C.K., and Tzen, J.T.C. 2003. Size and stability of reconstituted sesame oil bodies, 4
Biotechnol. Prog. 19, 1623–1626. 5
Perrier-Cornet, J.M., Marie, P., and Gervais, P. 2005. Comparison of emulsification efficiency of protein- 6
stabilized oil-in-water emulsions using jet, high pressure and colloid mill homogenization, J. Food Eng. 66(2), 7
211–217.
Pooga, M., Soomets, U., Hallbrink, M., Valkna, A., Saar, K., Rezaei, K., Kahl, U., Hao, J.X., Xu, X.J., Wiesenfeld-
8
Hallin, Z., Hokfelt, T., Bartfai, T., and Langel, U. 1998. Cell-penetrating PNA constructs regulate galanin recep- 9
tor levels and modify pain transmission in vivo, Nat. Biotechnol. 16, 857–861. 10
Rabe, S., Krings, U., and Berger, R.G. 2003. Influence of oil-in-water emulsion characteristics on initial dynamic 11
flavour release, J. Sci. Food Agric. 83, 1124–1133. 12
Ribeiro, H.S., Ax, K., and Schubert, H. 2003. Stability of lycopene emulsions in foods systems, J. Food Sci. 68(9),
2730–2734.
13
Robinson, D.G., Hinz, G., and Holstein, S.E. 1998. The molecular characterization of transport vesicles, Plant 14
Mol. Biol. 38, 49–76. 15
Romoscanu, A.I., and Mezzenga, R. 2005. Cross-linking and rheological characterization of adsorbed protein lay- 16
ers at the oil–water interface, Langmuir 21(21), 9689–9697. 17
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 macromolecules
18
into plant cells, Biochima et Biophysica Acta- Membranes, 1664, 230–240. 19
Schultz, S., Wagner, G., Urban, K., and Ulrich, J. 2004. High-pressure homogenization as a process for emulsion 20
formation, Chem. Eng. Technol. 27(4), 361–368. 21
Schwarz, K., Huang, S.W., German, J.B., Tiersch, B., Hartmann, J., and Frankel, E.N. 2000. Activities of antioxi- 22
dants are affected by colloidal properties of oil-in-water and water-in-oil emulsions and bulk oils, J. Agric. Food
Chem. 48, 4874–4882.
23
Sela, Y., Magdessi, S., and Garti, N. 1995. Release of markers from the inner phase of w/o/w emulsions stabilised 24
by silicone-based polymeric surfactants, J. Control. Release 33, 1–12. 25
Seo, M., Nie, Z.H., Xu, S.Q., Mok, M., Lewis, P.C., Graham, R., and Kumacheva, E. 2005. Continuous microflu- 26
idic reactors for polymer particles, Langmuir 21, 11614–11622. 27
Shamil, S., Wyeth, L.J., and Kilcast, D. 1991. Flavour release and perception in reduced fat foods. Food Qual.
Pref. 3(1), 51–60.
28
Shark, J.L. 1977. Encapsulation process utilizing microorganisms and products produced thereby, US Patent 29
4001480. 30
Shima, M., Tanaka, M., Kimura, Y., Adachi, S., and Matsuno, R. 2005. Enhancement in transport of a hydrophilic 31
marker through intestinal epithelial cell (Caco-2) monolayer by w/o/w multiple emulsion containing C8TG, 32
Food Hydrocolloids 19, 321–328.
Silvestre, M.P.C., Chaiyasit, W., Brannan, R.G., McClements, D.J., and Decker, E.A. 2000. Ability of surfactant
33
head group size to alter lipid and antioxidant oxidation in oil-in-water emulsions, J. Agric. Food Chem. 48, 34
2057–2061. 35
Simovic, S., and Prestidge, C.A. 2004. Nanoparticles of varying hydrophobicity at the emulsion droplet–water 36
interface: Adsorption and coalescence stability, Langmuir 20(19), 8357–8365. 37
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 Interface
38
Sci. 270, 221–228. 39
Surh, J., Decker, E.A., and McClements, D.J. 2006. Properties and stability of oil-in-water emulsions stabilized by 40
fish gelatin, Food Hydrocolloids 20, 596–606. 41
Suzuki, K., Fujiki, I., and Hagura, Y. 1998. Preparation of corn oil/water and water/corn oil emulsions using PTFE 42
membranes, 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 oil
43
bodies and their structural stabilisation to reconstituted oil bodies, Biosci. Biotechnol. Biochem. 66, 44
2146–2153. 45
Takahashi, Y., Yoshida, T., and Takashi, T. 1994. Process for producing a w/o/w-type multiple emulsion for medi- S46
cines, cosmetics, etc., US Patent 4985173. N47
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 80

80 Chapter 3

1 Taylor, A.J. 1996. Volatile flavour release from foods during eating, Crit. Rev. Food Sci. Nutr. 36(8), 765–784.
2 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-
3 tration and pH on the coalescence stability of beta-lactoglobulin emulsions: Experiment and interpretation,
4 Langmuir 21(11), 4842–4855.
5 Thill-Francis, L. 1993. Stable double emulsions containing finely divided particles. WO93/00007.
6 Trautwein, E.A., Duchateau, G.S.M.J.E., Lin, Y., Mel’nikov, S.M., Molhuizen, H.O.F., and Ntanios, F.Y.
7 2003. Proposed mechanisms of cholesterol-lowering action of plant sterols, Eur. J. Lipid. Sci. Technol. 105,
171–185.
8 Tzen, J.T.C., and Huang, A.H.C. 1992. Surface structure and properties of plant seed oil bodies, J. Cell Biol. 117,
9 327–335.
10 Tzen, J.T.C., Lie, G.C., and Huang, A.H.C. 1992. Characterisation of the charged components and their topology
11 on the surface of plant seed oil bodies, J. Biol. Chem. 267, 15626–15634.
12 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.
13 Valentova, H., and Pokorny, J. 1998. Effect of edible oils and oil emulsions on the perception of basic tastes,
14 Nahrung 42(6), 406–408.
15 Vanapalli, S.A., and Coupland, J.N. 2001. Emulsions under shear: The formation and properties of partially coa-
16 lesced lipid structures, Food Hydrocolloids 15(4–6), 507–512.
17 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.
18 van Rijn, C.J.M. 2004. Nano and micro engineered membrane technology. Elsevier, Amsterdam.
19 van Rooijen, G.J.H, and Moloney, M.M. 1995. Plant seed oil bodies as carriers for foreign proteins, Bio/Technol-
20 ogy 13, 72–77.
21 van Ruth, S.M., O’Connor, C.H., and Delahunty, C.H. 2000. Relationships between temporal release of
22 aroma compounds in a model mouth system and their physicochemical characteristics, Food Chem. 71,
393–399.
23 Villiere, A., Viau, M., Bronnec, I., Moreau, N., and Genot, C. 2005. Oxidative stability of bovine serum
24 albumin- and sodium caseinate–stabilized emulsions depends on metal availability, J. Agric. Food Chem. 53,
25 1514–1520.
26 Wedzicha, B.L. 1988. “Distribution of low-molecular-weight food additives”, in Dickinson, E., and Stainsby,
27 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.
28 Weel, K.G.C., Boelrijk, A.E.M., Burger, J.J., Jacobs, M.A., Gruppen, H., Voragen, A.G.J., and Smit, G. 2004a
29 Effect of emulsion properties on release of esters under static headspace, in vivo and artificial throat conditions
30 in relation to sensory intensity, J. Agric. Food Chem. 52, 6572–6577.
31 Weel, K.G.C., Boelrijk, A.E.M., Burger, J.J., Verschueren, M., Gruppen, H., Voragen, A.G.J., and Smit, G. 2004b.
32 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.
33 Wen, L., and Papadopoulos, K.D. 2000. Visualization of water transport in w1/o/w2 emulsions, Colloids Surf.
34 174, 159–167.
35 Wen, L., and Papadopoulos, K.D. 2001. Effects of osmotic pressure on water transport in w1/o/w2 emulsions,
36 J. Colloid Interface Sci. 235, 398–404.
37 Wendin, K, Ellekjaer, M.R., and Solheim, R. 1999. Fat content and homogenization effects of flavour and texture
of mayonnaise with added aroma, Food Sci. Technol. (Lebensmittel Wissenschaft and Technologie) 32,
38 377–383.
39 Wilson, C.E., and Brown, W.E. 1997. Influence of food matrix structure and oral breakdown during mastication on
40 temporal perception of flavour, J. Sens. Stud. 12(1), 69–86.
41 Wright, K.M., and Hills, B.P. 2003. Modelling flavour release from a chewed bolus in the mouth: Part II. The
42 release 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 the
43 mouth: Part I. Mastication, Int. J. Food Sci. Technol. 38, 351–360.
44 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-
45 terization of neutral-lipid-containing organelles and globuli-filled plastids from Brassica napus tapetum, Proc.
46S Natl. Acad. Sci. USA 94, 12711–12716.
47N
Lakkis_Ch_03-041-082 r1.qxd 3/29/07 2:42 PM Page 81

Emulsions as Delivery Systems in Foods 81

Yamamoto, Y, and Nakabayashi, M. 1999. Enhancing effect of an oil phase on the sensory intensity of salt taste of 1
NaCl in oil/water emulsions, J. Food Texture Stud. 30, 581–590. 2
Ye, A.Q., and Singh, H. 2001. Interfacial composition and stability of sodium caseinate emulsions as influenced
by calcium ions, Food Hydrocolloids 15(2) 195–207.
3
Yoshida, K., Sekine, T., Matsuzaki, F., Yanaki, T., and Yamaguchi, M. 1999. Stability of vitamin A in oil-in-water- 4
in-oil-type multiple emulsions, J. Am. Oil. Chem. Soc. 76(2), 195–200. 5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
S46
N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 83

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

4 Applications of Probiotic Encapsulation 1


2
in Dairy Products 3
4
5
Ming-Ju Chen and Kun-Nan Chen 6
7
8
9
10
Introduction 11
Most probiotics in the food supply are used in fermented milks and dairy products; in fact, 12
dairy products are the major carriers of probiotics available today. Probiotics can be defined 13
as living microbial supplements which can improve the balance of intestinal microorgan- 14
isms (Fuller 1992). This definition was broadened by Havenaar and Huis in’t Veld (1992) to 15
a “mono- or mixed-culture of live microorganisms which benefit man or animals by 16
improving the properties of the indigenous microflora.” The probiotic effect has been 17
attributed to the production of acid and/or bacteriocins, competition with pathogens and 18
enhancement of the immune system. Claimed benefits include controlling serum choles- 19
terol levels, preventing intestinal infection, improving lactose utilization in persons who are 20
lactose intolerant, and possessing anticarcinogenic activity. 21
Good probiotic viability and activity are considered essential for optimal functionality 22
(Mattila-Sandholm et al. 2002; Champagne and Gardner 2005). Furthermore, the ability of 23
microorganisms to survive and multiply in the host strongly influences their probiotic ben- 24
efits. The bacteria in a product should remain metabolically stable and active, surviving 25
passage through the upper digestive tract in large numbers sufficient enough to produce 26
beneficial effects when in the host intestines (Gilliland 1989). Adequate numbers of viable 27
cells, namely the “therapeutic minimum,” need to be regularly consumed in order to trans- 28
fer the probiotic effect to consumers. Survival of these bacteria during the product shelf life 29
until being consumed is therefore an important consideration. Suggested beneficial mini- 30
mum level for probiotics in yogurt is 106 cfu/mL (Robinson 1987; Kurman and Rasic 1991) 31
or the daily intake should be about 108 cfu/mL. Earlier studies indicated that some strains 32
of probiotics, especially Bifidobacterium spp., lack the ability to survive gastrointestinal 33
conditions (Berrada et al. 1991; Lankaputhra and Shah 1995). 34
Other studies have also reported low viability of probiotics in dairy products such as 35
yogurt and frozen dairy desserts (Iwana et al. 1993; Shah and Lankaputhra 1997; Schillinger 36
1999) due to the concentration of lactic acid and acetic acid (Samona and Robinson 1994), 37
low pH (Martin and Chou 1992; Klaver et al. 1993), the presence of hydrogen peroxide 38
(Lankaputhra and Shah 1996), and the oxygen content (Dave and Shah 1997). Methods for 39
protecting probiotics including selection of acid-resistant strains, control of over-acidification 40
of dairy products, and the addition of cysteine or an oxygen scavenger such as ascorbic acid 41
(Dave and Shah 1997) have been proposed by various studies (Dave and Shah 1998; 42
Adhikari et al. 2000; Krasaekoopt et al. 2003). Encapsulation has been investigated for 43
improving the viability of microorganisms in both dairy products and the intestinal tract 44
(Prevost and Divies 1988; Lacroix et al. 1990; Champagne et al. 1992). 45
Encapsulation is a physicochemical or mechanical process in which particles containing S46
active ingredients are covered by a layer of another material, providing protection and N47

83
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 84

84 Chapter 4

1 controlled release of the primary ingredients as well as making the ingredients more conve-
2 nient to work with (Thies 1996). The selection of different types of coating materials usu-
3 ally depends on the functional properties of the microcapsules and the coating process used
4 (Hegenbart 1993). For dairy and food applications, probiotic encapsulation in food grade,
5 porous matrices has been most widely used (Champagne et al. 1994). Spherical entrapment
6 beads are produced using spray-drying, extrusion, or emulsification techniques.
7 The following sections describe the techniques, effects, and applications of probiotic
8 encapsulation in dairy products. Published data on new techniques of probiotic encapsulation
9 with survival of probiotic capsules in dairy products and in the intestines are also discussed.
10
11
Techniques for Probiotic Encapsulation
12
13 Encapsulation of probiotics for use in dairy products or biomass production can be
14 achieved in two ways: physicomechanically and chemically. The probiotics are encapsu-
15 lated in the gas phase during physicomechanical procedures including spray-drying tech-
16 nique whereas, probiotic encapsulation is performed in liquid by thermal or ionotropic
17 gelation of the droplets including extrusion and emulsion techniques. All three techniques
18 have been proven to increase the survival of probiotics by up to 90% (Kebary et al. 1998).
19
20
Spray-Drying Technique
21
22 Among the well-known microencapsulation methods, spray-drying is most widely used in
23 the chemical, pharmaceutical, and food industries due to its inherent attributes such as high
24 production rates and relatively low operational cost (Gibbs et al. 1999). The principle of
25 spray-drying technique involves dissolving a polymer, in the continuous phase, which sur-
26 rounds the core material particles (encapsulant such as probiotics) inside the sprayed
27 droplets. The drying process causes this solution to shrink into a pure polymer envelope
28 enclosing the core material. The resulting capsules are obtained as free-flowing dry powder.
29 Table 4.1 shows probiotic encapsulation using the spray-drying technique in dairy prod-
30 ucts and biomass production. Various carrier matrices including starch (O’Riordan et al.
31 2001; Lian et al. 2003), gelatin (Lian et al. 2002, 2003), gum arabic (Lian et al. 2002,
32 2003), skim milk (Gardiner et al. 2002; Lian et al. 2003; Ananta et al. 2005), cellulose
33 acetate phthalate (CAP; Favaro-Trindade and Grosso 2002), whey protein (Picot and
34 Lacroix 2003, 2004), gum acacia (Desmond et al. 2002), and prebiotics (Ananta et al.
35 2005) have been reported and applied to various dairy products including yogurt (Picot and
36 Lacroix 2004), dry dairy beverages (O’Riordan et al. 2001), and cheddar cheese (Gardiner
37 et al. 2002). However, exposure to high air temperatures required to facilitate water evapo-
38 ration during the passage of the bacteria in the spray-drying chamber exerts a negative
39 impact on their viability and hampers their activity in the spray-dried product (Ananta et al.
40 2005). The survival of encapsulated microorganisms produced by spray-drying will be dis-
41 cussed in more detail in a later section.
42
43
Extrusion Technique
44
45 Extrusion is the simplest and most common technique used to produce probiotic capsules
46S with hydrocolloids (King 1995). The principle of this technique simply involves preparing
47N a hydrocolloid solution, adding the probiotic ingredient to the solution and dripping the cell
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 85

Applications of Probiotic Encapsulation in Dairy Products 85

Table 4.1. Probiotic encapsulation by spray-drying in dairy products and biomass production 1
2
Inlet and outlet
Probiotics Carrier matrix (%) temperature Application Reference 3
4
Bifidobacterium PL1 10% starch Inlet: 60–140°C Dry beverage O’Riordan
5
Outlet: 45°C et al. (2001)
6
L. paracasei 20% reconstituted Inlet: 175°C Cheddar Gardiner
skim milk Outlet: 68°C cheese et al. (2002)
7
8
L. acidophilus Cellulose acetate Inlet: 130°C Favaro-Trindade
phthalate and Grosso (2002) 9
B. lactis Outlet: 75°C
10
11
L. paracasei Gum acacia Inlet: 170°C Desmond
Outlet: 95–105°C et al. (2002) 12
B. longum 30% gelatin Inlet: 100°C Lian et al.
13
(2002, 2003) 14
B. infantis 35% soluble starch Outlet: 50–60°C 15
35% gum arabic 16
15% skim milk 17
B. breve 85% milk fat/5–15% Inlet: 160°C Picot and Lacroix 18
whey protein (2003) 19
B. longum 10% whey protein Outlet: 80°C 20
B. breve 85% milk fat/5–15% Inlet: 160°C Yogurt Picot and Lacroix 21
whey protein (2004) 22
B. longum Outlet: 80°C 23
L. rhamnosus GG 20% skim milk/ Outlet: 70–100°C Ananta et al. 24
oligofructose or (2005) 25
polydextrose 26
27
28
29
suspension through a syringe needle or nozzle spray machine in the form of droplets which 30
are allowed to free-fall into a hardening solution or setting bath. This extrusion technique 31
produces large particles with uniform particle size. 32
Table 4.2 shows probiotic encapsulation using extrusion techniques in dairy products and 33
biomass production. The common polymer used to produce probiotic encapsulation matrix 34
by extrusion technique is alginate (Krasaekoopt et al. 2003). Other food-grade encapsulation 35
materials like gellan gum and xanthan gum (Sun and Griffiths 2000; McMaster et al. 2005) 36
have also been proposed for encapsulating probiotics. Many dairy products including yogurt 37
(Prevost and Divies 1987; Sun and Griffiths 2000; Krasaekoopt et al. 2004; Iyer and Kailas- 38
apathy 2005), cheese (Prevost and Divies 1988), and cream (Prevost and Divies 1992) carry 39
encapsulated probiotics produced by extrusion. One of the major advantages of this method 40
is that the viscosity of the fluid does not limit capsule generation (Prüße et al. 2000). Fur- 41
thermore, the biological matter can be treated at lower temperatures. 42
43
44
Emulsion Technique
45
The emulsion technique has successfully been used to encapsulate lactic acid bacteria in S46
both batch (Lacroix et al. 1990) and continuous fermentation processes (Audet et al. 1992). N47
9
8
7
6
5
4
3
2
1

45
44
43
42
41
40
39
38
37
36
35
34
33
32
31
30
29
28
27
26
25
24
23
22
21
20
19
18
17
16
15
14
13
12
11
10

46S
47N
Table 4.2. Probiotic encapsulation by extrusion technique in dairy products and biomass production

86
Probiotics Supporting material (%) Hardening bath Special treatment Aplication Reference

L. delbrueckii 1.85% sodium alginate 1.5 M calcium chloride No Cheese Prevost et al. (1987)
ssp. bulgaricus
Streptococcus
thermophilus
L. plantarum 1.5% sodium alginate 0.05 M calcium chloride No Biomass production Kearney et al. (1990)
L. lactis 2% sodium alginate 0.1 M calcium chloride No Cream Prevost and Divies (1992)
L. casei 0.6% sodium alginate 1.0 M calcium chloride Chitosan Yoo et al. (1996)
1.0 M barium chloride
Lactococcus lactis 2% sodium alginate 0.05 M calcium chloride Chitosan Biomass production Zhou et al. (1998)
ssp. cremoris
B. bifidum 2% sodium alginate 0.2 M calcium chloride Poly-L-lysine Cui et al. (2000)
chitosan
B. longum 2–4% sodium alginate 0.1 M calcium chloride No Lee and Heo (2000)
B. infantis 0.75% gellan/1% 0.1 M calcium chloride No Yogurt Sun and Griffiths (2000)
xanthan gum
L. acidophilus 0.75–2% sodium alginate 0.1–1.0 M calcium chloride No Chandramouli et al. 2004
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 86

L. acidophilus 2% sodium alginate 0.05 M calcium chloride Chitosan Yogurt Krasaekoopt et al. (2004)
L. casei
B. bifidum
L. acidophilus 2% sodium alginate 0.05 M calcium chloride Sodium alginate Krasaekoopt et al. (2004)
poly-L-lysine
chitosan
L. casei
B. bifidum
L. bulgaricus 2% sodium alginate 0.5 M calcium chloride Chitosan Lee et al. (2004)
B. lactis 0.75% gellan/1% 0.1 M calcium chloride No Amasi McMaster et al. (2005)
xanthan gum (sour milk products)
L. acidophilus Sodium alginate Calcium chloride Hi-maize starch Yogurt Iyer and Kailasapathy
poly-L-lysine (2005)
Chitosan Raftiline®/Raftilose®
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 87

Applications of Probiotic Encapsulation in Dairy Products 87

The principle of these techniques is based on the relationship between the discontinuous 1
and the continuous phases. A small volume of the cell-polymer suspension (i.e., the discon- 2
tinuous phase) is added to a large volume of vegetable oil (i.e., the continuous phase). The 3
mixture is then homogenized to form a water-in-oil emulsion. Once the water-in-oil emul- 4
sion is formed, the water-soluble polymer must be insolubilized to form tiny gel particles 5
within the oil phase. The insolubilization method of choice depends on the type of support- 6
ing material used. The beads are harvested later by filtration. For encapsulation in an emul- 7
sion, an emulsifier and a surfactant are needed. Emulsifiers such as Tween 80 can break up 8
water and oil emulsions as well as prevent spheres from coalescing before breaking up the 9
emulsion. A surfactant such as sodium lauryl sulfate (SLS) is used to lower the surface ten- 10
sion in the coating matrix in order to reduce the size of the spheres. 11
Table 4.3 shows probiotic encapsulation using emulsion technique for dairy products 12
and biomass production. 13
Various supporting materials have been used to encapsulate probiotics by the emulsion 14
method including alginate (Sheu and Marshall 1993; Sultana et al. 2000; Truelstrup et al. 15
2002; Song et al. 2003; Shah and Ravla 2004), -carrageenan (Dinakar and Mistry 1994; 16
Adhikari et al. 2000, 2003), CAP (Modler and Villa-Garcia 1993), chitosan, and gelatin 17
(Peniche et al. 2003). This type of probiotic beads have been successfully applied to yogurt 18
(Adhikari et al. 2000; Sultana et al. 2000; Adhikari et al. 2003), cheddar cheese (Dinakar 19
and Mistry 1994), milk (Truelstrup et al. 2002), and ice cream (Sheu and Marshall 1993; 20
Shah and Ravla 2004). This technique provides both encapsulated and entrapped core 21
materials and is easy to scale up for large-scale production. 22
23
24
Advantages and Disadvantages of Various Probiotics
25
Encapsulation Techniques
26
A comparison of different encapsulation techniques is presented in Table 4.4. Both spray- 27
drying and extrusion (Krasaekoopt et al. 2003) are relatively simple techniques. Con- 28
versely, the emulsion technique based on the relationship between the discontinuous and 29
continuous phases is more complex. Although both spray-drying and emulsion techniques 30
are easier to scale up, Picot and Lacroix (2003) used an emulsification/spray technology to 31
produce microcapsules containing micronized skim milk powder dispersed in milk fat 32
droplets surrounded by an insoluble whey protein film. This technique is claimed to be 33
simple and can be easily scaled up for microencapsulation of dry probiotic cultures. 34
Encapsulation of probiotics using natural biopolymers such as calcium alginate, 35
-carrageenan, and gellan gum is currently applicable only on a laboratory scale (Doleyres 36
and Lacroix 2005). The high viscosity of these coating materials appears to hamper the effi- 37
ciency of encapsulation (Krasaekoopt et al. 2003). Scale-up production of encapsulated 38
probiotics via extrusion is more difficult due to the slow formation of beads (Krasaekoopt 39
et al. 2003). 40
The sizes of beads formed from spray-drying and emulsion are smaller than those pro- 41
duced by the extrusion method. With the extrusion method, the size of the capsules is highly 42
dependent on the viscosity of sodium alginate solution, the extruder orifice diameter, and the 43
distance between the syringe and the calcium chloride collecting solution (Smidsrod and 44
Skjak-Braek 1990). A higher concentration of sodium alginate results in significantly high 45
viscosity which leads to large particle sizes. Spherical beads, prepared by extrusion, are S46
approximately 2–3 mm in diameter, while those made by emulsification techniques have N47
9
8
7
6
5
4
3
2
1

45
44
43
42
41
40
39
38
37
36
35
34
33
32
31
30
29
28
27
26
25
24
23
22
21
20
19
18
17
16
15
14
13
12
11
10

46S
47N

88
Table 4.3. Probiotic encapsulation by emulsion in dairy products and biomass production

Concentration of
Probiotics supporting material (%) Continuous phase Special treatment Application Reference

L. bulgaricus 3% -carrageenan/ Soy oil No Biomass production Audet et al.(1989)


locust bean gum
S. thermophilus
Lc. lactis
L. bulgaricus 3% alginate Vegetable oil No Ice milk Sheu and Marshall
(1993)
B. bifidum 2% -carrageenan Vegetable oil No Cheddar Dinakar and Mistry
(1994)
B. longum 3% -carrageenan/ Soy oil No Maitrot et al. (1997)
locust bean gum
B. longum 2% -carrageenan Vegetable oil No Set yogurt Adhikari et al. (2000)
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 88

L. acidophilus 2% alginate Vegetable oil Hi-maize starch Yogurt Sultana et al. (2000)
Bifidobacterium spp.
B. adolescentis 3% alginate Vegetable oil No Milk Truelstrup et al. (2002)
B. breve
B. lactis
B. longum
L. casei 1% alginate Corn salad oil Microporous Song et al. (2003)
Glass Membrane
B. longum 2% -carrageena Vegetable oil No Stirred yogurt Adhikari et al. (2003)
L. bulgaricus Artificial oil Sesame oil/vegetable oil No Hou et al. (2003)
L. acidophilus 3% alginate Vegetable oil No Frozen dessert Shah and Ravla
(2004)
Bifidobacterium spp.
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 89

Applications of Probiotic Encapsulation in Dairy Products 89

Table 4.4. Advantages and disadvantages of encapsulation methods 1


2
Spray-drying Extrusion Emulsion
3
Scale-up Easy More difficult Easy 4
Encapsulating process Simple Simple More difficult
5
Variety of coating materials Many Few Many
Shape and size Uniform and small Uniform and large Non-uniform and small 6
Survival of microorganisms Dependent on the High High 7
carriers used and 8
temperature 9
10
bead diameters ranging from 25 µm to 2 mm. The actual bead size can be controlled by vary- 11
ing the speed of agitation and it also depends on the type of emulsifier used. 12
Probiotics encapsulated via spray-drying technique show lower survival rates during 13
drying and lower stability during storage (Ananta et al. 2005) than those produced by emul- 14
sion and extrusion, a result of their exposure to high air temperatures required to facilitate 15
water evaporation. 16
17
18
Effects of Encapsulation on Probiotic Survival 19
This section summarizes the factors affecting the survival of encapsulated probiotics. 20
21
22
Effect of Carrier Matrix on Probiotic Survival 23
24
Alginate 25
Alginate is a linear heteropolysaccharide of D-mannuronic and L-guluronic acids extracted 26
from various species of algae. The functional properties of alginate as a supporting material 27
are strongly associated with the composition and sequence of L-guluronic and D-mannuronic 28
acids. Divalent cations such as Ca2 preferentially bind to the polymer of L-guluronic acid 29
(Krasaekoopt et al. 2003). Calcium alginate is preferred over all other supporting materials 30
for encapsulating probiotics due to its simplicity, non-toxicity, biocompatibility, and low 31
cost (Sheu and Marshall 1993; Krasaekoopt et al. 2003). Solubilization of alginate gels by 32
sequestering calcium ions and releasing entrapped cells within the human intestines is 33
another advantage. The concentrations of sodium alginate and calcium chloride used to form 34
the beads vary and range between 1 and 3% alginate with 0.05~1.5 M CaCl2 (Prevost et al. 35
1988; Kearney et al. 1990; Cui et al. 2000; Chandramouli et al. 2004; Krasaekoopt et al. 36
2004). A very low level of alginate (0.6% alginate with 0.3 M CaCl2) was used to form a gel 37
by Jankowski et al. (1997). Nevertheless, alginate beads formed using low-viscosity alginate 38
solutions lack mechanical and physical stability (Smidsrod and Skjak-Braek 1990; Peirone 39
et al. 1998). 40
The use of alginate, however, is limited due to its low physical stability in the presence 41
of anti-gelling cations such as sodium and magnesium ions (Lee et al. 2004) or chelating 42
agents such as phosphate (Krasaekoopt et al. 2006). The latter share an affinity for calcium, 43
thus destabilizing the alginate gel (Smidsrod and Skjak-Braek 1990). Furthermore, under 44
low pH conditions, cross-linked alginate matrices can undergo degradation of the alginate 45
molecule and subsequent reduction in its molecular weight causing faster release of S46
entrapped active ingredients (Gombotz and Wee 1998). N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 90

90 Chapter 4

1 Specially Treated Alginates


2
Coating alginate beads with polycations and cross-linking with barium ions (Ba2+) instead
3
of calcium ions (Ca2) have been suggested for improving the mechanical stability of algi-
4
nate microcapsules (Thu et al. 1996; Gaumann et al. 2001; Koch et al. 2003; Krasaekoopt
5
et al. 2006).
6
Polycation-coated alginates: Coating alginate beads with polycations such as chitosan and
7
poly-L-lysine has been studied extensively for encapsulating probiotics (Cui et al. 2000; Canh
8
et al. 2004; Krasaekoopt et al. 2004; Lee et al. 2004; Krasaekoopt et al. 2006). Chitosan-
9
coated alginate capsules were produced by dropping an alginate solution into a mixture of
10
calcium chloride and chitosan solution (Krasaekoopt et al. 2004). Since chitosan (poly-(2-
11
amino-2-deoxy-β-D-glucopyranose)) is positively charged, it forms polyelectrolyte com-
12
plexes with alginates resulting in the formation of polyanionic polymer membranes which
13
are stable in the presence of calcium chelators or antigelling agents (Smidsrod and Skjak-
14
Braek 1990). Zhou et al. (1998) reported that suspending alginate capsules in a low molecu-
15
lar weight chitosan solution reduced cell release by 40%. On the contrary, Lee et al. (2004)
16
indicated that high molecular weight chitosan coating resulted in the highest survival for
17
Lactobacillus bulgaricus in simulated gastric juice and better stability at 22°C. Krasaekoopt
18
et al. (2006) studied the survival of probiotics encapsulated in chitosan-coated alginate
19
beads in yogurt and found that the survival of the encapsulated probiotic bacteria was higher
20
than free cells by approximately 1 log cycle. Lee et al. (2004) indicated that microencapsula-
21
tion of freeze-dried L. bulgaricus by chitosan-coated calcium alginate greatly improved the
22
viability of probiotics in simulated gastric and intestinal juices.
23
Alginate poly-L-lysine microcapsules’ high biocompatibility and strength make them
24
good candidates for food applications (Champagne et al. 1992; Larisch et al. 1994;
25
Krasaekoopt et al. 2004). Bifidobacteria loaded onto alginate poly-L-lysine microparticles
26
displayed enhanced survival of the probiotic bacteria during storage at 4°C (Cui et al.
27
2000). Krasaekoopt et al. (2004) compared the survival of microencapsulated probiotics
28
using different coating materials and found that chitosan-coated alginate beads provide bet-
29
ter protection for Lactobacillus acidophilus and Lactobacillus casei than did poly-L-lysine-
30
coated alginated beads in 0.6% bile salts.
31
Modification of alginates by succinylation (increased matrix anionic charge) or by
32
acetylation (increased matrix hydrophobicity) has also been suggested for stabilizing
33
encapsulated probiotics in acidic conditions (Le-Tien et al. 2004).
34
35
36
Prebiotics-Coated Alginates
37
38 Prebiotics are non-digestible food ingredients that beneficially affect the host by selectively
39 stimulating the growth and/or activity of one or a limited number of bacteria in the colon
40 (Gibson and Roberfroid 1995). Several studies (Bielecka et al. 2002; Chen et al. 2005a) have
41 confirmed that incorporation of prebiotics and calcium alginate as coating materials
42 provides better protection for probiotics in food and eventually the intestinal tract. Chen et
43 al. (2005a) incorporated prebiotics as coating materials for probiotic microencapsulation
44 and demonstrated that the addition of fructooligosaccharides (FSO), isomaltooligosaccha-
45 rides (IMO), and peptides in the walls of probiotic microcapsules provided improved protec-
46S tion for the active organisms. Probiotic counts remained at 106107 cfu g-1 for
47N microcapsules stored for one month and were then subjected to a simulated gastric fluid test
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 91

Applications of Probiotic Encapsulation in Dairy Products 91

and a bile salt test. Iyer and Kailasapathy (2005) reported that addition of Hi-maize starch to 1
capsules containing Lactobacillus spp. provided maximum protection under acidic condi- 2
tion. Moreover, by further coating the capsules with chitosan, the survival rate was signifi- 3
cantly increased under acidic and bile salt conditions. 4
5
6
Gellan Gum and Xanthan Gum
7
Gellan gum, a microbial polysaccharide derived from Pseudomonas elodea, is constituted 8
of a repeating unit of four monosaccharide molecules (glucose, glucuronic acid, glucose, 9
and rhamnose). The combination of gellan and xanthan gums to form bead is not only acid 10
resistant but also is stabilized by calcium ions (Norton and Lacroix 1990), which can pro- 11
tect cells from acid injury. Sun and Griffiths (2000) encapsulated Bifidobacterium spp. with 12
gellan-xanthan gum as the coating material and reported that gellan-xanthan beads were 13
highly acid-stable. At pH 2.5, the viable count of encapsulated probiotics decreased by only 14
0.67log in 30 min. while the survival of free cells dropped from 1.23  109 cfu mL-1 to an 15
undetectable level in the same period. 16
17
18
-Carrageenan and Locust Bean Gum
19
-Carrageenan is a natural polymer extracted from Irish moss and is commonly used in the 20
food industry. Formation of a gel using this polymer occurs because of temperature 21
changes. The cell suspension is mixed with the heat-sterilized polymer solution at 40–50°C 22
and gelation occurs on cooling to room temperature. The microcapsules are stabilized by 23
adding potassium ions. The encapsulation of Bifidobacterium bifidum in -carrageenan 24
beads maintained the cell viability for as long as 24 weeks of cheddar cheese ripening, with 25
no negative effects on the texture, appearance, or flavor (Dinakar and Mistry 1994). How- 26
ever, -carrageenan produces brittle gels which are not able to withstand stresses of inter- 27
nal bacterial growth and shear during agitation (Audet et al. 1988). 28
The combination of -carrageenan with locust bean gum, which produces more flexible 29
gels due to specific interactions between the two gums, was recently used to encapsulate 30
probiotics. The probiotics suspension was mixed with a -carrageenan-locust bean gum 31
solution, and the cell-polymer dispersion was then rapidly poured into vegetable oil with 32
agitation. The beads were washed and soaked in sterile KCl solution. Several researchers 33
(Maitrot et al. 1997; Audet, et al. 1988) combined -carrageenan with locust bean gum as 34
supporting material for encapsulation of probiotics and found that this coating material 35
was less sensitive to acid than alginate. Guoqiang et al. (1991) reported that a mixed gel 36
matrix of -carrageenan and locust bean gum showed significant stability for 3 months 37
in continuous lactic acid fermentation. However, the encapsulation of probiotics using 38
-carrageenan-locust bean gum as support material required potassium ions which can 39
damage cells of the probiotic bacteria (such as Streptococcus thermophilus, L. bulgaricus, 40
and Bifidobacterium longum) during fermentation (Audet et al. 1988). Furthermore, large 41
amount of potassium ions are not recommended in human diet. 42
43
44
Cellulose Acetate Phthalate
45
Cellulose acetate phthalate (CAP) is an enteric coating material used for controlling drug S46
release in the intestines and thus has a well-established safety record for pharmaceutical N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 92

92 Chapter 4

1 and dietary supplements applications. CAP is not soluble in water at pH values of less than
2 about 5.8.
3 The advantage of CAP is that it is insoluble at acidic pH (less than 5) but is soluble at pH
4 greater than 6. Nevertheless, encapsulation of bifidobacteria by CAP was found to be inef-
5 fective in preventing acid injury to bacteria in highly acidic yogurt (Modler and Villa-Garcia
6 1993). Fávaro-Trindade and Grosso (2002) encapsulated Bifidobacterium lactis and Lacto-
7 bacillus acidophilus using CAP as the coating material and concluded that CAP provided
8 good protection for both microorganisms in acid and bile solutions, conditions similar to
9 those of the intestine.
10
11
Chitosan
12
13 Chitosan is a cationic linear polysaccharide composed essentially of β(1-4)-linked glu-
14 cosamine units together with some proportion of N-acetylglucosamine units. Droplets of a
15 chitosan solution suspended in an oil phase can be hardened by cross-linking with glu-
16 taraldehyde (suspension cross-linking) via solvent evaporation or by the addition of poly-
17 valent anions such as sodium tripolyphosphate (TPP) or citrate (ionotropic gelation). The
18 stirring rate, temperature, level of the gelling agent, concentration of the surfactant poly-
19 mer, and the viscosities of the phases were reported to affect the size and morphology of the
20 particles (Peniche et al. 2003). However, inhibitory effects of chitosan on different types of
21 lactic acid bacteria were reported by Groboillot et al. (1993).
22
23
Others
24
25 Lian et al. (2002) investigated the survival of bifidobacteria after spray-drying with differ-
26 ent carrier matrices and indicated that the survival of microencapsulated bifidobacteria
27 after spray-drying varied with strains and was mainly dependent on the carriers used. In
28 addition, use of 10% gelation, gum arabic, and soluble starch resulted in the highest sur-
29 vival of bifidobacteria. O’Riordan et al. (2001) used modified waxy maize starch to encap-
30 sulate Bifidobacterium spp. with an average size of 5 µm by spray-drying and demonstrated
31 that maximum recovery yields were 30%. However, the starch-encapsulated Bifidobac-
32 terium spp. showed no improvement in viability compared with the control-free cells when
33 exposed to acidic conditions or when added to yogurt. They concluded that the modified
34 starches might not be suitable for use as an encapsulating material for probiotic strains.
35 Ananta et al. (2005) incorporated oligofructose-based or polydextrose-based skim milk in
36 a carrier matrix which resulted in a high level of survival for Lactobacillus rhamnosus
37 (LGG). A probiotic survival rate of 60% was achieved at an outlet temperature of 80°C.
38 Desmond et al. (2002) studied the survival of Lactobacillus paracasei in a mixture of
39 reconstituted skim milk and gum acacia followed by spray-drying and found ten-fold
40 greater survival than in the control group. Hou et al. (2003) developed a technique to pro-
41 tect lactic acid bacteria against simulated gastrointestinal conditions by encapsulating bac-
42 terial cells within artificial sesame oil emulsions.
43
44
Effect of Spray-Drying on Probiotic Survival
45
46S The survivability of the encapsulated probiotics is most significantly influenced by the exe-
47N cution of the spray-drying process as well as other factors. The survival of various lactic
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 93

Applications of Probiotic Encapsulation in Dairy Products 93

cultures affected by spray-drying have been carried out by various investigators (O’Riordan 1
et al. 2001; Lian et al. 2002; Lian et al. 2003; Picot and Lacroix 2003, 2004; Ananta et al. 2
2005). Different polysaccharides were used as the matrix and the nozzle temperature of the 3
spray dryer as well as the water activity of the microcapsules had a considerable impact on 4
the survival of probiotics. 5
The heat resistance of probiotic strains should be taken into account during the spray-dry 6
encapsulation of sensitive microorganisms. Picot and Lacroix (2004) dispersed fresh cells in 7
a heat-treated whey protein suspension followed by spray-drying and found a survival rate 8
of 26% for Bifidobacterium breve after spray-drying and 1.4% for the more heat-sensitive 9
B. longum. Lian et al. (2002) studied the survival of bifidobacteria after spray-drying and 10
found that Bifidobacterium longum B6 exhibited the least sensitivity to spray-drying and 11
showed the highest survival of 82.6% after drying with skim milk. 12
The outlet-air temperature is another major parameter affecting probiotic survival after 13
spray-drying with lower temperatures resulting in higher survival rates (Favaro-Trindade 14
and Grosso 2002; Ananta et al. 2005; Chen et al. 2006). Lian et al. (2002) reported that Bifi- 15
dobacterium spp. had the highest survival after drying at 50°C. Chen et al (2006) studied 16
the viability of probiotics after spray-drying at outlet air temperatures of 60, 70, and 80°C 17
and found that the survival of L. acidophilus and B. longum decreased as the outlet-air tem- 18
perature increased. However, the final total probiotic counts still remained above the rec- 19
ommended therapeutic minimum (107 cfu/g) after spray-drying at various outlet air 20
temperatures. Gardiner et al. (2002) spray-dried L. paracasei NFBE 338 Rifr with 20% 21
reconstituted skim milk at air inlet and outlet temperatures of 175°C and 68°C, respec- 22
tively, and found a probiotic survival rate of 84.5%. Ananta et al. (2005) assessed probiotic 23
injury sites in spray-drying by flow cytometry and found that the damage to cell mem- 24
branes was the key reason for cell death. Higher outlet temperature used for spry-drying 25
resulted in more serious disintegration of membranes. On the other hand, inactivation 26
caused by increased outlet-air temperatures varied with the carrier used. Lian et al. (2002) 27
indicated that using soluble starch as the carrier matrix significantly improved the probiotic 28
survival at a high outlet-air temperature, whereas skim milk showed the least effect. 29
30
31
Probiotic Survival in Dairy Products
32
An adequate number of viable cells, namely the “therapeutic minimum,” need to be con- 33
sumed regularly in order for consumers to experience the probiotic effects. Encapsulation 34
has been investigated for improving the viability of the microorganisms in dairy products 35
including fermented milk (Adhikari et al., 2000; Sultana et al. 2000; Sun and Griffiths 36
2000; Adhikari et al. 2003; Krasaekoopt et al. 2004; Picot and Lacroix 2004; Iyer and 37
Kailasapathy 2005), cheese (Dinakar and Mistry 1994; Desmond et al. 2002), and frozen 38
desserts (Sheu and Marshall 1993; Shah and Ravla. 2004). 39
40
41
Cheese
42
Introducing encapsulated probiotics in cheese not only enhances the storage viability 43
of probiotics but also improves the flavor of cheese. Research results (Dinakar and Mistry 44
1994; Desmond et al. 2002) have reported that cheese containing encapsulated Bifidobac- 45
terium spp. and L. paracasei did not differ from the control cheese in soluble protein, S46
flavor, appearance, texture, and normal microflora. The viabilities of both encapsulated N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 94

94 Chapter 4

1 Bifidobacterium spp. and L. paracasei in cheese were maintained for at least 6 months
2 and 3 months, respectively. In addition, acetic acid, a common metabolite of Bifidobac-
3 terium spp. and not preferred in dairy products, was not detected during ripening.
4
5
Frozen Dairy Desserts
6
7 It is difficult to incorporate probiotic bacteria into frozen desserts due to the acidity of the
8 products, high osmotic pressure, freeze injury, and exposure to air, as air is introduced dur-
9 ing freezing of these products (Shah and Ravla 2004). Thus, the application of microencap-
10 sulated probiotic bacteria to frozen dairy desserts may overcome these difficulties and
11 could produce useful markets and health benefits. Sheu et al. (1993) studied the survival of
12 culture bacteria in frozen desserts and indicated that the survival rate for encapsulated
13 L. bulgaricus in continuously frozen ice milk was approximated at 90% without a measura-
14 ble effect on the sensory characteristics.
15
16
Yogurt
17
18 Incorporation of probiotics has been shown to enhance the therapeutic value of yogurt.
19 However, the survival of probiotics in yogurt is low due to the prevailing low pH ranging
20 from 4.2 to 4.6 (Kailaspathy and Rybka 1997). Many studies have documented the positive
21 effects of encapsulation of probiotics and their survival in fermented dairy products
22 (Adhikari et al. 2000; Sultana et al. 2000; Sun and Griffiths 2000; Adhikari et al. 2003;
23 Krasaekoopt et al. 2004; Picot and Lacroix 2004; Iyer and Kailasapathy 2005). Of all
24 encapsulation techniques tested, chitosan-coated alginate beads were reported to offer no
25 enhanced protection for probiotics in yogurt stored at 4°C for 4 weeks (Krasekoopt et al.
26 2006).
27
28
Probiotic Survival in Gastrointestinal Conditions
29
30 Encapsulated probiotics should survive passage through the upper digestive tract in large
31 numbers in order to ensure desired beneficial effects in the host intestines (Gilliland 1989).
32 Various effects of encapsulation on the survival of bacteria under gastrointestinal condi-
33 tions have been reported (Table 4.5). The survival of encapsulated cells is strongly depen-
34 dent on the type and concentration of coating materials, bead size, initial cell numbers, and
35 bacterial species. Most studies have proven the advantages of encapsulating probiotics over
36 free cells under in vitro gastric conditions, others did not find any additional protection
37 under strongly acidic conditions (Rao et al. 1989; Sultana et al. 2000; O’Riordan et al.
38 2001; Truelstrup et al. 2002).
39 Several coating materials including sodium alginate (Lee and Heo 2000; Chandramouli
40 et al. 2004), sodium alginate with a polycation (Cui et al. 2000; Krasaekoopt et al. 2004;
41 Lee et al. 2004; Iyer and Kailasapathy 2005), gellan/xanthan gum (Sun and Griffiths 2000;
42 McMaster et al. 2005), artificial oil (Hou et al. 2003), gum arabic (Lian et al. 2003), and
43 whey protein (Picot and Lacroix 2004) showed good protection for encapsulating probi-
44 otics under gastrointestinal conditions. Lee and Heo (2000) studied the survival of
45 B. longum immobilized in alginate beads in simulated gastric juices and bile salt solutions
46S and found that the death rate of the probiotics in the capsules decreased proportionally with
47N an increase in the alginate concentration (13%), bead size (13 mm), and initial cell
Table 4.5. The effect of encapsulation on the survival of bacteria under gastrointestinal conditions

Survival under gastrointestinal


Probiotics Encapsulation method Coating materials conditions Reference
B. longum Extrusion 2–4% sodium alginate Depending on alginate Lee and Heo (2000)
concentration and bead size
B. bifidum Extrusion 2% sodium alginate with Higher than 106 cfu mL–1 Cui et al. (2000)
poly-L-lysine or chitosan
L. acidophilus Emulsion 2% alginate with Hi-maize starch Higher than 106 cfu mL–1 Sultana et al. (2000)
Bifidobacterium spp.
B. infantis Extrusion 0.75% gellan/1% xanthan gum Higher than 106 cfu mL–1 Sun and Griffiths (2000)
B. ruminantium Spray-drying 10% starch No counts detectable O’Riordan et al. (2001)
B. adolescentis Emulsion 3% alginate 8.2–1.0 log cfu mL–1 Truelstrup et al. (2002)
B. breve
B. lactis
B. longum
L. bulgaricus Emulsion Artificial oil Higher than 106 cfu mL–1 Hou et al. (2003)
B. longum Spray-drying 30% gelatin 87.15% Lian et al. (2003)
35% soluble starch 95.47%
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 95

35% gum Arabic 93.53%


15% skim milk 81.26%
B. infantis Spray-drying 30% gelatin 92.73% Lian et al. (2003)
35% soluble starch 92.70%
35% gum Arabic 89.17%
15% skim milk 65.16%
L. casei Emulsion 1% alginate with microporous Higher than 106 cfu mL–1 Song et al. (2003)
glass membrane
L. acidophilus Extrusion 1.8% sodium alginate 105–106 cfu mL–1 Chandramouli et al. (2004)
L. acidophilus Extrusion 2% sodium alginate with chitosan 1.5 × 106 cfu g–1 Krasaekoopt et al. (2004)
Alginate 1.3 × 104 cfu g–1
PLL-alginate 1.0 × 104 cfu g–1
L. casei Extrusion 2% sodium alginate with chitosan 1.6 × 106 cfu g–1 Krasaekoopt et al. (2004)
Alginate 6.7 × 103 cfu g–1
PLL-alginate 7.0 × 103 cfu g–1
L. bulgaricus Extrusion 2% sodium alginate with chitosan Higher than 106 cfu mL–1 Lee et al. (2004)
B. breve Emulsion/spray-drying 10% heat-denatured whey protein isolate 1.0log cfu mL–1 Picot and Lacroix (2004)
B. longum 3.8log cfu mL–1
L. acidophilus Extrusion Sodium alginate with poly-L-lysine Higher than 106 cfu mL–1 Iyer and Kailasapathy (2005)
or chitosan
Addition of Hi-maize starch

95
or Raftiline®/Raftilose®
B. lactis Extrusion 0.75% gellan/1% xanthan gum Higher than 106 cfu mL–1 McMaster et al. (2005)

N47
S46
45
44
43
42
41
40
39
38
37
36
35
34
33
32
31
30
29
28
27
26
25
24
23
22
21
20
19
18
17
16
15
14
13
12
11
10
9
8
7
6
5
4
3
2
1
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 96

96 Chapter 4

1 numbers. Similar results were also observed by Chandramouli et al. (2004). Further-
2 more, Sultana et al. (2000) reported that survival of probiotics in alginate-starch beads with
3 diameters of 1.0 mm did not improve after exposure to acidic and bile salt solutions.
4
5 Applications of Modern Optimization Techniques on the Optimal
6 Manufacturing Conditions for Probiotic Capsules
7
8 Factors that can influence the survival rate of the probiotic capsules have been discussed in
9 the above sections. Different ingredients constituting the probiotic capsules may also have
10 profound effects on the survival rate. In order to clarify the effects of these different ingre-
11 dients, experimental design can be carried out and response surface models developed.
12 Furthermore, modern optimization techniques can be applied to attain the optimal compo-
13 sition of the capsules.
14 The objective of this section is to demonstrate the application of two modern optimiza-
15 tion techniques for searching the optimal combination of coating materials for probiotic
16 microcapsules. The whole concept (Figure 4.1) includes:
17
18 1. Performing screening experiments and experimental design
19 2. Encapsulating the probiotics according to the experimental design
20 3. Building response surface models and formulating the optimization model
21 4. Performing optimization
22 5. Verifying the optimal manufacturing conditions.
23
24 A practical example of incorporating an additional prebiotic component to alginate matrix
25 is presented in the following to illustrate the entire scheme.
26
27 Performing Screening Experiments and Experimental Design
28
29 Theoretically, all factors that affect the physicochemical properties of a final product
30 should be included in the experimental design. However, if all the variables are included,
31 the search process may become cumbersome. Therefore, the potentially dominant parame-
32 ters must be identified by a screening process to limit the number of experiments needed to
33 a reasonable extent. After the screening experiments, the remaining screened factors are
34 used in the design.
35 The experimental design, which applies the statistical principles for data collection prior
36 to the experiment, has the main advantage of reducing the number of experimental trials
37 needed to evaluate multiple parameters and to determine their interactions (Porretta et al.
38 1995; Lee et al. 2000; Chen et al. 2005b). The response surface design, including the
39 Central Composite Design (CCD) and Box-Behnkin Design (BBD; Box and Behnkin
40 1960) provides more informative data from the least number of experimental runs
41 than from the traditional method. The CCD is a popular class of second-order design. This
42 design involves the use of a two-level factorial and 2k axial points with k being the number
43 of factors involved. On the other hand, the BBD is an effective three-level design based
44 on the construction of a balanced incomplete block design, and is an important alternative
45 to CCD.
46S In this study, survival of encapsulated probiotics (Lactobacillus spp. and Bifidobac-
47N terium spp.) was found to be dependent on the concentrations of alginates as well as the
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 97

Applications of Probiotic Encapsulation in Dairy Products 97

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
Figure 4.1. Research scheme for application of modern optimization techniques for 38
encapsulating probiotics. 39
40
41
42
43
three prebiotic coating materials (peptides, FOS, and IMO). These four components were 44
regarded as independent variables and therefore a four-variable BBD with six replicates at 45
the center point (total 30 trials) was selected to build the response surface models. The S46
coded and the nature variables and their respective levels are shown in Table 4.6. N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 98

98 Chapter 4

1 Table 4.6. Process variables and their levels in four variables—Box Behnkin Design
2
Level
3
4 Independent variable Symbol Coded Nature
5 –1 1.00
6 Sodium alginate concentration (%) X1 0 2.00
7 +1 3.00
–1 0.00
8
Peptides concentration (%) X2 0 0.50
9 +1 1.00
10 –1 0.00
11 FOS concentration (%) X3 0 1.50
12 +1 3.00
–1 0.00
13
IMO concentration (%) X4 0 1.50
14 +1 3.00
15
16
17
Encapsulating the Probiotics According to the Experimental Design
18
19 A schematic representation of the manufacturing process for probiotic microcapsules is
20 shown in Figure 4.2 and the process can be described as follows. Probiotic microcapsules
21 were prepared according to the BBD by mixing 4% (v/v) of culture concentrate (1% each
22 of L. acidophilus, L. casei, B. bifidum, and B. longum) with sodium alginate and the previ-
23 ously autoclaved (121°C, 15 min) prebiotics, FOS (03%), and IMO (03%), as well as
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S
47N Figure 4.2. Flow diagram for the preparation of probiotic microcapsules.
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 99

Applications of Probiotic Encapsulation in Dairy Products 99

peptides (01%). The mixture with cell suspension was injected through a 0.11 needle into 1
sterile 0.1 M CaCl2. The beads approximately 0.5 mm in diameter were allowed to stand for 2
1 hr for solidification, and then rinsed with, and subsequently kept in, sterile 0.1% peptone 3
solution at 4°C. Survival of the microencapsulated probiotics before and after simulated gas- 4
tric fluid test (defined as responses) was determined. The four responses were defined as viabil- 5
ity of Lactobacillus spp (L. acidophilus + L. casei.) before simulated gastric fluid test (SGFT), 6
viability of Bifidobacterium spp. (B. longum + B. bifidum) before SGFT, viability of Lacto- 7
bacillus spp. after SGFT, and viability of Bifidobacterium spp after SG 8
9
10
Building Response Surface Models and Formulating the 11
Optimization Model 12
13
Experimental data can be utilized to build mathematical models using linear, quadratic, or 14
cubic functions by the least square regression method, after which the fitted functions are 15
tested for adequacy and fitness using analysis of variance (ANOVA). Once an appropriate 16
approximating model has been derived, it can then be analyzed using various optimization 17
techniques to determine the optimum conditions for the process. 18
Model analysis and the Lack-of-Fit test can be used for the selection of adequate mod- 19
els, as outlined by Lee et al. (2000) and Weng et al. (2001). The model analysis compares 20
the validities of the linear, quadratic, and cubic models for the different responses accord- 21
ing to their F-values. A model with P-values (P>F) below 0.05 is regarded as significant 22
and the highest-order polynomial that is significant will be selected. The Lack-of-Fit test 23
demonstrates if the lack-of-fit between the experimental values and those calculated based 24
on the model equations can be explained by the experimental error. The model with no sig- 25
nificant lack-of-fit is appropriate for the description of the response surface. 26
In this example, the model analysis results (Table 4.7 and Table 4.8) show that the foll- 27
owing four equations, which represent three linear survival models (Lactobacillus spp. 28
before SGFT, Bifidobacterium spp. before SGFT and Bifidobacterium spp. after SGFT) 29
and one cubic model (Lactobacillus spp. after SGFT), appear to be the most accurate with 30
no significant lack-of-fit. 31
32
33
34
Table 4.7. Model analysis and lack-of-fit test for the viability of lactic acid bacteria for before
simulated gastric fluid test 35
36
La Bb 37
Model analysisc Lack-of-Fit testd Model analysis Lack-of-fit test 38
Source (P>F) (P>F) (P>F) (P>F) 39
Linear 0.0002** 0.3972 0.0013** 0.8444 40
Quadratic 0.5377 0.3595 0.4090 0.8743 41
Cubic 0.5023 0.2509 0.6494 0.9092 42
43
* Significant at 5% level.
** Significant at 1% level. 44
a L: L. acidophilus  L. casei.
b B: B. longum  B. bifidum.
45
c Model analysis selects the highest order polynomial where the additional terms are significant. S46
d Lack-of-Fit test wants the selected model to have insignificant lack-of-fit. N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 100

100 Chapter 4

1 Table 4.8. Model analysis and Lack-of-Fit test for the viability of lactic acid bacteria for after
2 simulated gastric fluid test
3 L B
4
Model analysis Lack-of-fit test Model analysis Lack-of-fit test
5 Source (P>F) (P>F) (P>F) (P>F)
6
7 Linear 0.0004** 0.0812** 0.0292* 0.4182
Quadratic 0.0161* 0.0631** 0.2185 0.4976
8 Cubic 0.0006** 0.1421 0.2918 0.6442
9
10 * Significant at 5% level.
** Significant at 1% level.
11
12
13 L
f bef  8 . 17  0 . 075 X 1  0 . 13 X 2  0 . 024 X 3 1 . 05 × 103 X 4 (1)
14
15
16 B
f bef  7 . 71 − 0 . 098 X 1  0 . 46 X 2  0 . 021 X 3  3 . 45  103 X 4 (2)
17
18
L  1.41  3.53X  8.89X  1.35X  0.68X  0.83X 2  1.19X 2
faft
19 1 2 3 4 1 2
20  0.23X 23  0.074X 24  5.89X1X2  0.029X1X3  0.65X1X4
21  1.46X2 X3  0.81X2X4  0.14X3X4  1.34X1X1X2  0.076X1X1X3
22  0.17X1X1X4  0.20X1X2X2  0.093X1X3X3  0.085X2X2X3
23
24  0.74X 2 X2 X4  0.48X2X3X3 (3)
25
26 B  7.35  0.045X  0.30X  0.065X  0.065X
f aft 1 2 3 4
(4)
27
28 L , f B , f L , and f B represent the functions for the survival of Lactobacillus spp.
29 where f bef bef aft aft

30 (superscript L) and Bifid obacterium spp. (superscript B) before (subscript bef) and after
31 (subscript aft) SGFT, respectively. The three-level BBD is incapable of forming the pure
32 cubic terms, that is, those with X3i, and equation (3) confirms this fact.
33 In order to search for a solution maximizing multiple responses, a composite fitness func-
34 tion (CFF) is defined as following:
35
36 1
⎛ m
⎞ m
37
38
CFF = ⎜

∏i =1
fi ⎟
⎠ (5)
39
40
41 where fi represents the ith function (response) and m denotes the total number of functions.
42 The term inside the parentheses in equation (5) is the product of all m functions. The com-
43 posite function combines m responses (m = 4 in our study) into one single function whose
44 maximum can then be sought by optimization techniques with each response contributing
45 equally to the CFF.
46S The relationship between the factors and the responses can be investigated by examining
47N the CFF contour plots created by holding constant two of the four independent variables.
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 101

Applications of Probiotic Encapsulation in Dairy Products 101

By fixing the peptides and FOS at three different levels, a three-dimensional plot of CFF 1
values as a function of sodium alginate and IMO can be produced. Figure 4.3 depicts that 2
the CFF values increase in accordance with the higher levels of FOS and peptides. On the 3
other hand, the higher IMO and alginate concentrations lead to lower CFF values when 4
FOS and peptides are 3 and 1%, respectively. Figure 4.3(c) shows clearly an optimal CFF 5
value of 8.172. 6
7
8
Performing Optimization 9
The CFF in equation (5) can be used as the objective function to be maximized in an 10
optimization problem, and the problem can be solved to find the optimal formulation for 11
probiotic microcapsules using optimization techniques. Optimization theory consists of a 12
body of numerical methods for finding and identifying the best candidate from a collection 13
of alternatives without having to explicitly evaluate all possible alternatives (Reklaintis et al. 14
1983). Among the optimization techniques, the steepest ascent (or descent) is commonly 15
used (see, for example, Stat-Ease, Inc., 2000), but the method is relatively inefficient and is a 16
local optimization technique capable of finding only local optima. Genetic Algorithms 17
(GAs), although even less efficient than the steepest ascent, are considered as global schemes. 18
The Sequential Quadratic Programming (SQP) technique is very powerful and efficient, and 19
with some modifications it can also perform global optimizations (Chen 2003). 20
21
22
Optimization Using the SQP Technique 23
A quadratic programming (QP) problem is an optimization problem involving a quadratic 24
objective function and linear constraints. The SQP method represents the current state-of- 25
the-art in non-linear programming methods (The Math Work Inc., 2000) and can be used to 26
solve a series of QP problems approximating the original non-linear programming prob- 27
lem. The basic scheme of an SQP technique can be expressed in the following steps 28
(Reklaintis et al. 1983; Chen 2003): 29
30
Step 1: Set up and solve a QP subproblem, giving a search direction. 31
Step 2: Test for convergence, stop if it is satisfied. 32
Step 3: Step forward to a new point along the search direction. 33
Step 4: Update the Hessian matrix in QP and go to step 1. 34
35
In order to search for the global optimum, the concept of multi-start global optimization 36
procedure (Snyman and Fatti 1987) may be combined with the SQP method. If F* denotes 37
the global maximum and r, the number of sample points falling within the region of conver- 38
gence of the current overall maximum F after n points have been sampled, then, under sta- 39
tistically non-informative prior distribution, the probability that F be equal to F* satisfies 40
the following relationship (Chen 2003): 41
42
43
Pr[FF*]  q(n, r)  1[(n1)!(2nr)!]/[(2n1)!(nr)!] (6)
44
45
A global optimization program equipped with a multi-start SQP technique was coded to S46
solve for the optimal solution in this example. The modified SQP with the multi-start ability, N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 102

(a)
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15 (b)
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30 (c)
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S
47N Figure 4.3. Response surface plots of survivability of probiotic microcapsules showing effects of
sodium 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 103

Applications of Probiotic Encapsulation in Dairy Products 103

which is capable of reaching the global optimum with great certainty, has been proven to be a 1
very efficient method (Chen et al. 2004). The program generates a series of uniformly 2
distributed random points for initial search, and then the SQP is applied to find the optimum 3
based on each subsequent initial point. If the probability of locating the global optimum 4
exceeds a preset value (99.99% in this example), the global optimum is considered found. 5
Otherwise, the next random, initial point is generated and the SQP re-executed. 6
A very high probability (>0.9999) in equation (6) was set to ensure the global optimum 7
would be attained. Figure 4.4 shows the evolution of the CFF values for a sequence of ran- 8
domly generated initial searching points and the optimal points found. The optimization 9
results clearly show that determination of the optima depends on the initial search points 10
11
12
13
(a) 14
15
Composite fitness function (CFF)

16
17
18
19
20
21
22
23
24
25
26
27
Number of function evaluations 28
29
30
(b) 31
32
Optimal composite function value

33
34
35
36
37
38
39
40
41
42
43
44
Initial searching point set 45
S46
Figure 4.4. (a) Evolution curve of CFF with 2% alginate, 0.5% peptides, 1.5% FOS and 1.5% IMO
as the initial searching point; (b) evolution curve of optimal CFF for randomly generated initial
N47
searching 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 104

104 Chapter 4

1 and there are three different local optimal CFF values identified from 20 randomly gener-
2 ated initial points. Of these local optima, the global optimal CFF is 8.172 with 99.99%
3 certainty. The global maximum corresponds to: 8.30log cfu for survival of Lactobacillus
4 spp. before SGFT, 8.01log cfu for survival of Bifidobacterium spp. before SGFT, 8.00log
5 cfu for survival of Lactobacillus spp. after SGFT, and 7.72log cfu for survival of Bifidobac-
6 terium spp. after SGFT. The highest optimal CFF value (8.172) was attained for 10 of 20
7 sets and the optimal point consists of independent variables at X1 1, X2 1, X33, and
8 X4 = 0. In other words, the optimal combination of the coating materials for the probiotic
9 microcapsules is 1% sodium alginate blended with 1% peptides, 3% FOS, and 0% IMO.
10
11
12 Optimization Using the Genetic Algorithms
13 Genetic Algorithms are search procedures that imitate the natural evolution process and
14 can be used for the computation of the global maximum or minimum of a function
15 (Mitchell 1996). Genetic algorithms differ from other search techniques in that they search
16 among a population of points and use probabilistic rather than deterministic transition
17 rules. As a result, genetic algorithms search more globally (Wang 1997).
18 GAs provide a very flexible framework and recently have been regarded as not only a
19 global optimization method but also a multi-objective optimization method in various
20 areas. Generally, the algorithms can be described in the following steps (Goldberg 1989;
21 Mitchell 1996):
22
23 Step 1: Start with a randomly generated population of chromosomes, each of which
24 defines a combination of the coating materials in this example.
25 Step 2: Calculate the fitness f (x) of each chromosome x in the population, with the fit-
26 ness being the CFF value of that combination of the coating materials.
27 Step 3: Repeat the following substeps until n offsprings have been created: (i) select a
28 pair of parent chromosomes from the current population, the probability of selection
29 being an increasing function of fitness; (ii) with crossover rate, cross over the pair at a
30 randomly chosen point to form two offsprings; (iii) mutate the two offsprings at a pre-
31 scribed mutation rate and place the resulting chromosomes in the new population;
32 (iv) replace the current population with the new population. Each iteration of this
33 process is called a generation. The above procedure is called the simple GA (SGA).
34
35 The Micro Genetic Algorithm (MGA) is a popular modification to SGA to optimize the
36 processing conditions (Chen et al. 2003). The essence of MGA is the lack of mutations and
37 the presence of re-starts. Due to these features, the algorithm converges rapidly to a local or
38 global maximum (Nikitas et al. 2001). The lack of mutations also results in a rapid decrease
39 of the variance of the cost values of the population. When the variance value falls below a
40 certain limit, a restarting process begins in which the chromosome with the highest CFF
41 value is retained and the rest N–1 chromosomes (N is the total number of chromosomes in
42 one generation) are replaced by randomly generated new ones.
43 The efficiency of the algorithms can be examined by the number of function evaluations
44 as follows:
45
46S
47N Number of function evaluations  Number of generations  Population size (7)
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 105

Applications of Probiotic Encapsulation in Dairy Products 105

A smaller number of function evaluations indicate a higher efficiency. 1


In the study of alginate microcapsules incorporated with prebiotics, the CFF was opti- 2
mized using MGA. The initial population consisting of 10 chromosomes (population size) 3
was generated at random and the crossover rate was set to 0.5. The chromosomes with 4
higher CFF values were selected and retained for the next generation. The maximum num- 5
ber of generations was set to 500 for the problem. Figure 4.5 shows the evolution curve of 6
the first 3000 function evaluations in searching for the global, optimal value. The MGA 7
produced rapidly increasing CFF during the early stage of the optimization process consist- 8
ing of a total of 5000 function evaluations, which is typical for MGA. 9
The chromosomes having the maximum CFF provided the optimal ratio of concentra- 10
tions of the coating materials. The optimal value (CFF = 8.172) was obtained after 1490 11
function evaluations during the process. 12
13
14
Verifying the Optimal Manufacturing Conditions 15
After the optimal processing condition is found by the SQP or MGA, repeated experiments 16
based on the condition should be conducted to verify the predicted optimum. The verifica- 17
tion results can then be analyzed using ANOVA from the SAS software package (SAS 18
Institute Inc., 1990), with Duncan’s multiple range test for significance to detect differences 19
between predicted values and observed values. In this example, the optimal production 20
condition for the coating composition, derived from the SQP and MGA, was the same. The 21
optimal combination of the coating materials for the probiotic microcapsules is 1% sodium 22
alginate blended with 1% peptides, 3% FOS, and 0% IMO. The four responses (survival of 23
Lactobacillus spp. and Bifidobacterium spp. before and after SGFT) and the CFF value 24
derived from the verification experiments are all very close to the SQP- or MGA-based pre- 25
diction, with no apparent significant differences (P 0.05) comparing the two sets. Both 26
SQP and MGA techniques may be used to determine the optimal combination of the coat- 27
ing materials for probiotic microcapsules. By comparing both methods, SQP was deemed 28
to be much more efficient than MGA at such a task. 29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
S46
Figure 4.5. Optimum composite function values using MGA. N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 106

106 Chapter 4

1 Practical Applications of Encapsulated Probiotics in Dairy Products


2
3 As discussed above, encapsulated probiotics have been used for accelerating cheese ripen-
4 ing, fortifying dairy products with beneficial bacteria as well as enhancing the shelf life and
5 bioavailability of this class of microorganisms in dietary supplements.
6 Several companies are currently involved in designing and manufacturing such products
7 to meet customers’ needs. Following are few examples of incorporating encapsulated pro-
8 biotics into real food systems:
9
10 Yogurt
11
12 For manufacturing set yogurt, homogenized whole milk and skim milk powder are blended
13 for a total solids content of 15–18%(w/w). The mix is pasteurized at 80–85°C for 30 min.
14 and cooled to 42°C before inoculation with a commercial freeze-dried starter culture con-
15 taining S. thermophilus and L. bulgaricus. The encapsulated probiotic cultures are then
16 added and the resulting mix is dispensed into containers and incubated at 42°C for 4–5 hr
17 until the pH reached 4.5. Finally, yogurts are stored at 4°C (Adhikari et al. 2000; Sultana
18 et al. 2000; Sun and Griffiths 2000; Krasaekoopt et al. 2004; Picot and Lacroix 2004; Iyer
19 and Kailasapathy 2005).
20 Stirred yogurt is manufactured in the same way except that the mix after inoculation is
21 incubated at 42°C for 4–5 hr until the pH reaches 4.5, added with 10% microencapsulated
22 probiotics, and then stirred and dispensed into containers. The probiotic yogurt is stored at
23 4°C (Adhikari et al. 2003). The probiotic counts of yogurts remained above 106 cfu/mL
24 and the final pH was 3.9–4.1 after one month of storage.
25 Commercialized yogurt products containing microencapsulated probiotics are also
26 available. Kaung-Chuan Inc. in Taiwan produces a bio-yogurt drink with probiotic micro-
27 capsules, which incorporate Bifidobacterium spp, are made by gelatin and have an average
28 size of 1–2 mm. The company claims that this product has intestinal benefits.
29
30 Cheese
31
32 Introducing encapsulated probiotics to cheese not only enhances the storage viability of
33 probiotics but also improves the cheese flavor.
34 For manufacturing cheddar cheese, raw whole milk is pasteurized and cooled to 31°C. The
35 freeze-dried mesophilis lactic starter culture is added at the rate of 5 g/100 g of milk. Curd
36 forms in approximately 30 min. and is cut with 0.65 cm wire knives. After a 15 min. healing
37 period, the temperature of the curd and whey mixture is raised to 37–38°C in 30 min. and then
38 maintained at that temperature for an additional 30 min. After the whey is drained, the curd is
39 cheddared to pH 5.2, and then milled, salted, followed by addition of the microencapsulated
40 probiotics and packing into hoops that are further ripened at 7°C for 6 months. Cheese con-
41 taining encapsulated Bifidobacterium was shown to possess similar flavor, texture, and
42 appearance compared to the control (Dinakar and Mistry 1994; Desmond et al. 2002).
43
44 Frozen Desserts
45
46S For manufacturing frozen ice milk, probiotics microencapsulated with 3% calcium alginate
47N (bead diameters > 30 µm) are blended with milk (5% fat) and the mix is frozen continuously
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 107

Applications of Probiotic Encapsulation in Dairy Products 107

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
Figure 4.6. Typical manufacturing process of fermented, frozen dairy desserts with microencap-
sulated probiotics. 36
37
38
39
40
41
in a freezer. Addition of microencapsulated probiotics has no measurable effect on the over- 42
run and the sensory characteristics of the products with 90% probiotic survival (Sheu et al. 43
1993). Sheu et al. (1993) manufactured fermented frozen dairy desserts by blending freeze 44
dried microencapsulated probiotics with yogurt and base mix, and then the mix was frozen 45
in a continuous freezer. Figure 4.6 details the process of incorporating encapsulated probi- S46
otic culture to a frozen milk-based dessert system. N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 108

108 Chapter 4

1 Summary
2
3 Encapsulated probiotics can be used in many dairy products such as yogurt, frozen
4 desserts, and cheese. In the encapsulated form, these sensitive microorganisms are pro-
5 tected from harsh environments including high levels of lactic and acetic acid, gastrointesti-
6 nal conditions, and freezing temperatures. Among encapsulation methods, spray-drying,
7 extrusion, and emulsion are the most common techniques for probiotic encapsulation.
8 However, the high cost of the process and the technical difficulty limit the large-scale appli-
9 cation of encapsulation technologies in the dairy industry.
10 Carrier matrices, encapsulation methods, and various dairy products to which the probi-
11 otic capsules are applied can influence the survival rate of the probiotics. Different ingredi-
12 ents constituting the probiotic capsules may also have profound effects on the survival rate.
13 In order to clarify the effects of these different ingredients, experimental design can be car-
14 ried out and response surface models developed. Furthermore, modern optimization tech-
15 niques can be applied to attain the optimal composition of the capsules. The two-stage
16 effort of obtaining a surface model using RSM and optimizing this model using SQP and
17 MGA techniques has been demonstrated to represent an effective approach.
18
19
References
20
21 Adhikari, K., A. Mustapha and I. U. Grün. 2000. Viability of microencapsulated Bifidobacteria in set yogurt dur-
22 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-
23 terium in stirred yogurt. Journal of Food Science 68(1): 275–280.
24 Ananta, E., M. Volkert and K. Knorr. 2005. Cellular injuries and storage stability of spray-dried Lactobacillus
25 rhamnosus GG. International Dairy Journal 15: 399–409.
26 Audet, P., C. Paquin and C. Lacroix. 1988. Immobilized growing lactic acid bacteria with k-carrageenan-locust
27 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
28 Streptococcus salivarius ssp. Thermophilus, Lactobacillus delbrueckii ssp. bulgaricus, and Lactococcus lactis
29 ssp. lactis in a whey permeate medium. Applied and Environmental Microbiology 55(1): 185–189.
30 Audet, P., C. Lacroix and C. Paquin. 1992. Effect of agitation rate on cell release rate and metabolism during con-
31 tinuous fermentation with entrapped growing. Biotechnology Techniques 6(3): 265–270.
32 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.
33 Bielecka, M., E. Biedrzycka and A. Majkowska. 2002. Selection of probiotics and prebiotics for symbiotic and
34 confirmation of their in vivo effectiveness. Food Research International 35: 125–131.
35 Box, G. E. P. and E. W. Behnkin. 1960. Some new three level designs for the study of quantitative variables. Tech-
36 nometrics 2: 455–475.
37 Canh, L., M. Millette, M. Mateescu and M. Lacroix. 2004. Modified alginate and chitosan for lactic acid bacteria
immobilization. Biotechnology and Applied Biochemistry 39: 347–354.
38 Champagne, C. P., Gaudy, D. Poncelet and R. J. Neufeld. 1992. Lactococcus lactis release from calcium alginate
39 beads. Applied and Environmental Microbiology 58: 1429–1434.
40 Champagne, C. P., Lacroix and I. Sodini-Gallot. 1994. Immobilized cell technologies for the dairy industry. Criti-
41 cal Reviews in Biotechnology 14(2): 109–134.
42 Champagne, C. P. and N. J. Gardner. 2005. Challenges in the addition of probiotic cultures to foods. Critical
Reviews in Food Science and Nutrition 45: 61–84.
43 Chandramouli, V., K. Kailasapathy, P. Peiris and M. Jones. 2004. An improved method of microencapsulation and
44 its evaluation to protect Lactobacillus spp. in simulated gastric conditions. Journal of Microbiological Methods
45 56: 27–35.
46S Chen, H. C., C. W. Lin and M. J. Chen. 2006. The effects of freeze drying and rehydration on survival of microor-
47N ganisms in Kefir. Asian-Australian Journal Animal Science 19(1): 126–130.
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 109

Applications of Probiotic Encapsulation in Dairy Products 109

Chen, M. J., K. N. Chen and C. W. Lin. 2003. Optimization of the viability of probiotics in a new fermented milk 1
drink by genetic algorithms for response surface modeling. Journal of Food Science 68: 632–638. 2
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.
3
Chen, M. J., K. N. Chen and C. W. Lin. 2005a. Optimization of incorporated prebiotics as coating materials for 4
probiotic microencapsulation. Journal of Food Science 70 (5): 260–267. 5
Chen, M. J., K. N. Chen and C. W. Lin. 2005b. Optimization on response surface models for the optimal manufac- 6
turing conditions of dairy tofu. Journal of Food Engineering 68(4): 471–480. 7
Chen, S. 2003. Robust design with dynamic characteristics using stochastic sequential quadratic programming.
Engineering Optimization 35(1): 79–89.
8
Cui, J., J. Goh, P. Kim, S. Choi and B. Lee. 2000. Survival and stability of Bifidobacteria loaded in alginate poly-L- 9
lysine microparticles. International Journal of Pharmaceutics 210: 51–59. 10
Dave, R. I. and N. P. Shah. 1997. Effectiveness of ascorbic acid as an oxygen scavenger in improving viability 11
of probiotic bacteria in yoghurts made with commercial starter cultures. International Dairy Journal 12
7: 435–443.
Dave, R. I. and N. P. Shah. 1998. Ingredients supplementation effects on viability of probiotic bacteria in yogurt.
13
Journal of Dairy Science 81: 2804–2816. 14
Desmond, C., R. P. Ross, E. O’Callaghan, G. Fitzgerald and C. Stanton. 2002. Improved survival of Lactobacillus 15
paracasei NFBC 338 in spray-dried powders containing gum acacia. Journal of Applied Microbiology 16
93: 1003–1011. 17
Dinakar, P. and V. V. Mistry. 1994. Growth and viability of Bifidobacterium bifidum in cheddar cheese. Journal of
Dairy Science 77: 2854–2864.
18
Doleyres, Y. and C. Lacroix. 2005. Technologies with free and immobilized cells for probiotic Bifidobacteria pro- 19
duction and protection. International Dairy Journal 15: 973–988. 20
Favaro-Trindade, C. S. and C. R. F. Grosso. 2002. Microencapsulation of L. acidophilus (La-05) and B. lactis 21
(Bb-12) and evaluation of their survival at the pH values of the stomach and in bile. Journal of Microencapsula- 22
tion 19(4): 485–494.
Fuller, R. 1992. Probiotics: The Scientific Basis. London: Chapman & Hall. p. 125.
23
Gardiner, G. E., P. Bouchier, E. O’Sullivan, J. Kelly, J. K. Collins, B.G. Fitzgerald, R. P. Ross and C. Stanton. 2002. 24
A spray-dried culture for probiotic cheddar cheese manufacture. International Dairy Journal 12: 749–756. 25
Gaumann, A., M. Laudes, B. Jacob, R. Pommersheilm, C. Laue, W. Vogt and J. Schrezenmeir. 2001. Xenotrans- 26
plantation of parathyroids in rats using barium alginate and polyacrylic acid multilayer microcapsules. Experi- 27
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-
28
national Journal of Food Sciences and Nutrition 50: 213–224. 29
Gibson, G. R. and M. B. Roberfroid. 1995. Dietary modulation of the human colonic microbial: Introducing the 30
concept of prebiotics. Journal of Nutrition 125: 1401–1412. 31
Gilliland, S. E. 1989. Acidophilus milk products: A review of potential benefits to consumers. Journal of Dairy 32
Science 72: 2483–2494.
Goldberg, D. E. 1989. Genetic Algorithms in Search, Optimization, and Machine Learning. Massachusetts: Addi-
33
son-Wesley Publishing Company. 34
Gombotz, W. R. and S. F. Wee. 1998. Protein release from alginate matrices. Advanced Drug Delivery Reviews 35
31: 276–283. 36
Groboillot, A. F., C. P. Champagne, G. F. Darling and D. Poncelet. 1993. Membrane formation by interfacial cross- 37
linking of chitosan for microencapsulation of Lactococcus lactis. Biotechnology and Bioengineering 42(10):
1157–1163.
38
Guoqiang, D., R. Kaul and B. Mattiasson. 1991. Evaluation of. alginate-immobilized Lactobacillus casei for lac- 39
tate production. Applied Microbiology and Biotechnology 36: 309–314. 40
Havenaar, R. and J. H. H. Huis in’t Veld. 1992. Probiotics: A General View. New York: Chapman and Hall. 41
pp. 209–224. 42
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
43
Lactobacillus delbrueckii ssp. bulgaricus in artificial sesame oil emulsions. Journal of Dairy Science 86: 44
424–428. 45
Iwana, H., H. Masuda, T. Fujisawa, H. Suzuki and T. Mitsuoka. 1993. Isolation and identification of Bifidobac- S46
terium ssp. in commercial yogurt sold. Europe. Mitsuokifidobacteria Microflora 12: 39–45. N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 110

110 Chapter 4

1 Iyer, C. and K. Kailasapathy. 2005. Effect of co-encapsulation of probiotics with prebiotics on increasing the via-
2 bility of encapsulated bacteria under in vitro acidic and bile salt conditions and in yogurt. Journal of Food Sci-
ence 70(1): 18–23.
3 Jankowski, T., M. Zielinska and A. Wysakowska. 1997. Encapsulation of lactic acid bacteria with alginate/starch
4 capsules. Biotechnology Techniques 11(1): 31–34.
5 Kailaspathy, K. and S. Rybka. 1997. Lactobacillus acidophilus and Bifidobacterium spp.—their therapeutic
6 potential and survival in yogurt. Australia Journal of Dairy Technology 52: 28–35.
7 Kearney, L., M. Upton and A. Loughlin. 1990. Enhancing the viability of Lactobacillus plantarum inoculum by
immobilizing the cells in calcium-alginate beads. Applied and Environmental Microbiology 56(10): 3112–3116.
8 Kebary, K. M., S. A. Hussein and R. M. Badawi. 1998. Improving viability of Bifidobacteria and their effect on
9 frozen ice milk. Egyptian Journal of Dairy Science 26 (2): 319–337.
10 King, A. H. 1995. Encapsulation of food ingredients: A review of available technology, focusing on hydrocolloids.
11 In Encapsulation and Control Release of Food Ingredients, eds. S. J. Risch and G. A. Reineccius, pp. 213–220.
12 Washington DC: American Chemical Society.
Klaver, F. A. M., F. Kingma and A. H. Weerkamp. 1993. Growth and survival of bifidobacteria in milk. Netherlands
13 Milk and Dairy Journal 47: 151–164.
14 Koch, S., C. Schwinger, J. Kressler, C. H. Heinzen and N. G. Rainov. 2003. Alginate encapsulation of genetically
15 engineered mammalian cells: comparison of production devices, methods and microcapsule characteristics.
16 Journal of Microencapsulation 20(3): 303–316.
17 Krasaekoopt, W., B. Bhandari and H. Deeth. 2003. Evaluation of encapsulation techniques for probiotics for
yogurt. International Dairy Journal 13: 3–13.
18 Krasaekoopt, W., B. Bhandari and H. Deeth. 2004. Comparison of texture of yogurt from conventionally treated
19 milk and UHT milk fortified with low-heat skim powder. Journal of Food Science 69(6): E276–E280.
20 Krasaekoopt, W., B. Bhandari and H. C. Deeth. 2006. Survival of probiotics encapsulated in chitosan-coated algi-
21 nate beads in yoghurt from UHT- and conventionally treated milk during storage LWT 39: 177–183.
22 Kurman, J. A. and J. L. Rasic. 1991. The health potential of products containing Bifidobacteria. In Therapeutic
Properties of Fermented Milk, ed. R. K. Robinson, 115–117. London: Elsevier Application Food Science
23 Series.
24 Lacroix, C., C. Paquin and J. P. Arnaud. 1990. Batch fermentation with entrapped growing cells of Lactobacillus
25 casei. I. Optimization of the rheological properties of the entrapment. Applied Microbiology and Biotechnology
26 32: 403–408.
27 Lankaputhra, W. E. V. and N. P. Shah. 1995. Survival of Lactobacillus acidopilus and Bifidobacterium spp. in the
presence of acid and bile salts. Cultured Dairy Products Journal 30: 2–7.
28 Lankaputhra, W. E. V. and N. P. Shah. 1996. Survival of Bifidobacteria during refrigerated storage in the presence
29 of acid and hydrogen peroxide. Milchwissenschaft 51: 65–70.
30 Larisch, B. C., D. Poncelet and C. P. Champagne. 1994. Microencapsulation of Lactococcus lactis subsp. cre-
31 moris. Journal of Microencapsulation 11(2): 189–195.
32 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.
33 Lee, K. and T. Heo. 2000. Survival of Bifidobacterium longum immobilized in calcium alginate beads in simulated
34 gastric juices and bile salt solution. Applied and Environmental Microbiology 66(2): 868–873.
35 Lee, J., L. Ye, W. O. Landen and R. R. Eitenmiller. 2000. Optimization of an extraction procedure for the quantifi-
36 cation of vitamin E in tomato and broccoli using response surface methodology. Journal of Food Composition
37 and Analysis 13: 45–57.
Le-Tien, C., M. Millette, M. Mateescu and M. Lacroix. 2004. Modified alginate and chitosan for lactic acid bacte-
38 ria immobilization. Biotechnology and Applied Biochemistry 39: 349–354.
39 Lian, W. D., H. C. Hsiao and C. C. Chou. 2002. Survival of Bifidobacteria after spray-drying. International Journal
40 of Food Microbiology 74: 79–86.
41 Lian, W. D., H. C. Hsiao and C. C. Chou. 2003. Viability of microencapsulated Bifidobacteria in simulated gastric
42 juice 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 cultures
43 of Bifidobacterium longum in k-carrageenan-locust bean gum gel. Biotechnology Techniques 11 (7): 527–531.
44 Martin, J. H. and K. M. Chou. 1992. Selection of Bifidobacterium ssp. for use as dietary adjuncts in cultured dairy
45 foods I. Tolerance to pH of yogurt. Cultured Dairy Products Journal 27: 21–26.
46S Mattila-Sandholm, T., P. Myllärinen, R. Crittenden, G. Mogensen, R. Fonden and M. Saarela. 2002. Technological
47N challenges for future probiotic foods. International Dairy Journal 12: 173–182.
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 111

Applications of Probiotic Encapsulation in Dairy Products 111

McMaster, L. D., S. A. Kokott, S. J. Reid and V. R. Abratt. 2005. Use of traditional African fermented beverages as 1
delivery vehicles for Bifidobacterium lactis DSM 10140. International Journal of Food Microbiology 102: 2
231–237.
Mitchell, M. 1996. An Introduction to Genetic Algorithms. London: The MIT Press.
3
Modler, H. W. and L. Villa-Garcia. 1993. The growth of Bifidobacterium longum in whey-based medium and via- 4
bility of this organism in frozen yogurt with low and high levels of developed acidity. Culture Dairy Products 5
Journal 28: 4–8. 6
Nikitas, P., A. Pappa-Louisi, A. Papageorgiou and A. Zitrou. 2001. On the use of genetic algorithms for response 7
surface modeling in high-performance liquid chromatography and their combination with the Microsoft Solver.
J. Chromatography A 942: 93–105.
8
Norton, S. and C. Lacroix. 1990. Gellan gum gel as entrapment matrix for high temperature fermentation 9
processes: A rheological study. Biotechnology Techniques 4: 351–356. 10
O’Riordan, K. I., D. Andrews, K. Buckle and P. Conway. 2001. Evaluation of microencapsulation of a Bifidobac- 11
terium strain with starch as an approach to prolonging viability during storage. Journal of Applied Microbiology 12
91: 1059–1066.
Peirone, M., C. J. Ross, G. Hortelano, J. L. Brash and P. L. Chang. 1998. Encapsulation of various recombinant mam-
13
malian cell types in different alginate microcapsules. Journal of Biomedical Materials Research 42: 587–596. 14
Peniche, C., W. Argüelles-Monal, H. Peniche and N. Acosta. 2003. Chitosan: An attractive biocompatible polymer 15
for microencapsulation. Macromolecular Bioscience 3: 511–520. 16
Picot, A. and C. Lacroix. 2003. Production of multiphase water-insoluble microcapsules for cell microencapsula- 17
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 survival
18
in simulated gastrointestinal conditions and in yogurt. International Dairy Journal 14: 505–515. 19
Porretta, A., A. C. Birzi and E. Vicini. 1995. Effects of ultra-high hydrostatic pressure treatments on the quality of 20
tomato juice. Food Chemistry 52: 35–41. 21
Prevost, H. and C. Divies. 1987. Fresh fermented cheese production with continuous pre-fermented milk by a 22
mixed culture of mesophilis lactic streptococci entrapped in Ca-alginate. Biotechnology Letters 9(11):
789–794.
23
Prevost, H. and C. Divies. 1988. Continuous pre-fermentation of milk by entrapped yogurt bacteria. I. Develop- 24
ment of the process. Milchwissenschaft 43: 621–625. 25
Prevost, H. and C. Divies. 1992. Cream fermentation by a mixed culture of lactococci entrapped in two-layer cal- 26
cium alginate gel beads. Biotechnology Letters 14(7): 583–588. 27
Prüße, U., J. Dalluhn, J. Breford and K. Vorlop. 2000. Production of spherical beads by Jet Cutting. Chemical
Engineering Technology 23(12): 1105–1110.
28
Rao, A. V., N. Shiwnarain and I. Maharaj. 1989. Survival of microencapsulated Bifidobacterium pseudolongum in 29
simulated gastric and intestinal juices. Canadian Institute of Food Science and Technology Journal 22(4): 30
345–349. 31
Reklaintis, G. V., A. Ravindran and K. M. Ragsdell. 1983. Engineering Optimization: Methods and Applications. 32
New York: John Wiley and Sons.
Robinson, R. K. 1987. Survival of Lactobacillus acidophilus in fermented products. Suid Afrikaans Tydskrif Vir
33
Suiwelunde 19(1): 25–27. 34
Samona, A. and R. K. Robinson. 1994. Effect of yogurt cultures on the survival of Bifidobacteria in fermented 35
milks. Journal of the Society of Dairy Technology 47: 58–60. 36
Schillinger, U. 1999. Isolation and identification of lactobacilli from novel-type probiotic and mild yogurts and 37
their 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 Bifidobacterium
38
ssp. in yogurt. International Dairy Journal 7: 349–356. 39
Shah, N. P. and R. R. Ravla. 2004. Selling the cells in desserts. Dairy Industries International 69(1): 31–32. 40
Sheu, T. Y. and R. T. Marshall. 1993. Microentrapment of lactobacilli in calcium alginate gels. Journal of Food 41
Science 54: 557–561. 42
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.
43
Smidsrod, O. and G. Skjak-Braek. 1990. Alginate as immobilization matrix for cells. Trends in Biotechnology 44
8(3): 71–78. 45
Snyman, J. A. and L. P. Fatti. 1987. A multi-start global minimization algorithm with dynamic search trajectories. S46
Journal of Optimization Theory and Applications 54(1): 121–141. N47
Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 112

112 Chapter 4

1 Song, S., Y. Cho and J. Park. 2003. Microencapsulation of Lactobacillus casei YIT 9018 using a microporous
2 glass 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 bacteria
3 with alginate-starch and evaluation of survival in simulated gastrointestinal conditions and in yogurt. Inter-
4 national Journal of Food Microbiology 62: 47–55.
5 Sun, W. and M. W. Griffiths. 2000. Survival of bifidobacteria in yogurt and simulated gastric juice following
6 immobilization in gellan-xanthan beads. International Journal of Food Microbiology 61: 17–25.
7 Thies, C. 1996. A survey of microencapsulation. In Microencapsulation: Methods and Industrial Applications, ed.
S. Benita, 1–19. New York: Marcel & Dekker.
8 Thu, B., P. Bruheim, T. Espevik, O. Smidsrod, P. Soon-Shiong and G. Skjak-Braek. 1996. Alginate polycation
9 microcapsules. I. Interaction between alginate and polycation. Biomaterials 17: 1031–1041.
10 Truelstrup-Hansen, L., P. M. Allan-Wojtas, Y. L. Jin and A. T. Paulson. 2002. Survival of Ca-alginate microencap-
11 sulated Bifidobacterium spp. in milk and simulated gastrointestinal conditions. Food Microbiology 19: 35–45.
12 Wang, Q. J. 1997. Using genetic algorithms to optimize model parameters. Environmental Modelling& Software
12: 27–34.
13 Weng, W., W. Liu and W. Lin. 2001 Studies on the optimum models of the dairy product. Kou Woan Lao using
14 response surface methodology. Asian-Australasian Journal of Animal Sciences 14(10): 1470–1476.
15 Yoo, I., G. H. Seong, H. N. Chang and J. K. Park. 1996. Encapsulation of Lactobacillus casei cells in liquid-core
16 alginate capsules for lactic acid production. Enzyme and Microbial Technology 19: 428–433.
17 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):
18 342–348.
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S
47N
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 113

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

5 Encapsulation and Controlled Release in 1


2
Bakery Applications 3
4
5
Jamileh M. Lakkis 6
7
8
9
Introduction 10
11
In commercial baking operations, high volumes of dough and batter premixes are prepared
12
for further distribution to stores and on-site baking. Maintaining good functionality and
13
overall quality of these products requires careful inactivation of the prevailing leavening
14
systems during storage and their controlled reactivation upon preparation and baking.
15
The basic ingredients in doughs and cake batters include flour, fat, eggs, and sweeteners.
16
These components play an important role in determining the functional and eating quality
17
of bakery products. Minor ingredients such as yeasts and chemical leavening agents, how-
18
ever, can have more dramatic effects on the overall quality and shelf life of these products.
19
Recent advances in microencapsulation and controlled release technologies have con-
20
tributed significantly to current availability and wide consumers’ acceptability of shelf-
21
stable bakery products. Bakery manufacturers have been keen on adopting these technologies
22
due to the tremendous cost savings provided by extending shelf life, eliminating fermenta-
23
tion stage, and shortening dough proofing time along with minimal impact on processabil-
24
ity of bakery products. These benefits can be better appreciated considering the huge
25
market of bakery products that was estimated at $300 billion worldwide in 2005 (Sosland
26
Publishing Co., Kansas City, MO).
27
This chapter discusses methods for encapsulating and controlling the release of chem-
28
ical and biological leaveners as well as other functional components of bakery systems
29
such as sweeteners, antimicrobial agents, dough conditioners, and flavors. Microencapsula-
30
tion technologies as well as coating materials available for bakery applications are also
31
discussed.
32
33
Encapsulation Technologies for Bakery Applications 34
35
A variety of encapsulation technologies have been adapted for bakery applications, mainly
36
hot melt particle coating and congealing via spray chilling. Embedding via extrusion and
37
liposome/vesicles, used to a much lesser extent in bakery applications, has been covered
38
elsewhere in this book; therefore, only particle coating and congealing are discussed here.
39
40
Hot Melt Particle–Coating Technology 41
42
Fluid bed coating is a well-established technology for encapsulating and controlling the
43
release of solid actives. The process consists, essentially, of spraying a solution or a molten
44
fluid onto particles of a substrate material undergoing encapsulation. Application of a film
45
to a solid is a very complex process and requires careful selection of substrates and coating
S46
materials as well as process conditions.
N47

113
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 114

114 Chapter 5

1 The solid substrate is placed in a container that is typically an inverted truncated cone
2 with a fine retention screen and an air distribution plate at its base. As the warm air flows
3 through the distribution plate, the particles become fluidized and are accelerated in an
4 upward flow where they encounter fine spray of the coating fluid. The coating spray nozzle
5 can be fitted (1) to the top (top-spray system); (2) to the bottom (bottom-spray system
6 referred to as Wurster); or (3) tangential to the base container (Figure 5.1a, b, c). The choice
7 of a suitable coating configuration should take into consideration the type of solid to be
8 coated (powder, pellets, etc.) as well as the desired film thickness and release properties.
9 Top-spray fluid beds are favored for high-throughput applications as well as for film uni-
10 formity. Bottom-spray (Wurster) systems are preferred for their high coating effectiveness
11 as well as their ability to form perfectly sealed films. This is critical for controlled release
12 applications. Tangential-spray systems (rotor pellet coating), on the other hand, are suitable
13 for coating pellets and rods (yeasts) but not small particles (sodium bicarbonate and other
14 chemical leaveners). In the tangential coating system, rotation of the base plate disc sets the
15 pellets into a spiral motion where they encounter the coating spray, thus coating concur-
16 rently to the powder bed. Very thick film layers can be applied using the rotor configuration.
17 In the Wurster system, film thickness varies with particle size within a batch; top- or
18 tangential-spray fluid beds rarely show this variation. This is due, in part, to the slow circu-
19 lation of lighter and/or smaller particles, a pattern inherent to the Wurster process
20 (Ichikawa et al., 1996).
21 Regardless of the coating unit configuration chosen, film formation around solid
22 particles cannot be achieved by a single pass through the coating zone, but requires many
23 such passes to produce complete particle coverage. The presence of any loose uncoated
24 actives can also have detrimental effects on the release mechanism and overall stability of
25 the finished product. Figure 5.2 shows a schematic of the steps involved in particle coating
26 and film formation in a fluid bed–coating unit.
27 Coat integrity and subsequent release of the active require careful combination of sev-
28 eral parameters such as air velocity, air temperature, spray rate, spray droplet size and so
29 on. Jozwiakowski et al. (1990) published an excellent paper detailing the impact of
30 substrate’s physicochemical properties on coating quality and efficiency in a fluid bed sys-
31 tem. Their study highlighted the importance of two types of interactions, namely
32
33
(a) (b) (c)
34
35
36
37
38
39
40
41
42
43
44 Bottom spray Tangential spray
45 Top spray (Wurster coating) (rotor pellet coating)
46S Figure 5.1. Various configurations of fluid bed–coating systems: (a) top spray, (b) bottom spray
47N (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 115

Encapsulation and Controlled Release in Bakery Applications 115

Spraying Wetting Congealing Coated particle 1


2
3
4
5
6
7
8
9
10
11
Particle coating droplets Film formation 12
13
Figure 5.2. Film formation principle in a fluid bed–coating system. (Courtesy of Glatt Air 14
techniques, with permission.) 15
16
17
particle–particle and particle–machine, and concluded that an ideal substrate should pos- 18
sess essential attributes such as spherical shape, uniform (high) bulk density, narrow 19
particle size distribution, and chemical stability. 20
21
22
Effect of Substrate’s Physicochemical Properties 23
It is critical to point out that film coating in a fluid bed system is applied on a weight basis. 24
Therefore, to achieve same film thickness, larger amounts of shell material are needed to coat 25
small particle cores (Madan et al. 1974). Coat thickness has been shown to be directly related 26
to substrate’s particle diameter but inversely proportional to its surface area (Table 5.1). 27
Particle shape, porosity, and friability can also play an important role in determining film 28
quality. Irregular-shaped particles such as crystals (salts, sodium bicarbonate) require 29
larger amounts of coating (in excess of 80% of microcapsule’s weight) and can most often 30
lead to nonuniform film formation. 31
In coating applications, particle–particle interactions manifest themselves in two differ- 32
ent phenomena, agglomeration and attrition. Fluidization of wet fine particle cores (<100 µm 33
diameter) under intensive motion in the bed vortex can lead to particle–particle collision and 34
agglomeration. The latter can be dramatically magnified when the fluid bed is operated at 35
temperatures too close to the melting temperature of the coating material or when using very 36
high rates of spray coating. Ideal core particle size for fluid bed encapsulation ranges from 37
38
39
Table 5.1. Effect of particle size on wall thickness. (Adapted from Madan et al., 1974, with
40
permission from the publisher.)
41
Calculated surface Number of particles 42
Particle diameter (µm) area of particles (n  10–4) Wall thickness, T (µm) 43
235 624 17.7 0.26  0.02 44
505 414 5.17 0.49  0.03 45
715 292 1.82 0.64  0.02 S46
840 249 1.12 1.31  0.13
N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 116

116 Chapter 5

1 100 to 800 µm (assuming optimal bulk density). Larger size particles or pellets (>1 mm)
2 can be coated readily, their repeated cycling in the bed may lead to particle abrasion and
3 attrition. Such cores should be coated for only short time intervals with minimum bed
4 movement during the warming period (Lehmann and Dreher, 1981).
5 Figure 5.3 shows a scanning electron micrograph of typical coated particle surrounded
6 by a lipid/wax wall material with the substrate particles completely engulfed in the
7 lipid/wax shell.
8
9
Spray Chilling
10
11 Fluid bed coating described above is, in essence, an enrobing mechanism whereby one or
12 few particles (100–400 µm) are enveloped in a coating film, forming a reservoir-like sys-
13 tem. As the temperature surrounding the capsule reaches the melting point of the wall
14 material, the entrapped particles are released. However, in the presence of slightest imper-
15 fections in the shell material, the actual release tends to shift to “burst-like” behavior. The
16 latter can have detrimental consequences upon storage and preparation of dough or batter
17 systems, resulting in premature or uncontrolled release of the encapsulated active.
18 Spray chilling is an alternative technique that has been used for years in manufacturing
19 stable pharmaceutical capsules with a unique matrix release mechanism. This technique is
20 a solvent-free spray-drying method for encapsulating water-sensitive actives. In this
21 process, fine particles (typically <100 µm) are dispersed in a hot melt fluid (waxes, fatty
22 acids) to form a homogeneous dispersion. The latter is atomized via spraying through a
23 pressurized single nozzle into a cooled chamber. The chamber temperature is set below the
24 melting point of the mixture or its individual components using nitrogen or carbon dioxide
25 gas. Ideally, spray chilling results in the formation of uniform spherical micropellets with
26 smooth surfaces that are water-impermeable but not water-resistive. These qualities are
27 essential for better mixing owing to reduced surface tension between the microcapsule’s
28 hydrophobic surface and the batter’s aqueous environment. Due to the absence of solvent
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S Figure 5.3. Surface morphology of a coated solid particle. (Courtesy of Balchem Corp., with
47N permission.)
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 117

Encapsulation and Controlled Release in Bakery Applications 117

evaporation in spray chilling operations, the particles are generally non-porous and 1
mechanically-strong so that they remain intact upon agitation (Gherbre-Sellassie, 1989). 2
Nanosized particles can also be prepared using this process (Eldem et al., 1991). Additional 3
coating is often applied to spray chilled congealed particles to ensure complete coverage of 4
the microparticle and to eliminate undesirable interactions of exposed actives with their 5
surroundings during storage and dough/batter preparation. 6
Conventional spray dryers with cooled air inlets can be used for spray chilling. The 7
apparatus consists of two main parts: (1) a cooling chamber and (2) an atomizer. For effec- 8
tive spray chilling, it is recommended that the dispersion matrix has a very narrow melting 9
range so that the particles can be held together during spraying. 10
Critical conditions for manufacturing uniform congealed capsules are: (1) low viscosity 11
of the active and molten fat dispersion. Das and Gupta (1988) suggested that an ideal vis- 12
cosity should be around 24 cP at 55°C and (2) high atomization speed to increase the per- 13
centage of small micropellets (Scott et al., 1964; Deasey, 1984). 14
Release of actives from spray chilled microcapsules takes place via erosion and leaching 15
through the matrix. Surfactants (depending on type and concentrations) can also dramati- 16
cally affect matrix dissolution rates. John and Becker (1968) demonstrated that addition of 17
4% of the non-ionic surfactant sorbitan monooleate resulted in enhanced release rates from 18
a wax-congealed matrix; however, increasing the monooleate concentration to 10% led to a 19
reduction in rate of release. 20
Spray chilling suffers from one main drawback, that is, the rapid cooling rates can some- 21
times crystallize the triglyceride matrix in the unstable α-polymorphic form, leading to the 22
formation of disordered chains with undesirable orientation and, subsequently, low barrier 23
properties. 24
25
26
High-Pressure Congealing (Beta Process)
27
To overcome these drawbacks, a modified method was advanced by Verion Inc. (Redding, 28
1995; Vaghefi et al., 2001). The process involves forming an active-matrix dispersion (e.g., 29
sodium bicarbonate dispersed in molten fat or wax) and further subjecting the mixture to 30
high pressure (40,000–50,000 psi) for few seconds to intimately compress the mixture. The 31
sodium bicarbonate/fat mixture is then discharged through a spray nozzle into a chilling 32
zone to congeal the molten fat material around the particles. The mixture is allowed to 33
cycle through the system for multiple passes depending on the active load and/or desired 34
capsule matrix consistency. The high pressure and high shear applied result in favorable 35
changes in the polymorphic structure of the treated fat or wax and in shifting its polymor- 36
phic structure into the stable beta (β)-form, thus the name Beta process. 37
Redding (1995) studied the impact of heat, pressure, and their combinations on changes 38
in the differential scanning calorimetric (DSC) profiles of tristearin (Figure 5.4) using the 39
β-process system. Commercially obtained native triglyceride displayed a β-melting peak at 40
72°C. Upon melting and further resolidification, the DSC profile showed, in addition to the 41
β-peak, a new peak at 59.84°C corresponding to the α-form. Heating the tristearin to 42
145°C along with pressure application (4400 psi) led to the complete elimination of the 43
α-peak and the dominance of a stable β-peak at 75.73°C. 44
Similar to low-pressure congealing, complete coverage of the active particles located 45
on the microparticle surface can be ensured by applying an outer coating layer via fluid-bed S46
or other coating techniques. Capsules prepared, using this process, normally follow true N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 118

118 Chapter 5

1
(a)
2
3

Heat flow (w/g)


4
5
6
7
8
9 72°C
10 20 40 60 80 100
11 Temperature (°C)
12
13 (b) 16
14
Heat flow (w/g)

15 14
16
12
17
18 10 59.84°C
19 8
20 77.86°C
21 6
20 40 60 80 100
22 Temperature (°C)
23
24 (c)
25 20.5
26 20
Heat flow (w/g)

27 19.5
28 19
29 18.5
30 18
31 17.5 75.73°C
32 17
33 20 40 60 80 100
34 Temperature (°C)
35
36 Figure 5.4. Effect of temperature and pressure on polymorphic profile of stearine: (a) native,
37 (b) melted and resolidified, and (c) treated at 145°C and 4400 lb/in.2. (Reproduced from Redding,
38 1995, with permission from the publisher.)
39
40 zero-order release mechanism, a result of the slow erosion from the microcapsules that
41 form “tortuous” paths throughout the matrix. The shell material is not swellable and does
42 not rely on osmotic pressure to release the core material.
43
44
Film-Forming Materials
45
46S A variety of fats and waxes are available for hot melt coating of leavening systems
47N (Table 5.2) Lipid-based coating materials are available as pure components or most often as
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 119

Encapsulation and Controlled Release in Bakery Applications 119

Table 5.2. Source and melting temperatures of selected group of waxes, lipids, and resin 1
compounds used in particle-coating applications. (Adapted from various sources.) 2
Product Source Melting point (°C) 3
4
Beeswax Bees 61–64
Carnauba wax Tree of life 82–86
5
Candelilla wax Candelilla plant 65–69 6
Shellac Laccifer lacca insect 115–120 7
Lauric acid Coconut oil 44 8
Capric acid Coconut oil 31.6 9
Myristic acid Coconut oil, butter fat 54.4
Stearic acid Most fats/oils 69.6
10
Behenic acid Peanut oil 80 11
Palmitic Most fats/oils 62.9 12
Stearine Partially hydrogenated 61–64 13
Cottonseed oil 14
Stearine Partially hydrogenated 66–70
Soybean oil
15
16
17
18
functionally optimized composites (Kanig and Goodman, 1962; Kester and Fennema, 19
1989a). Blends of lipids (different hydrophobicity, chain length, hardness, melting point, 20
etc.), lipids and waxes, and lipids and polysaccharides can be adequately formulated to 21
encapsulate actives for bakery and other food applications. 22
23
24
Waxes
25
Natural and synthetic waxes have been used in particle-coating applications. The most 26
commonly used materials include: paraffin, carnauba, candelilla, beeswaxes and/or wax 27
emulsions. 28
Paraffin wax: Paraffin wax is derived from the wax distillate fraction of crude petroleum. It 29
is composed of hydrocarbon fractions of generic formula CnH2n+2 ranging from 18 to 32 30
carbon units (Hernandez, 1994). Refined paraffin waxes can be used in specific coating 31
applications (21 CFR, Code of Federal Regulations, 184.1973). 32
Carnauba wax: Carnauba is a plant-derived exudate from the leaves of the Tree of Life 33
(Copernica Cerifera) found mostly in Brazil. Carnauba wax consists mostly of saturated 34
wax acid esters with 24–32 hydrocarbons and saturated long-chain monofunctional alco- 35
hols such as myricyl cerotate alcohols C9H59CH2OH. 36
Carnauba wax is the hardest natural wax available (hardness 4.7 cm  102 for a 37
50 g/60 sec/25oC) and has the highest melting point and specific gravity of commonly 38
found natural waxes (82–86oC). It is added to other waxes to increase melting point, hard- 39
ness, toughness, and luster. However, carnauba wax is very brittle and lacks elasticity. Car- 40
nauba wax is allowed for specific applications in food systems (21 CFR 184.1978). 41
Beeswax: Beeswax is a secretory product of honey bees and is the basic material for comb 42
building. Beeswax is harvested after removal of the honey by draining or centrifuging and 43
further melting with hot water, steam, or solar heating. The wax is separated from impuri- 44
ties by treating with diatomaceous earth and activated carbon. Beeswax is made up essen- 45
tially of long-chain alcohols (C24–C33), hydrocarbons (C25–C33), and long-chain acids S46
(C24–C34). Beeswax is very plastic at room temperature (melting point 61–65oC) but N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 120

120 Chapter 5

1 becomes brittle at colder temperatures. It is soluble in most other waxes and oils (Tulloch,
2 1970; Bennett, 1975). Beeswax is considered a GRAS substance and is allowed for direct
3 use with some limitations (21 CFR 184.1973).
4 Candelilla wax: Candelilla wax is obtained from the candelilla plant that grows mostly in
5 Mexico and southern Texas. The wax is prepared by immersing the plant in boiling water
6 containing sulfuric acid and further skimming off the surface, refining, and bleaching. Its
7 degree of hardness is intermediate between beeswax and carnauba. It contains small
8 amounts of esters and free acids. This wax sets very slowly and takes several days to reach
9 maximum hardness. Candelilla wax is considered GRAS and is allowed for certain food
10 uses (21 CFR 184.1976).
11 Wax macro- and microemulsions: Carnauba and beeswax owing to their high content of
12 alcohol and ether groups, can be microemulsified to form effective coating materials.
13 Waxes are dispersed in water to form macro- or microemulsions via a process commonly
14 known as inversion (Wineman, 1984).
15
16
Resins and Rosins
17
18 Shellac: Shellac resin is a secretion by the insect Laccifer lacca and is mostly produced in
19 central India. This resin consists of a complex mixture of aliphatic alicyclic hydroxyl acid
20 polymers, that is, aleuritic and shelloic acids. It is soluble in alcohol and alkaline media.
21 Shellac resins can be blended with waxes to form improved moisture-barrier properties and
22 increased gloss for coated products. Shellac is not GRAS; it is only permitted as an indirect
23 food additive in food coatings and adhesives (21 CFR 175.300). Shellac is rich in car-
24 boxylic acid residues and is highly water insoluble (Sward, 1972).
25
26
Glycol Polymers
27
28 Polyethylene glycols such as Carbowax (different grades, that is, viscosities 3350, 4600,
29 8000) possess desirable coating properties, mainly their resistance to abrasion. Levels of
30 15–40% were found to be useful for coating yeasts and extending their viability (Percel,
31 1988).
32
33
Fats and Glycerides
34
35 A wide variety of commercially available triglycerides are used in coating applications.
36 Naturally occurring food-grade fats are derived from animal or plant origin. Animal
37 triglycerides differ from plant triglycerides not only in the ratios of saturated to unsaturated
38 carboxylic acids or their chain length but also in the location of the unsaturated fatty acid in
39 the glyceryl molecule. Variations in the carboxylic acid chain length, their melting profile,
40 degree of saturation, degree of esterification, purity grades as well as their crystalline struc-
41 ture can have a significant impact on coating processability as well as the performance of
42 the encapsulated product.Natural oils and fats used in coating applications consist of one or
43 more of the three major fatty acid groups; these are lauric, palmitic, and oleic–linoleic
44 groups:
45 Lauric acid group: Fatty acids of this group are highly saturated, rich in short-chain fatty
46S acids (8, 10, and 14 carbon chain length), and are very stable. They contain 40–50% lauric
47N acid on average. Oleic and linoleic acids constitute the majority of the unsaturated
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 121

Encapsulation and Controlled Release in Bakery Applications 121

fractions, while saturated ones are essentially made up of palmitic and stearic acids. 1
Examples of this group include palm seed oil, canola, coconut, babassu, and palm kernel 2
oils. Lauric-acid-based fatty acids have relatively low melting points (~44°C). Hydro- 3
genated canola oil, for example, is composed of a triglyceride with extremely symmetrical, 4
nonrandom structure, resulting in a hard and dry fat with good flowability at room temper- 5
ature. Coconut oil has melting point of 24°C, while its hydrogenated counterpart melts at 6
33°C (Bailey, 1952). 7
Palmitic acid group: This group is represented by palm oil (from palm pulp) and palm ker- 8
nel oil (from kernel). Palm oil contains 32–47% palmitic acid and 40–52% oleic acid. Palm 9
oil has equal concentrations of the saturated and unsaturated fatty acids. Most of the 10
triglycerides (85%) of palm oil contain unsaturated fatty acids at the 2-position of the glyc- 11
erol backbone (Bailey, 1952). 12
Oleic/linoleic acid group: Commercially important oils in this group include corn, cotton- 13
seed, peanut, olive, sunflower, safflower, and rice bran oil. These oils can be hydrogenated 14
to form plastic fats with different degrees of hardness. Most oils in this group are short- and 15
medium-chain unsaturated fatty acids. Only the highly-hydrogenated versions of these 16
fatty acids can be effective in particle coating applications. 17
18
19
Characteristics of Wax and Fat-Coating Materials 20
21
Chain Length
22
Long-chain fatty acids such as stearic (C 18:0) and palmitic (C 16:0) acids, by virtue of 23
their high melting and apolar properties, have been used extensively in food coating appli- 24
cations (Hagenmaier and Baker, 1991; Greener and Fennema, 1993; Kester and Fennema, 25
1989b; McHugh and Krochta, 1994). Due to their strong H-bonding, long-chain fatty acids 26
such as stearyl alcohols, stearic acid and beeswax crystallize into platelet-like dense 27
microstructures, commonly associated with effective moisture barrier properties (Figure 5.5). 28
Longer-chain triglycerides (higher than 18 carbon atoms) such as arachidonic or behenic 29
acids, however, show higher permeability presumably due to the heterogenous structure of 30
the polymer network. 31
32
33
Polarity
34
Stearyl alcohol, a polar molecule, is a better barrier than its fatty acid counterpart, a result 35
of the lower affinity of the hydroxyl group for water than carbonyl and carboxyl groups. 36
However, in most applications, other factors such as chain structure and its conformation 37
should be taken into consideration when choosing a lipid barrier material. 38
39
40
Degree of Unsaturation
41
The degree of unsaturation plays a considerable role in defining the crystal structure of 42
triglycerides and their mobility. For example, the area occupied by a molecule of oleic acid 43
in a monolayer film is 0.48 nm2, whereas for stearic acid, it is 0.23 nm2 (Kamper and 44
Fennema, 1984). Despite this fact, oleic acid displays greater mobility owing to the double 45
bond that favors the diffusivity of water molecules compared to stearic acid, which is a fully S46
saturated carboxylic acid (Gontard et al., 1994). N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 122

122 Chapter 5

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15 Figure 5.5. Cross section of wax-coated particle showing platelet-like microstructure. (Courtesy
16 of Balchem Corp., with permission.)
17
18
19
20
Solid Fat Index
21
22 Solid fat index (SFI) has been directly related to fat hardness and its spreadability (Bailey,
23 1952; Vreeker et al., 1992). High solid fat concentration (0–30%) improves barrier proper-
24 ties of fat-based films but increases their hardness (Narine and Margoni, 2002). High solid
25 fats have better barrier properties than their corresponding liquid or semi-solids due to the
26 formation of dense structure and greater volume that limits the diffusion of water (Perron
27 and Ollivon, 1992). However, at very high solid fat concentration, higher than the critical
28 value, permeability could increase due to structural defects within the film.
29
30
Hydrophobicity
31
32 Hydrophobicity has been used as a criterion for predicting water barrier properties of fats
33 and waxes. Kester and Fennema (1989a, b) proposed the following order of decreasing
34 moisture barrier effectiveness of waxes and fats: beeswax, stearyl alcohol, acetyl glycerols,
35 hexatriacontane, tristearin, and stearic acid, a reflection of their decreased hydrophobicity.
36 Avner and Blatt (1990) indicated that despite the similarities in melting temperatures
37 between hydrogenated castor oil and calcium stearate/stearic acid blend (melting
38 point~86°C), hydrogenated castor oil provided higher thermal stability and barrier proper-
39 ties due to the superior hydrophobic character of castor oil.
40
41
Polymorphism (Crystallization Behavior)
42
43 Triglyceride molecules are naturally arranged in a “tuning fork” structure where three fatty
44 acids are more or less parallel, one pointing in the opposite direction of the others. How-
45 ever, the complexity and flexibility of triglyceride molecules allow different crystalline
46S packing of the same ensemble of molecules, leading to the existence of different poly-
47N morphs. During crystal growth, such molecules will pack more easily side to side than end
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 123

Encapsulation and Controlled Release in Bakery Applications 123

to end with glyceryl alcohols. The resulting individual crystals appear needle shaped under 1
the microscope (Bailey, 1952; van de Tempel, 1961). 2
Fat polymorphism, that is, their ability to crystallize under several forms, also affects 3
their barrier efficiency. Triglycerides crystallize into α (hexagonal), β (triclinic), and β′ 4
(mainly orthorhombic) forms. The latter two crystals are stabilized by London forces 5
occurring between the aliphatic chains, which give rise to dense networks. No such forces 6
occur in the α-form. The carbon atom can rotate (small angles), leading to a hexagonal 7
nonstable structure (Sato and Kuroda, 1987 and therein; Ubbelohde, 1978). The β-form, 8
represented by lard and tallow, has the highest melting point and highest order (large and 9
coarse crystals 25–50 µm long). β′-crystals, however, provide the most functional form 10
owing to their small size (less than 1 µm), thin needle-shaped morphology, and interlocking 11
structure (Roberts et al., 2000). Polymorphic states of a substance have different physical 12
properties but on melting yield identical liquids since these states are merely due to differ- 13
ences in packing of the constituent molecules upon crystallization. 14
15
16
Melting Point 17
18
Most naturally occurring fats show multiple melting points reflected in several melting
19
peaks in their DSC profile. Low melting fats most often result in inferior coating qualities
20
due to their poor flowability and tendency to form clumps. On the other hand, high melting
21
fats such as palm stearine may cause difficulties in spray coating encapsulation due to their
22
stiffness and lack of plasticity. The melting point of polymorphic forms of stearine range
23
from 65°C (α) to 70°C (β′) and 72°C (β) (Bailey, 1952).Low melting lipids are useful for
24
coating heat-labile actives such as yeasts, enzymes, vitamins, probiotic bacteria and so on.
25
High melting lipids, on the other hand, are suitable for encapsulating chemical leavening
26
agents, acids, and other less heat-sensitive actives.
27
Despite the positive impact of high melting fats on fluid bed–coating efficiency, coating
28
performance of a given fat is a function of the melting profile and not necessarily the tem-
29
perature of the melting peak. It has been a common practice to source pure fats/waxes
30
(fewer components) in order to provide a single narrow-shaped peak; in practice coating
31
conditions, in particular the rate of fat/wax cooling, are as critical in determining the sharp-
32
ness or broadness of the melting peak.
33
The mechanical properties of lipid-based films can be improved by modifying either the
34
film’s melting profile or its structural properties. Two classes of materials have been used to
35
modify the performance of fats, namely, waxes and plasticizers:
36
37
1. Waxes can be successfully used to raise the melting point of natural fats, thereby reduc- 38
ing tackiness and enhancing their coating performance. For such applications, it is criti- 39
cal to maintain the fat/wax blend under continuous mixing and at temperatures slightly 40
above the wax-melting point to avoid wax gravity settling and subsequent plugging of 41
the spray nozzle. In the petroleum industry, the temperature at which wax crystals start 42
to appear when temperature falls to a critical level referred to as wax appearance tem- 43
perature, WAT (Azvedo and Teixeira, 2002). Prolonged heating of wax/fat mixtures, 44
however, may lead to fat oxidation; therefore caution should be exercised and whenever 45
possible, incorporating antioxidants or blanketing the hot melt container with nitrogen S46
can be very effective in retarding fat oxidation and/or degradation. N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 124

124 Chapter 5

1 2. Plasticizers such as acetoglycerides, glycerin, monoglycerides, alcohols, phospholipids


2 (lecithins) and ester derivatives of glycerols can be used to lower the melting point of
3 fats and waxes, thus facilitating their atomization from the spray nozzle. Plasticizers can
4 also help in reducing film brittleness and enhancing its flexibility and mechanical prop-
5 erties without any significant impact on the melting point of the wax. Litwinenko et al.
6 (2004) reported that addition of glycerol increased the mean crystal size of fats up to a
7 concentration where the crystal size decreased with increased glycerol concentration
8 (Table 5.3).
9
10 Films made from molten beeswax are often smooth and uniform, whereas those made with
11 alcohol are rough and irregular with apparent large size globules (Greener and Fennema,
12 1993). Incorporation of polysaccharide molecules such as ethyl cellulose into fat-based
13 films can be used to provide additional film toughness. The latter is presumably a result of
14 the proper orientation of the wax crystallites parallel to the polysaccharide support base.
15
16
17 Ideal Properties of Encapsulated Particles for Bakery Applications
18 Ideal microcapsules manufactured for baking applications should possess the following
19 essential properties.
20
21
22 Good Barrier Properties
23
24 Fats of large and closely packed crystals are favored for their high moisture barrier proper-
25 ties. As discussed above, barrier properties are a combination of fat/wax chain length,
26 polarity, polymorphic form, film flexibility, and so on.
27
28
Flexibility
29
30 Fat crystals form a particular class of soft materials that demonstrate yield stress and vis-
31 coelastic properties, that is, plastic-like materials (Narine and Marangoni, 1999; 2002).
32 Waxes, on the other hand, form stiff films that tend to become fragile, especially if stored
33 for long periods. Film stiffness could also lead to microcapsule fracture and rupture during
34 blending.
35
36
37 Table 5.3. Effect of glycerol on microstructural parameters determined by image analysis.
(Adapted from Litwinenko et al., 2004, with permission from the publisher.)
38
39 Crystallization temperature (C) Glycerol (%) Mean particle size (µm2) Number of particles (N)
40 Following storage for 24 h at 5°C 0.00 40 4845
41 0.03 30 4630
42 0.10 86 3300
43 0.25 69 3646
44 Following storage for 15 min at 5°C 0.00 163 1386
45 0.03 302 685
0.10 326 679
46S 0.25 150 1555
47N
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 125

Encapsulation and Controlled Release in Bakery Applications 125

Mechanical Strength 1
2
Good mechanical properties are critical for processing intact microcapsules without fracture. 3
Edible fats have inherently poor mechanical strength. The latter can be enhanced by incorpo- 4
rating waxes, by modifying their hydrophobic character (Narine and Marangoni, 1999), or by 5
addition of polar substances such as hydrocolloids (Chen and Nussinovitch, 2000). 6
7
Surface Morphology 8
9
Smooth surfaces that are free of cracks or crevices are ideal for formulating bakery prod-
10
ucts with microencapsulated actives and for controlling their release, that is, by retarding
11
microcapsule leaking and premature release.
12
13
Particle Size Distribution 14
15
Uniform particle size distribution is critical for many applications, especially in situations
16
where only a very narrow melting window is available. Very large microparticles or micro-
17
capsules can cause localized effects such as failure to distribute evenly and eventually form
18
immobile melted masses of barrier material. Very small particles, on the other hand, will have
19
a large surface area which speeds up melting of the coating material upon baking the product.
20
21
Film Thickness 22
23
In encapsulation and film coating, one critical decision to be made is how much coating is
24
necessary to achieve desired properties in the finished product. Applied films should be
25
thick enough to overcome surface imperfections but not to modify the release behavior of
26
the microcapsule. For stable dough and batter systems, coatings in the range of 50–95% are
27
common. It should be noted that high film thickness can modify the release of actives in an
28
unpredictable manner and at extreme levels, the release rate tends to become dependent on
29
the size of the core, regardless of the film thickness.
30
31
Melting Properties 32
33
Choice of suitable melting properties of the coating material should take into consideration
34
the bakery product’s desired storage and processing conditions. Figure 5.6 shows a typical
35
temperature/time profile of baked batter in a conventional oven. An ideal leavening micro-
36
capsule for this application should have a melting profile that closely mirrors that of the
37
baked product. Melting of the shell material should commence as the product temperature
38
approaches or attains 200ºF (93ºC) and should be completed around the first 13–14 min
39
of the baking cycle, that is, before the baked product structure is fully set.
40
41
Miscellaneous Examples of Encapsulation and Controlled Release 42
in Bakery Applications 43
44
Yeasts
45
Yeasts used in baking applications include mainly Saccharomyces cerevisiae, Saccha- S46
romyces rosei, Saccharomyces exiguous, and Candida milleri. In the absence of oxygen, N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 126

126 Chapter 5

1 Muffin temperature development

Batter temperature (°F)


2 250
3 200
4
150
5
6 100
7 50
8
0
9
1 3 5 7 9 11 13 15 17 19 21 23 25
10
Time in oven (min)
11
12 Figure 5.6. Temperature development of baked muffin over time.
13
14 yeast cells metabolize carbohydrates, via multiple intermediate steps, to produce alcohol,
15 energy, and carbon dioxide gas (Equation 1). The latter is essential for building volume and
16 cell structure in the baked product. Yeast activity takes place during dough preparation and
17 can sharply increase as temperature rises upon preparation and baking (rate doubles for
18 every 10°C rise in temperature).
19
20 Yeast fermentation
21 C6H12O6 → 2C2H5OH  2CO2  234 kJ (1)
22
23 Baker’s yeast is available in many forms: active dry yeast (ADY), inactive dry yeast,
24 compressed yeast, cream yeast, crumbled yeast, and protected dry yeast (Fleischmann’s,
25 2001). Although active dry yeast (ADY) has a relatively low moisture content (7.3–8.3%),
26 it is sensitive to oxygen and is always distributed in hermetically sealed pouches under vac-
27 uum or flushed with liquid nitrogen. Cream yeast and compressed yeast are usually stored
28 at subfreezing temperatures to maintain their activities. However, problems arise if any of
29 these yeast forms is incorporated into a dough system before freezing because of uncon-
30 trolled fermentation and dough expansion (Reed and Nagodawithana, 1991).
31 Many attempts to improve the keeping qualities of Baker’s yeast have been documented
32 (Pelletier and Roger, 1989). Addition of hydrophilic agents (starch, locust bean gum) to
33 yeast preparations to bind up water and potentially retard endogenous metabolic processes
34 has not proven to be very effective. Other technical approaches involve immobilization in
35 chemically cross-linked chitosan beads or activated carriers (Donova et al., 1993;
36 Markvicheva et al., 1991; Freeman and Dror, 1994; Shimon et al., 1991). Luca et al.
37 (1979)) patented a process for treating fresh yeast with hydrophobic silicon dioxide
38 (0.2–1%) to form a colloidal dispersion that was claimed to help maintain the yeast stable
39 at high moisture contents. Soltis and Sell (1998) developed a method for encapsulating
40 Baker’s (cream) yeast and forming a shelf-stable product that can be safely stored at ambi-
41 ent conditions, thus eliminating the need for costly refrigerated storage. Their method con-
42 sists essentially of pre-adsorbing the high moisture cream yeast onto food fiber, such as
43 grain or bean hulls, followed by coating with a thermoplastic hydrophobic material. The
44 system was claimed to provide a means for delaying the release of active yeast cells until
45 later in the baking process.
46S Newly developed forms of ADY (high ADY) possess high fermentative power and are
47N instantized to allow their incorporation into dough systems without rehydration. However,
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 127

Encapsulation and Controlled Release in Bakery Applications 127

this process results in porous granules that may expose the yeast to undesirable conditions, 1
resulting in their inactivation within three days. Percel (1988) devised a method for preserv- 2
ing the viability of instant yeast via coating (15–40% w/w) with polyethylene glycol of dif- 3
ferent molecular weights (Carbowax 3350, 4600 and 8000) under mild temperatures of 4
54–63°C using fluid bed coating. These experiments showed that Carbowax 4600 provided 5
most immediate release of the yeast in cold water while Carbowax 8000 resulted in yeast 6
micropartculates with most abrasion resistance and stability (several weeks in dry mixes 7
compared to few days for the unencapsulated yeast). Fuglsang et al. (2002) used liposome 8
systems to entrap yeast and inhibit their release at ambient temperature. Upon heating from 9
25 to 60°C, the lipid undergoes phase change, thus releasing the encapsulated yeast material. 10
11
12
Chemical Leaveners
13
Manufacturing consistent quality bakery products, especially in large volume operations, 14
relies on using chemical leaveners such as baking powders for cakes, muffins, and cookies 15
and to replace yeasts in bread and dough formulations. Chemical leavening agents operate 16
differently from yeast; while yeast requires sitting time after thawing and prior to baking to 17
produce carbon dioxide, a chemical leavening system produces CO2 gas during baking to 18
expand the dough and create cellular microstructure. 19
Chemical leavening agents comprise a long list of acids and alkalis that vary in their reac- 20
tivity, stability, solubility, as well as other attributes. (Church and Dwight, 1999). Leavening 21
systems comprise two main components, leavening acid such as sodium acid pyrophosphate 22
(SAP) and base pair such as sodium bicarbonate (soda) that when allowed to react in an aque- 23
ous medium (in some cases in the presence of heat) results in the formation of CO2. In such 24
systems, the bicarbonate will provide the gas while the acid will control the reaction rate. 25
Common leavening agents are classified into three categories: (1) slow acting, (2) fast 26
acting, and (3) double acting. Sodium aluminum sulfate (SAS) and monocalcium phos- 27
phate monohydrate (MCP) form a double-acting system. Slow-acting systems include a 28
combination of soda and SAP, where baking temperatures affect CO2 release. In fast-acting 29
systems (soda and MCP), CO2 evolution and its release occur effectively during mixing and 30
standing. Deciding which component of the leavening system to encapsulate will depend 31
greatly, among other aspects, on whether the product is a dry mix, bread dough, or a high 32
water activity batter. In high water activity formulations, encapsulating the soda component 33
is more feasible since the acid component can help provide additional antimicrobial protec- 34
tion to the batters. 35
Chemical leavening agents are sensitive to moisture but much less to storage and prepara- 36
tion temperatures; therefore, encapsulating chemical leaveners in a hydrogel system, for 37
example, would not be an option owing to the risk of dissolving the active prior to its point of 38
application. Hydrophobic coatings such as fats and waxes constitute the most commercially 39
viable encapsulating media. Upon heating, the shell melts, thus allowing the leavening 40
active to become available and ultimately to release CO2 needed for building volume and 41
cell structure of the baked product. The rate of capsule melting and CO2 evolution are criti- 42
cal parameter for determining volume, density, and textural qualities of the baked product. 43
This step must occur within narrow limits for some applications such as in the preparation of 44
canned doughs. Manipulating the release rate can be achieved by choosing suitable fats 45
and/or waxes with adequate melting temperatures as well as melting profile, coat thickness, S46
encapsulation technology, and release mechanism (reservoir vs. matrix vs. combination). N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 128

128 Chapter 5

1 For most bakery systems, encapsulating chemical leaveners via particle coating requires
2 the application of fairly high levels of coating materials onto the crystalline core; ideal
3 coat:core ratios range from 50:50 to 95:5 to ensure complete coverage of the active’s sur-
4 face. Canned self-sealing doughs represent an exceptional application, where the presence
5 of small amounts of partially-coated or uncoated leavening is desirable. In such situations,
6 the partially-coated or uncoated agents react to form CO2 during or immediately following
7 packaging of the can, thus purging out air and providing a seal from inside the package. The
8 fully-coated portion of the leavening microcapsules is preserved for further reaction during
9 baking. Care should be taken not to allow excessive evolution of CO2 and over-expansion
10 of the pressurized dough.
11 Huang et al (1989) cited an interesting advantage of encapsulating leaveners for
12 microwave baked products i.e. their ionic interactions with the dough components and the
13 subsequent positive impact on reducing gluten toughness and starch firmness, important
14 attributes to forming stable matrix for stabilizing generated CO2.
15 Assessing the stability of encapsulated sodium bicarbonate is achieved by tracking the
16 release of carbon dioxide in sealed packages or containers as well as changes in pH,
17 appearance, and other sensory attributes of the stored product. Figure 5.7 shows a compari-
18 son of the risograph gas evolution in refrigerated doughs made using two commercially
19 encapsulated soda products and one unencapsulated control, with E-soda 1 displaying
20 highest stability (lowest CO2 release during storage) while unencapsulated soda showing
21 the least stability (Domingues et al., 2003).
22 Pacifico (2003) suggested that leach rate upon baking (rate at which an encapsulated
23 agent seeps out from the capsule) can be a useful indicator of the functionality of encapsu-
24 lated leaveners. Accordingly, high leach rate of congealed and coated actives may enhance
25 their reactivity and does not necessarily imply ineffective encapsulation.
26 Several other publications and inventions have surfaced in the last decade claiming
27 ingredients and methods for manufacturing chemically-leavened shelf stable bakery
28
29
30
31 60
32 free CO2
33 50
34
35 40
CO2 (%)

36 E2-Soda
30
37
38 20
39
40 10 E1-Soda
41
0
42
0 200 400 600 800 1000 1200 1400
43
44 Time
45 Figure 5.7. Release of carbon dioxide from refrigerated dough package made with
46S unencapsulated and two encapsulated soda samples (E1-soda and E2-soda) and stored at
47N 45°F for six weeks. (Reproduced from Domingues, 2003.)
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 129

Encapsulation and Controlled Release in Bakery Applications 129

products (Book et al., 2000; Chung and Lavault, 1995; El-Afandi and Citti, 2006; 1
Kringelum et al., 1999; LaBell, 1999; Dorko and Penfield, 1993; Redding, 1995; Redding 2
and Bruce, 2002; Tuazon and Foster, 1992; Wu et al., 2000); each of these inventions is 3
directed to a specific bakery application. 4
5
6
Dough Conditioners
7
Bread staling and loss of freshness are major economical hurdles for the baking industry. Sev- 8
eral underlying mechanisms have been speculated, with the amylose molecular rearrange- 9
ments theory being the most plausible. During baking, starch is gradually transitioned from 10
an amorphous structure to a partially crystalline state, a result of inter- or intramolecular 11
interactions via H-bonding of the amylose and amylopectin fractions. Upon recrystallization 12
(retrogradation), starch releases water and the crumb becomes very firm and stale. 13
Storing baked products at room temperature or under high (safe) relative humidity can 14
delay staling, but only for few hours. Emulsifiers are commonly used to help retard staling, 15
though not very effectively; their mechanism of action is believed to be via softening the 16
bread and reducing its firmness rather than retarding starch retrogradation. 17
Amylases that modify starch responsible for staling can be used effectively for increasing 18
shelf life of bread via hydrolysis of the glycosidic linkages in polyglucans. Most commonly 19
used α-amylases are derived mainly from Aspergillus oryzae, Bacillus subtilis, and Bacillus 20
stearothermophilus. These enzymes act on damaged starch particles, thus lowering dough 21
viscosity and producing fermentable sugars necessary for larger bread volume and softer 22
loaves. The Bacillus-derived amylases, however, are fairly heat stable and therefore do not 23
get inactivated during baking, resulting in excessive breakdown of starch and the formation 24
of very moist and sticky crumbs that are difficult to control. Encapsulating these amylases, 25
therefore, can help control their enzymatic activity and reduce their damaging effect on 26
starch. Encapsulating amylases in lipid films has been suggested for their sustained release 27
during baking and shelf life of the baked product (Cole, 1983; Horn, 2002; Schuster et al. 28
2001; Mori et al., 2002). 29
To ensure even distribution of dough ingredients, manufacturers may sometimes extend 30
the time of mixing. In certain formulations, this overmixing can result in doughs beyond 31
their peak viscosity, thus adversely affecting their viscoelastic properties. Fuglsang et al. 32
(2002) developed a process for encapsulating xylanase enzymes (dough strengtheners) into 33
micelles to help reduce dough stickiness and improve its handling and machinability. 34
Release of the enzyme was designed to be initiated by melting the coating material during 35
leavening or early baking where enzyme activity is desired. Pan breads and flat breads are 36
the most common target applications. 37
38
39
Antimicrobial Agents
40
A wide range of antimicrobial agents has been used traditionally for controlling microbial 41
growth in shelf-stable bakery products such as sorbates, benzoates, propionates, paraben, 42
nisin, fumaric acid, and in some cases food grade metabolites produced by Propionibac- 43
terium sp. However, effective concentrations of these substances can dramatically affect the 44
flavor, color, odor, and textural attributes of bakery products. Another drawback of using 45
antimicrobial agents is their negative impact on viability of yeasts and enzymes used in S46
bakery systems. Acidic antimicrobial agents, sorbates in particular, can also disturb the N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 130

130 Chapter 5

1 chemical-leavening balance of doughs and batters. Their reducing power can lead to irre-
2 versible changes in the rheological properties of dough and batter systems.
3 An alternate approach for retarding microbial growth of doughs and batters is via pH
4 adjustment (reducing pH depending on type of bacteria and product storage conditions).
5 Although reducing pH can inhibit growth of bacteria and other microorganisms, it has no
6 effect on growth of fungus. Generally, a pH of 5 is acceptable for refrigerated doughs,
7 whereas ambient shelf-stable products require a lower pH (<4). The latter can have an unfa-
8 vorable reducing effect on the rheological properties of the dough or batter systems.
9 Natamycin is an extremely effective natural antifungal polyene macrolide that can be
10 produced by fermentation of the bacterium Streptomyces natalensis. However, its activity
11 can be negatively affected by extreme pH conditions and in the presence of metals. In addi-
12 tion, natamycin is very antagonistic to yeasts and moulds. Several encapsulation and con-
13 trolled release formulations have been documented in the patent literature for mitigating
14 these undesirable effects (Kringleum, 1999; Thomas et al., 2005) mainly via coating the
15 preservative with a food-grade high-melting hydrophobic substance and its further disper-
16 sion into bread dough system. Methods for encapsulating natamycin using a variety of
17 matrices via extrusion, liposomes, coacervation have been developed (Thomas et al., 2005)
18 and claimed sustained release of the antifungal agent into yeast-leavened dough with no
19 adverse effects on the yeast. Koontz and Marcy (2003) reported successful entrapment of
20 natamycin into a γ-cyclodextrin molecule and formation of stable natamycin/γ-cyclodextrin
21 inclusion complex, despite the incomplete lodging of the bulky natamycin in the
22 γ-cyclodextrin host.
23
24
Flavors
25
26 Encapsulation of flavors has been used in baking applications to retard flavor losses during
27 baking and/or eliminate their undesirable interactions with dough components. One group
28 of flavors including cinnamon, cloves, allspice, and nutmeg is known to have negative
29 effect on yeast-leavened doughs, resulting in deteriorating the yeast’s rising properties as
30 well as overall quality of the final baked product. The S-containing major components of
31 cinnamon oil can also have a negative impact on gluten development due to their interac-
32 tions with disulfide moieties of gluten. Black et al. (1988), developed an encapsulation
33 composition that is claimed to allow incorporation of cinnamon flavor to baked products
34 and to retard the associated undesirable interactions. Wampler (1995) patented a coacervation-
35 based flavor composition to deliver a high payload (70–95% flavor oil). via a cross-linked
36 gelatin shell. The composition was claimed to be stable during mixing with other dough
37 ingredients and subsequent baking.
38
39
Sweeteners
40
41 Conklin et al. (1987) developed a microencapsulated particulate sweetening system that is
42 thermally stable and is suitable for bakery applications. The composition is essentially a
43 water-swellable structure containing aspartame/acid core. Co-packing the food acid with
44 aspartame can create adequate pH (<5) conditions in the aspartame’s microenvironment,
45 thus stabilizing the dipeptide. The composition was claimed to be made very compact to
46S reduce wetting via capillary and can also be coated with a hydrophobic substance to delay
47N but not block the diffusion of water.
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 131

Encapsulation and Controlled Release in Bakery Applications 131

Further Remarks 1
2
• Generally speaking, encapsulation of leavening agents, especially chemical leaveners, 3
requires applications of higher levels of coating than most traditional applications (up to 4
95% coating may be required). 5
• Reproducibility of production-scale microcapsules is often an issue, especially for 6
moisture-labile actives such as sodium bicarbonate. Regardless of the technology or 7
materials used, leaky capsules may still be generated. Severity of this problem varies 8
with the end-product application. 9
• Manufacturers should be aware of potential differences in performance of encapsulated 10
ingredients in conventional baking compared to microwave baking. 11
• Judging the stability of an encapsulated leavening system should be done by monitoring 12
changes in CO2 generated as well as pH of the dough/cake batter. The latter can be most 13
accurately be determined few hours after preparing the dough or batter. 14
• Incorporating additional amounts of leavening acid into the formula (slightly higher 15
than needed during baking) may help make up for any initial neutralizing reaction that 16
might have occurred upon mixing the batter. 17
• For shelf-stable doughs or batters packaged in containers, injection of an inert gas such 18
as nitrous oxide that is partially soluble in the dough can help produce extra amounts of 19
gas bubbles to compensate for any build up of viscosity and density of batters. The latter 20
may result from reactions with the protein and starch components of the dough/batter 21
system. 22
• Chemical reactivity of leavening systems does not necessarily stop in dry mixes. Con- 23
densation reactions can be a problem even at very low moisture levels, which can accel- 24
erate other reactions. The role of water in low moisture systems can be more 25
challenging. 26
• The choice of a technology and/or material(s) for encapsulating actives for bakery appli- 27
cations depends on a host of factors such as type of finished product, desired packaging 28
and shelf life, release trigger, site, rate of release, cost and so on. For successful micro- 29
capsule design, it is imperative to determine whether the active is sensitive to moisture, 30
water vapor, oxygen, high temperatures, pH, or other environmental parameters. 31
32
33
References 34
Avner, R. and Blatt, Y. 1990. Microcapsules containing food additives and their use. EP 411,326 A2. 35
Azvedo, L.F. and Teixeira, A.M. 2002. A critical review of the modeling of wax deposition mechanisms. Presented 36
at the AIChE 2002 Spring National Meeting, New Orleans, LA, 10–14 March, 2002. 37
Bailey, A.E. 1952. Melting and Solidification of Fats. New York, Interscience Publishers. 38
Bennett, H. 1975. Industrial Waxes. Chemical Publ. Co., New York, NY.
39
Black, M., Popplewell, L.M. and Porzio, M. 1988. Controlled release encapsulation composition. US Patent
5,756,136. 40
Book, S., Corliss, G. and Heidolph, B. 2000. Process and formulation for a chemically leavened dough or bakery 41
product. US Patent 6,149,960. 42
21 CFR, Code of Federal Regulations(§.184.1973; § 184.1978 and §184.1976). 43
Chen, S. and Nussinovitch, A. 2000. Permeability & roughness determinations of wax-hydrocolloid coatings &
44
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. 45
Church and Dwight Co, Inc.1999. Leavening Sales brochure. S46
Cole, M. 1983. Antistaling baking composition. US Patent 4,416,903. N47
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 132

132 Chapter 5

1 Conklin, J.R., Gressgott, H.L. and Wolford, T.D. 1987. Thermally stable particulate artificial sweeteners.
2 EP 0,229,730 A2.
Das, S.K. and Gupta, B.K. 1988. Optimization of controlled drug release through micropellitization. Drug Dev.
3 Ind. Pharm. 14(12): 1673–1697.
4 Deasey, P.B. 1984. In Microencapsulation and Related Drug Processes (P.B. Deasy, Ed.), Marcel Dekker, New
5 York, p. 181.
6 Domingues, D.J. 2003. Chemical leavened doughs and related methods. WO 03/020,044 A1.
7 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-
8 lactam gel: a novel matrix for entrapment of microorganisms. Biotechnol. Tech. 7(6): 415-422.
9 Dorko, C.L. and Penfield, M.P. 1993. Melt point of encapsulated sodium bicarbonates: effect on refrigerated batter
10 and muffins baked in conventional and microwave ovens. J. Food Sci. 58(3):574–578.
11 El-Afandi, A. and Citti, J. 2006. Refrigerated dough and product in low pressure container. US Patent
12 2006/0,177,558 A1.
Eldem, P., Speiser, P. and Hincal, A.A. 1991. Optimization of spray-dried and –congealed lipid micropellets and
13 characterization of their surface morphology by scanning electron microscopy. Pharm. Res. 8(1): 47–54.
14 Fleischmann’s Yeasts. 2001. Product directory.
15 Freeman, A. and Dror, Y. 1994. immobilization of “disguised” yeast in chemically cross-linked chitosan beads.
16 Biotechnol and Bioeng. 44: 1083-1088.
17 Fuglsang, C., Callisen, T. and Budolfsen, G. 2002. Dough composition comprising a lipid-encapsulated enzyme.
WO 02/19,828 A1.
18 Gherbe-Sellassie, I. 1989. In Pharmaceutical Pelletization Technology (I. Gherbe-Sellassie, Ed.), Marcel Dekker,
19 New York, p. i.
20 Gontard, N., Duchez, C., Cuq, J.L. and Guilbert, S. 1994. Edible composite films of wheat gluten and lipids: water
21 vapor permeability and other physical properties. Int. J. Food Sci. Technol. 29: 39–50.
22 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
23 Chem. 41: 283–287.
24 Hernandez, E. 1994. Edible Coatings from Lipids & Resins. Chapter 10: 279- in Edible Coatings & Films to
25 improve Food Quality (J. Krochta, E.A. Baldwin and M.O. Nisperos-Carriedo, Eds.). Technomic Publishing
26 Co., Lancaster, Basel., pp. 279–303.
27 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,
28 A.M., Rogers, D.E. and Saguy, I. 1989. Starch-based products for microwave cooking or heating. EP 0,617,896 A2.
29 Ichikawa, H., Kaneko, S. and Fukumori, Y. 1996. Coating performance of aqueous composite lattices with
30 N-ispropylacrylamide shell and thermosensitive permeation properties of their microcapsule membrane. Chem.
31 Pharm. Bull. 44(2): 383–391.
32 John, P.M. and Becker, C.H. 1968. Surfactant effects on spray-congealed formulations of sulfaethylthiadiazole-
wax. J. Pharm. Sci. 57(4): 584–589.
33 Jozwiakowski, M.J., Jones, D.M. and Franz, R.M. 1990. Characterization of a hot-melt fluid bed coating process
34 for fine granules. Pharm. Res. 7(11): 1119–1126.
35 Kamper, S. and Fennema, O. 1984. Water vapor permeability of an edible fatty acid bilayer film. J. Food Sci. 49:
36 1482–1485.
37 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
38 stearyl alcohol film. JAOCS 66: 1154–1157.
39 Koontz, J.L. and Marcy, J.E. 2003. Formation of natamycin:cyclodextrin inclusion complexes and their character-
40 ization. J. Agric. Food Chem. 51: 7106–7110.
41 Kringelum, E. 1999. Compositions containing encapsulated food additives and their use. WO 99/08,553.
42 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.
43 Int. J. Pharm. Tech. and Prod. Mfr. 2(4): 31–43.
44 Litwinenko, J.W., Singh, A.P. and Marangoni, A.G. 2004. Effects of glycerol and Tween 60 on the crystallization
45 behavior, mechanical properties and microstructure of a plastic fat. Crystal Growth and Design 4(1): 161–168.
46S Luca, S.F., Thommel, J. and Bronn, W.K. (1979). Free-flowing powdered fresh baker’s yeast preparation and
47N method of producing it. US Patent 4,160,040.
Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 133

Encapsulation and Controlled Release in Bakery Applications 133

Madan et al. 1974. Microencapsulation of a waxy solid: wall thickness and surface appearance studies. J. of Phar- 1
maceutical Sci. 63(2): 280–284. 2
Markvicheva, E.A., Kuz’kina, Pashkin, I.I., plechko, T.N., Kirsh, Y.E. and Zubov, V.P. 1991. A novel technique for
entrapment of hybridome cells in synthetic thermally reversible polymers. Biotechnol. Tech. 5(3): 223-226.
3
McHugh, T.H. and Krochta, J.M. 1994. Milk protein-based edible films and coatings. Food Technol. 48(1): 4
97–103. 5
Mori, S., Sato, K. and Tanaka, N. 2002. Dough composition and preparation thereof. US Patent 6,355,282 B1. 6
Narine, S.S. and Marangoni, A.G. 1999. Relating structure of fat crystal networks to mechanical properties: a 7
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
8
Food and Nutrition Research 44: 33-145. 9
Pacifico, C.J. 2003. Chemical leavening ingredient. US Patent 2003/0,031,773 A1. 10
Pelletier, R. and Roger, F. 1989. Process for using bakery additives and bakery yeast. WO 89/00,009. 11
Percel, P.J. 1988. Encapsulated yeast. US Patent 4,719,114. 12
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.
13
Redding, B.K. 1995. Method for entrapment of liquids in transformed waxes. US Patent 5,460,756. 14
Redding, B.K. and Bruce, K. 2002. Ready-to-use food product. WO 02/11,544 A1. 15
Reed, G. and Nagodawithana, T.W. (Eds.). 1991. Bakers yeast production, pp. 261–314. In Yeast Technology, 2nd 16
edition. AVI Publishers, New York. 17
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.
18
Sato, K. and Kuroda, T. 1987. kinetics of melt crystallization and transformation of tripalmitin polymorphs. 19
J AOCS 64(1): 124–127. 20
Schuster, E., Sprossler, B. and Hofmeister, J. 2001. Process for making baked articles that retain freshness. US 21
Patent 6,254, 903 B1. 22
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.
23
Shimon, L.M., Kotorman, M. and Sayani, B. 1991. immobilization of yeast alcohol dehdrogenase on a p-benzo- 24
quinone-actiavted silicate carrier. Prikladnaya Biokhimiya i Mikrobiologiya 27 (1): 86-90. 25
Soltis, J. and Sell, J.L. 1989. Yeast composition. US Patent 5,70,669. 26
Sward, G.G. 1972. Natural resins. Am. Soc. Test Mat: 77-91. 27
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.
28
Tuazon, M.T. and Foster, L.C. 1992. Souffle mix. EP 0,545,025 B1. 29
Tulloch, A.P. 1970. The composition of beeswax and other waxes secreted by insects. Lipids 5(2): 247-258. 30
Ubbelohde, A.R. 1978. In The molten State of Matter, John Wiley & Sons, Chichester. 31
Vaghefi, F., Lee, J. and Nalamothu, V. 2001. Zero-order release and temperature-controlled microcapsules and 32
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
33
Sci. 16: 284–296. 34
Vreeker, A., Hoekstra, L.L., deb Boer, D.C. and Agterof, W.G.M. 1992. The fractal nature of fat crystal networks. 35
Colloids and Surfaces 65: 185–189. 36
Wampler, D. 1993. Aqueous liquid flavor oil capsules, method of making and using in foods. EP 0,633,732 B2. 37
Wetzel, C.R. and Bell, L.N. 1998. Chemical stability of encapsulated aspartame in cakes without added sugar.
Food Chem. 63(1): 33-37.
38
Wineman, R.D. 1984. Water emulsion fruit and vegetable coatings based on waxes. Eastman Chem. Prod. Publi- 39
cation # F-257A. 40
Wu, C., Creek, J.L., Wang, K., Carlson, R.M., Cheung, S. and Tang, P.J. 2002. Measurement of wax deposition in 41
paraffin solutions. Presented at the AIChE 2002 Spring National Meeting, New Orleans, LA 10–14 March. 42
43
44
45
S46
N47
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 135

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

6 Encapsulation Technologies for 1


2
Preserving and Controlling the Release 3
of Enzymes and Phytochemicals 4
5
6
7
Xiaoyong Wang, Yan Jiang, and 8
Qingrong Huang 9
10
11
12
13
14
Introduction
15
According to a report from Business Communication, Inc. (http://www.bccresearch.com), 16
the functional food industry in the US was valued at $20.2 billion in 2002 or 4 percent 17
of the total food industry. Driven by both increasing fortification with healthy food in- 18
gredients and consumer demand for novel food products, the functional food market is 19
expected to increase at an average growth rate of 13.3 percent, bringing the market value 20
to $37.7 billion by 2007. The development of functional foods with good bioavailability 21
and eating qualities, however, requires methods for protecting these sensitive compo- 22
nents from harmful environmental conditions and masking the taste of some of these 23
components. 24
Encapsulation is the technique by which one material or a mixture of materials is coated 25
with or entrapped within another material or system (Green and Scheicher, 1955). Encap- 26
sulation can also be used to mask undesirable odors and bitter tastes of food ingredi- 27
ents. The coated material is called active or core material, and the coating material is called 28
shell, wall material, carrier, or encapsulant. Encapsulation technology is well devel- 29
oped and accepted within the pharmaceutical, chemical, cosmetic, and food industries 30
(Augustin et al., 2001; Heinzen, 2002). Many encapsulation techniques have been devel- 31
oped, such as spray drying, spray chilling and cooling, coacervation, fluidized bed coating, 32
liposome entrapment, rotational suspension separation, and extrusion and inclusion 33
complexation (Madene et al., 2006). 34
The widely used wall materials include polysaccharides and proteins, the key 35
components in both natural and processed foods (Tolstoguzov, 1991). Such polymers 36
have critical impact on the structure and stability of food systems through their gelling, 37
thickening, and surface-stabilizing functional properties. Proteins and polysaccha- 38
rides are usually used in composites, especially when the creation of new products 39
is required. Intrinsic functional properties of individual components and their interactions 40
determine the final structure, texture, and stability of food materials. Understanding such 41
interactions, especially between proteins and polysaccharides, is important not only 42
for manufacturing cost-effective functional ingredients, but also for designing novel 43
foods and for controlling their structural and textural impact on fabricated foods (Sanchez 44
et al., 1997). 45
S46
N47

135
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 136

136 Chapter 6

1 Complex Coacervate-Based Controlled Release Systems


2
3 One way to create controlled release encapsulation systems is through the use of complex
4 coacervates formed by proteins and polysaccharides. The basic science behind the
5 coacervation process is well developed and understood. Coacervation is divided into
6 “simple” or “complex” processes. The former involves only one macromolecule and may
7 result from the addition of a dehydrating agent that promotes polymer–polymer interac-
8 tions over polymer–solvent interactions. In the latter case, two or more oppositely charged
9 macromolecules or colloidal species are present to generate phase separation. In solution,
10 polysaccharide and proteins may undergo two types of phase separation at above or below
11 the isoelectric points of proteins: (i) the solid–liquid phase separation called precipitation
12 (Kokufuta et al., 1981); and (ii) the liquid–liquid phase separation called coacervation
13 (Burgess and Carless, 1984). The coacervate is the denser phase that is relatively concen-
14 trated in macromolecules and is in equilibrium with the relatively dilute macromolecular
15 liquid phase (Bungenberg, 1949).
16 The general picture for protein/polysaccharide coacervation from previous studies is that
17 protein molecules initially bind to polysaccharide chains to form primary soluble complexes
18 at first critical pH (pHc), and complex coacervate droplets, which ultimately settle at the bot-
19 tom to generate the dense coacervate phase, are formed at second critical pH (pH). Primary
20 complex formation, initiated at pHc, is viewed as a microscopic transition on the molecular
21 scales, whereas coacervate droplet formation at pH is viewed as a global phase transition.
22 A typical phase diagram of bovine serum albumin (BSA)/-carrageenan mixtures is
23 shown in Figure 6.1. Three regions are observed in the pH titration curve: (i) at pH > 6.2,
24 there is no change in turbidity; (ii) at pH < 6.2, turbidity starts to increase, which is identified
25 as the intercept of the soluble complex (pHc); and (iii) at pH < 4.8, turbidity increases signifi-
26 cantly with the decrease of pH, which corresponds to the phase separation point (pH).
27 Because coacervates formed by polysaccharides and oppositely charged proteins are mainly
28 driven by the long-range character of the electrostatic interaction, physicochemical parame-
29 ters affecting such interactions, such as pH, ionic strength, polysaccharide linear charge den-
30 sity, protein surface charge density, rigidity of the polysaccharide chain, size of the protein,
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S Figure 6.1. Plot of turbidity versus pH for mixture of bovine serum albumin (BSA) and
47N -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 137

Preserving and Controlling the Release of Enzymes and Phytochemicals 137

and protein/polysaccharide ratio, strongly influence the formation of the complexes (Burgess 1
and Singh, 1993; Hansen et al., 1971; Hugerth and Sundelof, 2001; Xia and Dubin, 1994). 2
Although extensive studies have been focused on the phase boundaries of protein/ 3
polysaccharide coacervation, understanding the structure of protein/polysaccharide coacer- 4
vates is still quite lacking (Doublier et al., 2000; Turgeon et al., 2003). With the help of confo- 5
cal scanning laser microscopy, Sanchez et al. (2002) found that the internal structure of 6
-lactoglobulin/gum arabic coacervates was vesicular or sponge-like, exhibiting numerous 7
spherical inclusions of water depending on the initial mixing ratio. Using small-angle X-ray 8
scattering, Weinbreck et al. (2004a) found that whey protein/gum arabic complex coacervates 9
were dense and structured and could be tuned by pH, protein/polysaccharide ratio, and ionic 10
strength. They also studied the viscoelastic properties of whey protein/gum acacia coacer- 11
vates and verified that whey protein/gum acacia complex coacervates had the highest viscos- 12
ity at pH = 4.0, which was ascribed to the strongest electrostatic interaction between whey 13
protein and gum acacia (Weinbreck et al., 2004b). Recently, our group has investigated the 14
dynamic rheolgical properties of BSA/-carrageenan complex coacervates. Figure 6.2 shows 15
typical profile of small deformation oscillatory measurements of BSA/-carrageenan com- 16
plex coacervates at 0.1 M NaCl concentration and 10:1 protein/polysaccharide ratio, with 17
pH = 4.5. The storage modulus (G) was found to be more than two times greater than the loss 18
modulus (G), wherein the two moduli are almost independent of angular frequency () at 19
 > 0.5 rad/s. The high value of G when compared to G indicates that BSA/-carrageenan 20
complex coacervates have a highly interconnected gel-like network structure with mainly 21
elastic behavior, which agrees with the rheological properties of simple coacervates like gela- 22
tin (Mohanty and Bohidar, 2005). At similar frequencies, sweep measurements for whey 23
protein/gum Arabic coacervates, G was reported to be three to seven times higher than G, an 24
indication of the highly viscous character of whey protein/gum Arabic coacervates (Weinbreck 25
et al., 2004a). Coacervates of BSA with synthetic polyelectrolyte poly(diallyldimethylammo- 26
nium chloride) also show viscous nature, with G larger than G in the high-frequency range 27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
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 (Lee S46
et al., 2003). N47
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 138

138 Chapter 6

1 (Bohidar et al., 2005). Therefore, different viscoelastic properties in different systems reflect
2 the characteristics of protein/polymer pair and thus distinct coacervate structure.
3 Small-angle neutron scattering (SANS) experiments, which were performed at the
4 intense pulsed neutron source at Argonne National Laboratory, Argonne, IL, have been used
5 to illustrate the structure of complex coacervates formed by -lactoglobulin and pectin. The
6 SANS results for -lactoglobulin/pectin coacervates (30:1 w/w) at two different salt concen-
7 trations are shown in Figure 6.3(a). All the curves show a peak of shoulder at intermediate
8 scattering vector range, indicating the electrostatic repulsion of proteins bound onto pectin
9 chains. From the maximum of the peaks in the structure factor curves [Figure 6.3(b)], the
10 distance between bound proteins (d) could be determined: d = 7.6 nm at 0.05 M NaCl is
11 found to be smaller than d = 8.7 nm at 0.1 M NaCl. The strong correlation between peak
12 maxima and their position with salt concentration may be an indication of the more het-
13 erogenous and less-structured nature of these coacervates at higher salt concentration.
14 The concept behind protein/polysaccharide complex coacervate-based controlled release
15 delivery systems arises from the pH-triggered phase separation of protein/polysaccharide
16 complexes from the initial mixed solutions, and the subsequent deposition of the newly
17 formed coacervate phase surrounding the active ingredients (Gouin, 2004). If needed, the
18 coacervate shell can be cross-linked using an appropriate chemical or enzymatic cross-
19 linker. A large number of protein/polysaccharide complex systems, such as gelatin/gum aca-
20 cia (Ijichi et al., 1997; Rabiskova and Valaskova, 1998), gelatin/carboxymethylcellulose
21 (Bakker et al., 1999), -lactoglobulin/gum acacia (Schmitt et al., 2000), and guar/dextran
22 (Simonet et al., 2002), have shown good properties for microencapsulation application.
23 Coacervation is typically used in the encapsulation of flavor oils (Soper, 1995), but can
24 also be adapted for the encapsulation of fish oils (Lamprecht et al., 2001), vitamins
25 (Junyaprasert et al., 2001), enzymes (Dubin et al., 1998), and dietary supplements.
26 To improve the appeal of frozen baked foods upon heating, flavor oil was entrapped in
27 complex coacervate microcapsules using gelatin and gum arabic (Yeo et al., 2005). The
28 design criteria of these systems include: (i) the odor should not be released while the food is
29 frozen or, if thawed, before it is cooked; and (ii) the odor should be released upon heating.
30 The rate of homogenization was found to affect the size of oil cores encapsulated within the
31 microcapsules, whereas the polyion concentrations affected the number of core aggregates
32 consisting of a microcapsule. The comparison of homogenization speed between 3000 and
33
34
(a) (b)
35 0.05 M NaCl 0.05 M NaCl
36 100 0.1 M NaCl 100 0.1 M NaCl

37
S (q)
I (q)

38 10
39 10
40 1
41
42
0.01 0.1 1 0.01 0.1 1
43
q (A–1) q (A–1)
44
45 Figure 6.3. Small-angle neutron scattering intensity profiles from -lactoglobulin/pectin
46S coacervates (30:1 w/w) at different salt concentrations: (a) scattering curves; (b) structure
47N factor curves.
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 139

Preserving and Controlling the Release of Enzymes and Phytochemicals 139

9000 rpm shows that the microcapsules prepared with a lower homogenization speed were 1
less resistant to heating. A possible explanation for this result is that slower homogenization 2
formed microcapsules having single large cores, which may make them more vulnerable to 3
damage than multivesicular microcapsules produced by higher speed homogenization. 4
Microcapsules prepared with higher concentrations of polyions were less resistant to the 5
release conditions. This result may be due to the fact that lower solution concentrations 6
resulted in bigger agglomerates of oil droplets, which were harder to completely break down. 7
8
9
Encapsulation and Controlled Release of Food Enzymes
10
Enzymes are catalytic proteins that are capable of great specificity and reactivity under 11
physiological conditions. Like most proteins, they are highly susceptible to physiological 12
parameters such as pH and heat and to chemicals like denaturation agents. They are com- 13
monly encapsulated and immobilized in food processing, biomedical examination, and 14
antibody labeling. However, they are normally contaminated by proteases, which may gen- 15
erate unpredictable or inaccurate results. Most often, these enzymes are obtained from bac- 16
teria or other biomaterials, which subsequently complicates their purification and the 17
formation of protease-free enzymes. The existence of protease in enzymes prevents the use 18
of proteins as wall materials for enzyme encapsulation. 19
Recently, we have developed an inexpensive, fast, and convenient method for encapsulat- 20
ing food enzyme (Jiang and Huang, 2004) through the direct formation of complex coacer- 21
vate with negatively charged polysaccharide such as -carrageenan. -Amylase was used as 22
a model enzyme to form coacervates with -carrageenan to microencapsulate -amylase. 23
The -amylase encapsulation efficiency and free -amylase were defined as: 24
25
Encapsulated enzyme 26
Encapsulation efficiency %  (1)
27
Total enzyme
28
29
Non-encapsulated enzyme
Free enzyme %  (2) 30
Total enzyme 31
32
Our results show that a -carrageenan to -amylase ratio of 1 to 2 resulted in very high 33
encapsulation efficiency (>99 percent) of -amylase as shown in Figure 6.4. -Amylase 34
released from coacervates also maintained the same catalytic activity as the enzyme control, 35
while unencapsulated -amylase lost most of its enzymatic activities after exposure to low pH 36
(i.e., 3) for half an hour. Enzyme kinetics, therefore, can be described by Michaelis–Menten 37
equation, 38
K 39
1 1 1
 m
(3) 40
V Vmax [ S ] Vmax 41
42
Here Km is the Michaelis–Menten constant and Vmax is the maximum hydrolysis rate. Km and 43
Vmax were determined from equation (3). Table 6.1 shows that for coacervate-encapsulated 44
-amylase, even after being treated with acid, -amylase displayed negligible change in enzy- 45
matic activities after being released. However, -amylase without coacervate protection S46
almost lost its enzymatic activities, as evidenced by significantly lower values of Km and Vmax. N47
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 140

140 Chapter 6

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15 Figure 6.4. Encapsulation efficiency curve. Encapsulation has the highest efficiency of about
16 99.3 percent at the ratio of -carrageenan/ -amylase 1:2 in 0.01 M NaCl.
17
18
19
Table 6.1. Enzymatic kinetics of -amylase with different treatments
20
21 Enzyme Km (g) Vmax (g/min)
22 1 Untreated enzyme (control) 1.08 0.3
23 2 Encapsulated, acid treated, released 1.07 0.28
24 3 Unencapsulated, acid treated 0.04 0.0036
25
26
27
28 These results suggest that the enzyme encapsulation through complex coacervation is an effi-
29 cient method to protect the enzyme from denaturation.
30
31
Encapsulation and Controlled Release of Phytochemicals
32
33 Phytochemicals have received much attention in recent years from the scientific
34 community, consumers, and food manufacturers due to their potential in lowering blood
35 pressure, reducing cancer risk factors, regulating digestive tract activity, strengthening
36 immune systems, regulating growth, controlling blood sugar concentration, lowering
37 cholesterol levels and serving as antioxidants. The scientific evidence supporting these
38 health-promoting claims of phytochemicals is growing steadily (Wildman, 2001).
39 Although the use of phytochemicals in capsules and tablets is abundant, their effect is
40 frequently diminished or even lost due to their lack of solubility in water, vegetable oils or
41 other food-grade solvents. In addition, insufficient gastric residence time, low permeability
42 and solubility within the gut, as well as instability under conditions encountered in product
43 processing (temperature, oxygen, light) or in the gastro-intestinal tract (pH, enzymes,
44 presence of other nutrients) limit the activity and potential health benefits of phytochemical
45 molecules (Bell, 2001). The delivery of these molecules will therefore require availability
46S of protective mechanisms that can maintain the active molecular form until the time of
47N consumption and to deliver this form to the physiological target within the organism.
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 141

Preserving and Controlling the Release of Enzymes and Phytochemicals 141

1
2
3
4
5
6
7
8
9
10
11
12
13
Figure 6.5. Color changes of epigallocatechin gallate (EGCG) at different pHs after 1-day 14
storage. 15
16
17
To overcome instability, poor water solubility and bioavailability of phytochemicals, 18
encapsulation techniques have been employed to bring about effective amounts of the intact 19
active component to desired target sites in the body. Ideally, actives such as phytochemicals 20
should be stable and intact under stomach acidic conditions, but readily bioavailable under 21
prevailing alkaline conditions of the small intestines (Ho et al., 1992; Salah et al., 1995; 22
Havsteen, 1983). 23
Tea catechins, one of the typical flavonoid components of green tea, have been shown 24
to possess desirable physiological activities such as antioxidants, anti-AIDS virus, anti- 25
mutagenic, anti-carcinogenic, probiotic, anti-microbial and anti-inflammatory (Havsteen, 26
1983; Nakagawa et al., 1999). One of the major challenges with utilizing tea catechins 27
is their poor oral bioavailabilities. Epigallocatechin gallate (EGCG), the most important 28
component of catechins contained in green tea, can readily undergo extensive glucoronida- 29
tion, sulfation, methylation and ring fission in humans, mice and rats (Yang et al., 2002; 30
Nakagawa et al., 1997; Suganuma et al., 1998; Cauturla et al., 2003). In addition, it can eas- 31
ily undergo oxidation at neutral to alkaline pH, especially at high temperatures. Figure 6.5 32
demonstrates progressive increase in color intensity (browning) of EGCG solutions with 33
increased pH after only one-day storage. Oxidation of EGCG solutions at different pH 34
levels and temperatures can be accurately monitored by tracing their absorption at wave- 35
length of 290 nm using UV spectroscopy. Upon oxidation of EGCG, its absorption wave- 36
length was found to gradually shift to longer wavelength (317 nm). 37
In our laboratories, we attempted to preserve the stability and bioavailability of tea 38
catechins (EGCG) via complex coacervation in carrageenan/gelatin-A (Jiang and Huang, 39
2004). The encapsulation efficiency of EGCG in these coacervates was determined 40
by high performance liquid chromatography (HPLC) and found to be as high as 89.4% 41
(Figure 6.6). In vitro release of coacervate encapsulated EGCG was also studied in artificial 42
stomach and intestinal juices at 37ºC for 2 hrs and 4 hrs, respectively. The active (EGCG) 43
did not show any release under acidic stomach conditions (confirmed by UV spectra), 44
but was totally released in the first 15 minutes of incubation in artificial intestinal juice 45
(Figure 6.7). S46
N47
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 142

142 Chapter 6

1 12
2
3 10
4
5 8
6
mg/ml

7 6 Efficiency
89.44%
8
9 4
10
11 2
12
13 0
Encapsulated
14 Un-encap
+Un-encap
15
16 Figure 6.6. Encapsulation efficiency of complex coacervate-encapsulated epigallocatechin
gallate (EGCG) as determined by high-performance liquid chromatography (HPLC).
17
18
19
20
21
22 No catechins
23 Stomach were released
24 in 2 hrs.
25
26
27
28
29
30
31 Small intestine Within 20 min.,
32 all catechins
33 were released.
34
35
36 Figure 6.7. In vitro release of tea catechins in artificial stomach and intestinal juice.
37
38
39
Encapsulation of Phytochemicals by Nanoemulsions
40
41 Nanoemulsions are a class of extremely small emulsion droplets that can be transparent or
42 translucent with a bluish coloration (Nakajima, 1997; Solans et al., 2005; Sonneville-
43 Aubrun et al., 2004). They are usually available in the range of 50-200 nm. Similar to tradi-
44 tional macro-emulsions, two types of nanoemulsions can be prepared, namely oil-in-water
45 (O/W) and water-in-oil (W/O) nanoemulsions. Although emulsions are thermodynamically
46S unstable systems, nanoemulsions, owing to their characteristic size, may possess high
47N kinetic stability against sedimentation or creaming. Nanoemulsions can be prepared by the
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 143

Preserving and Controlling the Release of Enzymes and Phytochemicals 143

so-called dispersion or high-energy emulsification methods using high shear stirring, high- 1
pressure homogenization and ultrasound generators (Walstra, 1983). Other methods such 2
as condensation or low-energy emulsification and phase inversion temperature could pro- 3
duce nanoemulsion almost spontaneously (Rang & Miller, 1999). 4
Nanoemulsions have been investigated for their ability to transport phytochemicals 5
(Solans et al., 2005). The mechanism takes place via large reduction in gravitational force 6
and Brownian diffusion, thus preventing any creaming or sedimentation, followed by steric 7
stabilization and prevention of droplet flocculation or its coalescence. Nanoemulsions also 8
offer other advantages for encapsulating water-soluble (entrapped in the core) and water 9
insoluble (incorporated at the interface or the oil phase) substances that can be designed for 10
slow release applications (Garti et al., 2003; Shefer and Shefer, 2003). This approach was 11
claimed to enhance bioavailability of oil-soluble or water-soluble phytochemicals. 12
Curcumin, an FDA-approved food additive, is widely used as a preservative and yellow 13
coloring agent for foods, drugs, and cosmetics. Curcumin has also been shown to possess 14
unique anti-inflammatory activity (Reddy et al., 2004; Huang et al., 1988, 1994). However, 15
orally administered curcumin is plagued with low systemic bioavailability (Pan et al., 16
1999). Recently, we developed o/w nanoemulsion for encapsulating curcumin (Wang and 17
others, unpublished). Figure 6.8 shows photomicrographs of curcumin regular- and nano- 18
sized- emulsions with the latter exhibiting unique homogeneous droplet size distribution. 19
Using particle size analysis, average diameter of curcumin nanoemulsion droplets was 20
found to be 65 nm. 21
The mouse ear inflammation model is commonly used to test the bioavailability 22
of anti-inflammatory agents in vivo. In such studies, topical application of 12-O-tetrade- 23
canoylphorbol-13-acetate (TPA) can rapidly induce edema of mouse ear in a dose- and 24
time-dependent manner. Earlier studies in our laboratory have shown that oral administra- 25
tion of anti-inflammatory agents such as aspirin and garcinol can inhibit TPA-induced 26
edema in mouse ears. We have also reported that various levels of garcinol were found in 27
serum, ear, liver, lung and colon after oral administration of garcinol by female CD-1 mice 28
for several hours. In addition, oral administration of aspirin or garcinol by gavages resulted 29
in marked inhibition of TPA-induced edema in mouse ears. 30
In contrast, oral administration of curcumin, a poor bio-available anti-inflammatory 31
agent, had little or no effect on TPA-induced edema of mouse ears. However, oral adminis- 32
tration of two different preparations of curcumin emulsion (10 mg curcumin in 1 ml) 33
prepared by either high speed homogenization (regular) or high pressure homogenization 34
(nanoemulsion) to mice by gavages at 30 min prior to topical application of TPA has 35
markedly inhibited TPA-induced edema of mouse ears by 43 and 85%, respectively. 36
37
38
Bioconjugation of Phytochemicals
39
Nanoparticles are defined as submicronic (<1 μm) colloidal systems made of polymers 40
both biodegradable and non-biodegradable. Nanocapsules, one type of nanoparticles, are 41
vesicular systems in which actives such as flavonoids can be confined to a cavity, generally, 42
an oily or aqueous core surrounded by a unique polymeric membrane. Nanospheres, on the 43
other hand, are matrix systems in which the active is dispersed throughout the particles. 44
Initial research on colloidal carriers was mainly focused on liposomes which are very diffi- 45
cult to produce or stabilize for practical applications. In contrast, nanopraticles owing to S46
their unique stability can potentially be superior carriers compared to liposome. N47
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 144

144 Chapter 6

1
2
3
4
5
6
7
8
9
10
11
12
(a) (b)
13
14
Figure 6.8. Microscope images of curcumin normal emulsions (left) and nanoemulsions (right).
15
16
17 Activities of polyphenols such as their anti-oxidative power, circulation time in the
18 human body and other activities can be reduced upon exposure to environmental stresses
19 such as moisture, heat, and oxidation (Hagerman et al., 1998; Kurisawa et al., 2003). We
20 have attempted to preserve polyphenol activities, in particular their anti-oxidative ability by
21 means of synthesizing poly(catechin)via enzyme-catalyzed oxidative coupling using horse-
22 radish peroxidase as a catalyst (Shin and Huang, unpublished results). The poly-catechin
23 showed great improvement in antioxidative activity such as radical scavenging activity
24 against the superoxide anion and inhibition effects against free radical induced oxidation of
25 low-density lipoprotein, compared to the catechin monomer. In addition, poly-(catechin)
26 showed very high inhibition effects on xanthine oxidase activity, whereas the catechin
27 monomer showed very low inhibition effects.
28
29 Conclusion
30
31 One of the most important stakes in the health promotion industry is the efficient encapsu-
32 lation of highly valuable phytochemicals. Taking advantage of nanoscale particles,
33 nanoemulsions and nanoparticles, provide excellent vehicles for encapsulating phytochem-
34 icals and to preserve their stability and bioavailability. Another unique encapsulation tech-
35 nique for such applications is complex coacervation. Numerous protein/polysaccharide
36 pairs have been demonstrated to provide controlled-release of phytochemicals in vitro as
37 well as in vivo. Conjugation of phytochemicals can also play a promising role in encapsu-
38 lating large-scale actives and in their effective utilization in food systems. Indeed, the
39 choice of an appropriate technique, however, depends on the properties of the active com-
40 pounds, the degree of stability required during storage and processing, desired release
41 properties, maximum obtainable phytochemical load as well as production cost.
42
43 References
44
Augustin, M. A., Sanguansri, L., Margetts, C., Young, B. 2001. Microencapsulation of food ingredients. Food
45 Aust., 53, 220–223.
46S Bakker, M. A. E., Galema, S. A., Visser, A. 1999. Microcapsules of Gelatin and Carboxymethylcellulose. Euro-
47N pean Patent Application EP 0 937 496 A2, Unilever NV, NL; Unilever PLC, GB.
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 145

Preserving and Controlling the Release of Enzymes and Phytochemicals 145

Bell, L. N. 2001. Stability testing of nutraceuticals and functional foods. In: Wildman, R. E. C. (Ed.), Handbook of 1
Nutraceuticals and Functional Foods (pp. 501–516). New York: CRC Press. 2
Bohidar, H., Dubin, P. L., Majhi, P. R., Tribet, C., Jaeger, W. 2005. Effects of protein–polyelectrolyte affinity and
polyelectrolyte molecular weight on dynamic properties of bovine serum albumin-poly (diallyldimethylammo-
3
nium chloride) coacervates. Biomacromolecules, 6, 1573–1585. 4
Bungenberg de Jong, H. G. 1949. In: Kruyt, H. R. (Ed.), Colloid Science, Vol. II. Amsterdam: Elsevier. 5
Burgess, D. J., Carless, J. E. 1984. Microelectrophoretic studies of gelatin and acacia for the prediction of complex 6
coacervation. J. Colloid Interface Sci., 98, 1–8. 7
Burgess, D. J., Singh, O. N. 1993. Spontaneous formation of small sized albumin/acacia coacervate particles.
J. Pharm. Pharmacol., 45, 586–591.
8
Cauturla, N., Vera-samper, E., Villalain, J., Mateo, C. R., Micol, V. 2003. The relationship between the antioxidant 9
and the antibacterial properties of galloylated catechins and the structure of phospholipid model membranes. 10
Free Radic. Biol. Med., 34, 648–662. 11
Cho, Y. H., Huang, Q. R. (submitted for publication) Temperature Triggered Release of Curcumin from Complex 12
Coacervation with Gelatin A and Alginate.
Doublier, J.-L., Garnier, C., Renard, D., Sanchez, C. 2000. Protein–polysaccharide interactions. Curr. Opin.
13
Colloid Interface Sci., 5, 202–214. 14
Dubin, P. L., Muhoberac, B. B., Xia, J. 1998. Preparation of Enzyme–Polyelectrolyte Coacervate Complexes and 15
their Properties. US 5834271A. 16
Garti, N., Aserin, A., Spernath, A., Amar, I. 2003. Nano-Sized Self-Assembled Structured Liquids. 17
US20030232095.
Gouin, S. 2004. Microencapsulation: industrial appraisal of existing technologies and trends. Trends Food Sci.
18
Technol., 15, 330–347. 19
Green, B. K., Scheicher, L. 1955. Pressure Sensitive Record Materials. US Patent No. 2, 217, 507, NCR C. 20
Hagerman, A. E., Riedl, K. M., Jones, G. A., Sovik, K. N., Ritchard, N. T., Hartzfeld, P. W., Riechel, T. L. 1998. 21
High molecular weight plant polyphenolics (Tannins) as biological antioxidants. J. Agric. Food Chem., 46(5), 22
1887–1892.
Hansen, P. M. T., Hidalgo, J., Gould, I. 1971. Reclamation of whey protein with carboxymethylcellulose. J. Dairy
23
Sci., 54, 830–834. 24
Havsteen, B. 1983. Flavonoids, a class of natural products of high pharmacological potency. Biochem. Pharmacol., 25
32, 1141–1148. 26
Heinzen, C. 2002. Microencapsulation solves time dependent problems for foodmakers. Eur. Food Drink Rev. 3, 27
27–30.
Ho, C. T., Lee, C. Y., Huang, M. T. (Eds.). 1992. Phenolic Compounds in Food and their Effects on Health.
28
I: Analysis, Occurrence, and Chemistry. ACS Symp. Ser. 506, Washington, D.C.: American Chemical Society. 29
Huang, M. T., Lou, Y. R., Ma, W., Newmark, H. L., Reuhl, K. R., Conney, A. H. 1994. Inhibitory effects of dietary 30
curcumin on forestomach, duodenal and colon carcinogenesis in mice. Cancer Res., 54, 5841–5847. 31
Huang, M. T., Smart, R. C., Wong, C. Q., Cooney, A. H. 1988. Inhibitory effect of curcumin, chlorogenic acid, caf- 32
feic, and ferulic acid on tumor promotion in mouse skin by 12-O-tetradecanoylphorbol-13-aceate, Cancer Res.,
48, 5941.
33
Hugerth, A., Sundelof, L.-O. 2001. The effect of polyelectrolyte counterion specificity, charge density, and confor- 34
mation on polyelectrolyte–amphiphile interaction: The carrageenan/furcellaran-amitriptyline system. Biopoly- 35
mers, 58, 186–194. 36
Ijichi, K., Yoshizawa, H., Uemura, Y., Hatate, Y., Kawano, Y. 1997. Multi-layered gelatin/acacia microcapsules by 37
complex 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-
38
tein/polysaccharide coacervates. 39
Jiang, Y., Huang, Q. R. 2004. Microencapsulation and controlled-release of food enzyme using protein–- 40
polysaccharide coacervates. Polym. Prepr., 45(2), 464. 41
Junyaprasert, V. B., Mitrevej, A., Sinchaipanid, N., Boonme, P., Wurster, D. E. 2001. Effect of process variables on the 42
microencapsulation 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-
43
ride) with acidic groups in the polyion complex between human carboxyhemoglobin and potassium poly(vinyl 44
alcohol) sulfate. Macromolecules, 14, 1178–1180. 45
Kurisawa, M., Chung, J. E., Kim, Y. J., Uyama, H., Kobayashi, S. 2003. Amplification of antioxidant activity and S46
xanthine oxidase inhibition of catechin by enzymatic polymerization. Biomacromolecules, 4(3), 469–471. N47
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 146

146 Chapter 6

1 Lamprecht, A., Schafer, U., Lehr, C. M. 2001. Influences of process parameters on preparation of microparticle
2 used as a carrier system for W-3 unsaturated fatty acid ethyl esters used in supplementary nutrition. J. Microen-
capsul., 18, 347–357.
3 Lee, J. Y., Ruengruglikit, C., Huang, Q. R. 2003. Interactions between Carrageenan and BSA: (1) Effects of linear
4 charge density and ionic strength. Polym Prepr., 44, 289–290.
5 Madene, A., Jacquot, M., Scher, J., Desobry, S. 2006. Flavour encapsulation and controlled release. Int. J. Food
6 Sci. Technol., 41, 1–21.
7 Mohanty, B., Bohidar, H. B. 2005. Microscopic structure of gelatin coacervates. Int. J. Biol. Macromol., 36,
39–46.
8 Nakagawa, K., Miyazawa, T. 1997. Absorption and distribution of tea catechin, (-)-epigallocatechin-3-gallate, in
9 the rat. Nutr. Sci. Vitaminol., 43, 679–684.
10 Nakagawa, K., Ninomiya, M., Okubo, T., Aoi, N., Juneja, L. R., Kim, M., Yamanaka, K., Miyazawa, T. 1999. Tea
11 catechin supplementation increases antioxidant capacity and prevents phospholipid hydroperoxidation in
12 plasma of human. J. Agric. Food Chem., 47, 3967–3973.
Nakajima, N. 1997. Microemulsions in cosmetics. In: Solans, C., Kunieda, H. (Eds.), Industrial Applications of
13 Microemulsions (pp. 175–197). New York: Marcel Dekker.
14 Pan, M. H., Huang, T. M., Lin, J. K. 1999. Biotransformation of curcumin through reduction and glucuronidation
15 in mice. Drug Metab. Dispos., 27, 486–494.
16 Rabiskova, M., Valaskova, J. 1998. The influence of HLB on the encapsulation of oils by complex coacervation.
17 J. Microencapsul., 15, 747–751.
Rang, M. J., Miller, C. A. 1999. Spontaneous emulsification of oils containing hydrocarbon, nonionic surfactant,
18 and oleyl alcohol. J. Colloid Interface Sci., 209, 179–192.
19 Reddy, R. S., Rao, C. V. 2004. Chemoprevention of colon cancer by curcumin. In: Meskin, M. S., Bidlack, W. R.,
20 Davies, A. J., Lewis, D. S., Randolph, R. K. (Eds.), Phytochemicals: Mechanisms of Action (pp. 177–192).
21 Boca Raton, FL: CRC Press.
22 Salah, N., Miller, N. J., Paganga, G. 1995. Polyphenolic flavanols as scavengers of aqueous phase radicals and as
chain breaking antioxidants. Arch. Biochem. Biophys., 322, 339–346.
23 Sanchez, C., Schmitt, C., Babak, V. G., Hardy, J. 1997. Rheology of whey protein isolate-xanthan mixed solutions
24 and gels. Effect of pH and xanthan concentration. Nahrung, 41(6), 336–343.
25 Sanchez, C., Mekhloufi, G., Schmitt, C., Renard, D., Robert, P., Lehr, C.-M., Lamprecht, A., Hardy, J. 2002. Self-
26 assembly of -lactoglobulin and acacia gum in aqueous solvent: structure and phase-ordering kinetics. Lang-
27 muir, 18, 10323–10333.
Schmitt, C., Sanchez, C., Despond, S., Renard, D., Thomas, F., Hardy, J. 2000. Effect of protein aggregates on the
28 complex coacervation between beta-lactoglobulin and acacia gum at pH 4.2. Food Hydrocolloids, 14, 403–413.
29 Soper, J. C. 1995. Utilization of coacervated flavors. ACS-Symposium Series, No. 590, 104–112.
30 Shefer, A., Shefer, S. 2003. Multicomponent Controlled Release System for Oral Care, Food Products, Nutraceti-
31 cal, and Beverages. US20030152629.
32 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.
33 Solans, C., Izquierdo, P., Nolla, J., Azemar, N., Garcia-Celma, M. J. 2005. Self-assembly of surfactants and phos-
34 pholipids at interfaces between aqueous phases and thermotropic liquid crystals. Curr. Opin. Colloid Interface
35 Sci., 10, 102–110.
36 Sonneville-Aubrun, O., Simonnet, J. T., L’Alloret, F. 2004. Nanoemulsions a new vehicle for skincare products.
37 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](-)-
38 epigallocatechin gallate, a cancer preventive tea polyphenol, in mouse tissue, Carcinogenesis, 19, 1771–1776.
39 Tolstoguzov, V. B.1991. Functional properties of food proteins and role of protein–polysaccharide interaction.
40 Food Hydrocolloids, 4, 429–468.
41 Turgeon, S. L., Beaulieu, M., Schmitt, C., Sanchez, C. 2003. Protein–polysaccharide interactions: phase-ordering
42 kinetics, 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-
43 ervates. Biomacromolecules, 5, 1437–1445.
44 Weinbreck, F., Wientjes, R. H. W., Nieuwenhuijse, H., Robijn, G. W., de Kruif, C. G. 2004b. Rheological proper-
45 ties of whey protein/gum arabic coacervates. J. Rheol., 48, 1215.
46S Wildman, R. E. C. 2001. In: Wildman, R. E. C. (Ed.), Handbook of Nutraceuticals and Functional Foods. New
47N York: CRC Press.
Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 147

Preserving and Controlling the Release of Enzymes and Phytochemicals 147

Xia, J., Dubin, P. L. 1994. Protein–polyelectrolyte complexes. In: Dubin, P. L., Bock, J., Davis, R., Schulz, D. N., 1
Thies, C. (Eds.), Macromolecular Complexes in Chemistry and Biology (pp. 247–271). Berlin: Springer- 2
Verlag.
Yang, C. S., Maliakal, P., Meng, X. 2002. Inhibition of carcinogenesis by tea. Annu. Rev. Pharmacol. Toxicol., 42,
3
25–54. 4
Yeo, Y., Bellas, E., Firestone, W., Langer, R., Kohane, D. E. 2005. Complex coacervates for thermally sensitive 5
controlled release of flavor compounds. J. Agric. Food Chem., 53, 7518–7525. 6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
S46
N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 149

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

7 Microencapsulation of Flavors by 1
2
Complex Coacervation 3
4
5
Curt Thies 6
7
8
9
10
11
Introduction 12
The encapsulation of flavors was first reported in the 1930s when it was observed that a 13
volatile substance, isopropanol, was retained by a spray-dried particle (Thies, 1999). This 14
observation catalyzed the development of spray dry flavor encapsulation, a technology 15
responsible today for the daily production of tons of encapsulated flavor products globally. 16
Reineccius (2004) and others (Brenner, 1983; Re, 1998; Liu et al., 2001) have discussed 17
spray dry encapsulation technology in some detail. 18
Although spray drying is currently the dominant flavor encapsulation technique, a num- 19
ber of alternate encapsulation technologies exist and offer a potential means of producing 20
unique flavor-loaded microcapsules. Complex coacervation encapsulation procedures fall 21
into this category. Accordingly, this contribution is a discussion of various aspects of com- 22
plex coacervation encapsulation technology and the encapsulation of flavors for food prod- 23
ucts. Only complex coacervation processes based on food-grade polymers are considered 24
here. Although fragrances are not considered in this contribution, much of the discussion is 25
also applicable to the encapsulation of fragrances as well as other complex core materials. 26
27
28
Flavor Encapsulation 29
The preparation of flavor-loaded microcapsules is a complex task. It is much more compli- 30
cated than it appears at first glance, because flavor microcapsules must meet a series of 31
requirements. One requirement is the production of microcapsules that retain the desired 32
properties of the flavor encapsulated. Each flavor is a unique and complex mixture of many 33
compounds. These compounds have a broad range of structures with vapor pressures, sol- 34
vent solubility, and stability that differ significantly. Any useful encapsulation technology 35
must be able to accommodate this variability. Ideally, the chemical composition of the fla- 36
vor is unchanged by the encapsulation process, and the encapsulated flavor is identical in 37
all respects to the unencapsulated flavor. In reality, this is generally not the case. Encapsula- 38
tion processes typically change the chemical composition of a flavor in some way. Loss of 39
more volatile or more water-soluble components during an encapsulation process is common. 40
Such losses can have a significant effect on the desired olfactory properties of the flavor. 41
Flavor-loaded microcapsules must contain enough active agents to cause the desired 42
effect. The amount of flavor required varies with the nature of the flavor and intended food 43
product. Although flavor impact can be altered by varying the flavor loading of a capsule of 44
fixed size as well as by varying capsule size, the degree of variation may be limited by the 45
nature of the food product. For example, capsule size variations may be limited by the need S46
N47

149
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 150

150 Chapter 7

1 to retain structural integrity during processing while having the ability to be ruptured by
2 chewing. Variations in amount of flavor carried by a capsule of fixed size may be limited
3 by the capsule formation process as well as the retention or barrier properties of the cap-
4 sule shell.
5 Stability of flavor-loaded capsules during processing and storage is an issue that must be
6 addressed. Such capsules must have acceptable stability from the time of formation to con-
7 sumption of the food product. Stability during processing of capsules as they are incorpo-
8 rated into a food product can be a problem if this involves high shear or a combination of
9 high temperature and shear. Shelf life stability after incorporation and storage in a food
10 product is important as is the ability of capsules to release their contents during food prepa-
11 ration or consumption. This series of stability requirements is imposing and often limits
12 actual capsule performance. Factors that affect capsule stability include oxygen, moisture,
13 heat, and light. In principle, the shell of a capsule should protect an encapsulated flavor
14 from these agents, but deficiencies in either the capsule shell or the material(s) from which
15 the shell is prepared may cause a shell to provide inadequate protection. Shell materials
16 typically used to form food-grade flavor capsules may experience major property changes
17 during food-processing steps that involve heat and moisture. Of course, flavor-loaded
18 microcapsules must meet specifications imposed by governmental regulatory agencies
19 responsible for food safety. This requirement puts a restriction on the shell materials that
20 can be used. It also limits the nature and amount of processing agents used in a capsule-
21 formation process.
22 In summary, the complex series of specifications associated with the formation of flavor-
23 loaded microcapsules makes their preparation an interesting field of study. Much can be
24 done in order to produce capsules that more closely approach the degree of perfection
25 desired. Studies by various workers of the diffusion barrier properties of candidate shell
26 materials merit review, because they provide much insight into the properties of such mate-
27 rials and help one develop a realistic appreciation for the limitations of specific shell mate-
28 rials and capsule formation processes (Menting and Hoogstad, 1967; Kerkof and Thijssen,
29 1974; Thijssen, 1975; Rulkans and Thijssen, 1978; Goubet et al., 1998). Although most of
30 these involve spray drying and freeze drying studies, the results obtained are applicable to
31 capsules formed by any encapsulation process.
32
33
Complex Coacervation
34
35 Before discussing complex coacervation encapsulation processes, it is appropriate to con-
36 sider the nature of complex coacervate systems and some of their characteristic features.
37 Complex coacervation is the liquid/liquid phase separation that occurs when solutions
38 of two or more oppositely charged polyelectrolytes are mixed under suitable conditions.
39 Two liquid phases are formed: the coacervate phase and the supernatant or equilibrium
40 liquid phase. The coacervate phase is a relatively concentrated polymer solution that par-
41 ticipates in a complex coacervation encapsulation system. It is in this phase that capsule
42 shell forms. The supernatant phase is a dilute polymer solution and serves as the continu-
43 ous phase in which capsule formation occurs. Dilution favors complex coacervation and is
44 a property that distinguishes complex coacervation from other polymer phase-separation
45 phenomena.
46S Complex coacervation is affected by many variables. Bungenberg de Jong’s experimental
47N studies in the 1930s and 1940s provide much useful background data about the phenomenon
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 151

Microencapsulation of Flavors by Complex Coacervation 151

(Bungenberg, 1949). Since then, a variety of workers have considered various aspects of 1
coacervation including theoretical analyses based on polymer solution thermodynamics 2
(Burgess, 1990; Veis, 1970; Schmitt et al., 1998). Although this information provides a guide 3
for developing complex coacervation microencapsulation procedures, it is important to rec- 4
ognize that conditions that optimize the degree of coacervation in a specific system may not 5
be conditions under which useful microcapsules can be formed. For example, these condi- 6
tions may produce a complex coacervate that is too viscous to yield acceptable capsules. 7
8
9
Selected Properties of Complex Coacervates
10
Many complex coacervation systems suitable for the production of microcapsules exist. In 11
virtually all cases, gelatin is the polycation used. A wide range of polyanions is used. Each 12
system operates under a unique set of conditions and has a unique set of properties once 13
formed. This reflects differences in nature and frequency of ionic groups distributed along 14
the chains of the polymers involved in a specific complex coacervation procedure. Differ- 15
ences in polymer chain structure and molecular weight (MW) are other factors that influ- 16
ence coacervation. 17
One of the polyelectrolytes used in a typical complex coacervation encapsulation proce- 18
dure is a natural polymer with a complex molecular structure. For example, gelatin polymer 19
molecules are made up of a number of different amino acids with different pendent groups. 20
Because anionic and cationic pendent groups are distributed along the polymer chain, gela- 21
tin is a polyampholyte. The cationic groups are primary amino groups, while the anionic 22
groups are carboxyl groups. The degree of ionization of these ionic groups varies with pH, 23
so the net charge carried by a gelatin molecule varies with pH. 24
Gelatins formed by acid hydrolysis of collagen are classified as Type A or acid precursor 25
gelatins. Alkaline hydrolysis yields Type B or alkaline precursor gelatins. The isoelectric 26
point (pI) of Type A gelatins is typically 8–9, while the typical pI of Type B gelatins is 4–5. 27
The reduced pI value of Type B gelatins is caused by hydrolysis of pendant amide groups 28
under alkaline conditions. The number of primary amino groups distributed along a gelatin 29
chain is essentially independent of the hydrolysis procedure. Although both types of 30
gelatins produce complex coacervates suitable for microcapsule formation, Type A gelatins 31
historically have been used most. Significantly, for gelatin to carry a net cationic charge, 32
it must be at a pH lower than its pI. 33
Although gelatin is the polycation involved in the formation of complex coacervates 34
used in microencapsulation processes, many different polyanions are used. They differ 35
greatly in anion group distribution along a polymer chain as well as the nature of this 36
group. This is particularly true of natural polymers that carry an anionic group. Gum arabic 37
(GA) and alginate, two polysaccharides, derive their anionic character from carboxyl 38
groups distributed along their polymer chains. In GA, such groups are located on short- 39
chain branches hanging off the primary polymer chain. Approximately 20% of the sugar 40
units in GA contain a carboxyl group. In contrast, alginate molecules are linear polymer 41
chains and every sugar in the chain has a carboxyl group. The degree of ionization of 42
carboxyl groups is a strong function of pH and steadily decreases as pH decreases. The 43
anionic character of sodium polyphosphate, an inorganic material, is due to the phosphate 44
group. Carrageenan, a polysaccharide with a linear chain, has sulfate groups distributed 45
along its chain. The average number of sulfate groups per sugar unit varies with the type of S46
carrageenan. N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 152

152 Chapter 7

1 Because the polymers used to form complex coacervates differ significantly in composi-
2 tion and structure, properties of complex coacervates formed by different polymers differ
3 significantly. In order to illustrate this point, Table 7.1 contains degree of coacervation and
4 enrichment data at 50°C for three gelatin-based complex coacervation systems used to pre-
5 pare microcapsules: gelatin/gum arabic (GGA), gelatin/polyphosphate (GP), and gelatin/
6 sodium alginate (GAlg) (Commandur et al., 1989). Degree of coacervation (ρ) is defined as
7 the fraction of total polymer in the system that is in the coacervate (Veis and Aryani, 1960).
8 Enrichment (ε) is defined as the ratio of polymer concentration in the coacervate phase to
9 that in the supernatant phase (Veis and Aryani, 1960). GGA and GP coacervates were
10 formed by interacting 285 bloom Type A gelatin with GA and sodium hexametaphosphate,
11 respectively. GAlg coacervates were formed by interacting 231 bloom Type A gelatin with
12 a hydrolyzed alginate.
13 Each coacervation system was studied at three initial solids concentrations and three pH
14 values in order to illustrate how changes in these variables affect coacervate formation.
15
16 Table 7.1. Tabulation of degree of coacervation (ρ) and enrichment data (ε) at 50ºC for several
17 coacervation systems
18
Coacervate system Initial solids w/v (%) pH Degree of coacervation Enrichment
19
20 GGA 3.96 4 0.86 22
21 3.3 4 0.88 34.8
2.83 4 0.89 47.7
22
23 3.96 4.2 0.78 11.3
24 3.3 4.2 0.81 17
2.83 4.2 0.81 21.2
25
26 3.96 4.4 0.81 12.1
27 3.3 4.4 0.8 16.6
2.83 4.4 0.83 24.2
28
29
GAlg 2.11 4 0.86 66.7
30 1.81 4 0.83 68
31 1.59 4 0.82 71.7
32
2.11 4.2 0.66 25.3
33 1.81 4.2 0.73 42.5
34 1.59 4.2 0.7 54.7
35
2.11 4.4 0.78 35.7
36 1.81 4.4 0.81 53
37 1.59 4.4 0.79 54.7
38
39 GP 5 4 0.74 11.5
40 4.54 4 0.78 15.5
4.17 4 0.78 20.9
41
42 5 4.2 0.72 9.4
43 4.54 4.2 0.71 11.3
4.17 4.2 0.74 14.5
44
45 5 4.4 0.61 4.8
46S 4.54 4.4 0.57 5.7
4.17 4.4 0.65 10.2
47N
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 153

Microencapsulation of Flavors by Complex Coacervation 153

Indeed, at sufficiently high concentration and pH values, complex coacervation does not 1
occur. Initial solids content is defined as the total polymer solids in the system at the time 2
when complex coacervation occurred. The data in Table 7.1 were obtained by using initial 3
solids of 1.6–5 w/w% and pH values of 4.0–4.4 pH. Although these initial solids and pH 4
values are typical for many coacervation systems used to make microcapsules, situations 5
exist where suitable coacervates will form when values of one or both parameters fall out- 6
side these ranges. 7
The gelatin/polyanion ratio used for forming gelatin-based complex coacervates varies 8
with ionic equivalent weight of the polyanion(s) used. The GGA complex coacervate data 9
in Table 7.1 were formed by using a 1:1 w/w ratio of gelatin and GA, because both poly- 10
mers have an ionic equivalent weight of roughly 1000. Alginates have an ionic equivalent 11
weight of approximately 180, so the gelatin/alginate the ratio used was 3.7:1 w/w. The w/w 12
gelatin/polyphosphate ratio was 9:1. It is high, because polyphosphates have a low ionic 13
equivalent weight. 14
In all cases, the polyanion ratios reported here are those typically used by the author to 15
produce complex coacervates suitable for microcapsule formation. Complex coacervates 16
will form when a coacervation system contains excess gelatin or polyanion, but it is com- 17
mon practice to use gelatin/polyanion ratios that approach ionic equivalency. Although the 18
data in Table 7.1 are for coacervation systems based on one polyanion, mixtures of several 19
chemically different polyanions can be used to produce gelatin-based coacervates suitable 20
for microcapsule formation. This enables one to develop a broad range of complex coacer- 21
vate systems suitable for microcapsule formation. 22
The data in Table 7.1 (Commandur et al., 1989) show that ρ and ε at 50°C are affected by 23
the nature of the polyanion involved in coacervate formation, system pH, and initial solids 24
content of the system. For all coacervation systems examined, values of ε at constant pH 25
increase as the initial solids content decreases. This reflects the increase in intensity of 26
coacervation upon solution dilution, a characteristic feature of complex coacervation. In 27
contrast, other polymer phase-separation phenomena such as polymer/polymer incompati- 28
bility and salting out (simple) coacervation are favored by increasing the concentration of 29
the molecules involved. 30
Values of ρ for the GGA system fall between 0.8 and 0.9 over the range of initial solids 31
and pH values examined. Thus, in these GGA systems, 80–90 w/v% of the polymers were 32
concentrated in the coacervate phase. The GGA coacervate phases formed are 15–26 vol% 33
of total system volume and have a solids content of 10–14 w/v%. Solids content of the 34
supernatant phase was 0.3–0.9 w/v%. Since values of ε found for the GGA systems range 35
from 11 to 35, a high degree of polymer partitioning was achieved. At constant initial 36
solids, values of ε decrease as pH increases. 37
GP coacervate phases occupy 13–21 vol% of total system volume and have a solids con- 38
tent of 13–21 w/v%. GP ρ values of 0.6–0.8 and ε values of 5–21 are lower than the range 39
of ρ and ε values found for GGA and GAlg systems. Thus, the GP coacervate system 40
achieves a lower degree of polymer partitioning. The solids content of GP supernatant 41
phases range from 1 to 2.3 w/v%, considerably higher than that observed with GGA and 42
GAlg systems. Because gelatin concentrations of approximately 2 w/v% approach the con- 43
centration at which gelatin solutions gel, operating conditions of an encapsulation process 44
based on GP must be adjusted to keep the supernatant solids concentration below 2 w/v%. 45
The solids content of a GAlg coacervate phase at 50°C varies from 15 to 22 w/v% while S46
the solids content of a GAlg supernatant phase varies from 0.3 to 0.9 w/v%. The GAlg N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 154

154 Chapter 7

1 coacervate is 4–7 vol% of total system volume, considerably lower than the values
2 observed with GGA or GP coacervates. Values of ρ for the GAlg systems are 0.7–0.9, a
3 range similar to but broader than that observed with GGA coacervates. The 25–72 range of
4 ε values for GAlg coacervate systems is higher than that observed with GGA or GP sys-
5 tems. Thus, GAlg coacervate systems more effectively concentrate or partition into the
6 coacervate phase the polymers involved in complex coacervation. At first glance, this is
7 surprising, because the volume of the GAlg coacervate phase is much smaller than that of
8 the GGA and GP coacervate phases, while the solids content of the GAlg coacervate phase
9 is similar to that of the GGA and GP coacervate phases. Closer analysis leads to the recog-
10 nition that the initial solids content of the GAlg system is measurably lower than that of the
11 GGA and GP systems. Thus, the GAlg coacervate phase contains a higher percentage of
12 polymers present in the GAlg system, even though it occupies a smaller volume fraction
13 than the GGA and GP coacervate phases and has a solids content similar to these systems.
14 The ρ and ε data reported in Table 7.1 provide valuable insight into the nature of three
15 complex coacervate systems, but they reveal nothing about coacervate rheology or the tem-
16 perature at which the coacervates gel. Coacervate rheology is a primary variable that
17 affects capsule shell formation and capsule aggregation. Accordingly, the viscosity of a
18 number of coacervate phases was measured over a range of temperatures by capillary
19 viscometry (Commandur et al., 1989). Table 7.2 summarizes results of these measure-
20 ments. The data show that the sodium alginate and GA solutions used to form GGA and GP
21 coacervates have a viscosity of 3–6 cS at 50°C. This viscosity increases as the temperature
22 is reduced to 30°C, but the viscosity increase caused by cooling is not pronounced, because
23 neither polymer alone gels on cooling. In contrast, viscosity of the gelatin solutions exam-
24 ined steadily increases as the solution temperature is reduced from 50°C to 35°C. The
25 viscosity of most such solutions becomes unstable at 32°C. That is, the recorded viscosity
26 steadily increases toward infinity as the time at 32ºC increases. This viscosity increase is
27 due to the onset of gelation.
28 Not shown in Table 7.2 are viscosity data for GGA, GP, and GAlg supernatant phases
29 that exist in equilibrium with the GGA, GP, and GAlg coacervate phases for which viscos-
30 ity data were obtained. Most supernatant phases have a viscosity below 1 cS at tempera-
31 tures ranging from 50°C to 35°C and provide no indication that they will gel on further
32 cooling. Exceptions are two GP supernatant solutions isolated from pH 4.4 GP coacervate
33 systems. The viscosity of both solutions remained below 1.5 cS as they were cooled to
34 35°C, but the upward slope of their temperature–viscosity plots suggests that both will ulti-
35 mately gel.
36 GGA coacervate viscosity at pH 4.4 and 50°C ranged from 23 to 58 cS, i.e., 2 to 5 times
37 greater than the 11.3 cS viscosity of a 10% solution of 285 bloom Type A gelatin at 50ºC.
38 Viscosity of the GGA coacervates steadily increases as the system is cooled. They either
39 gel or become unstable due to onset of gelation as the temperature falls below 35°C.
40 Decreasing the pH of GGA coacervate formation from 4.4 to 4.0 increases coacervate vis-
41 cosity at all temperatures examined.
42 The viscosity of GP coacervates at 50°C is 47–373 cS, measurably higher than the vis-
43 cosity of GGA coacervates. Reducing the pH of a GP coacervate from 4.4 to 4.0 causes a
44 major increase in coacervate viscosity and raises the GP coacervate gelation temperature
45 above 35°C.
46S The viscosity of all GAlg coacervates at 50°C is 20–60 times higher than that of most
47N GGA and GP coacervate phases at 50°C. Although GAlg coacervate phases have a very
Table 7.2. Changes in viscosity of various coacervate systems at different temperatures (from Commandur et al., 1989, with permission)

Viscosity (cS)
Initial solids Solids
System (w/v%)a pH (w/v%)b 50°C 45°C 40°C 37°C 35°C 32.5°C 30°C

GGA coacervate 3.96 4.4 10.4 23 26 29 35 46 107 Gelled


2.83 4.4 12.1 39 42 47 62 96 Unstable Gelled
GGA coacervate 3.96 4 13.2 39 42 48 79 97 143 Gelled
2.83 4 14.3 58 63 74 Unstable Gelled
GP Coacervate 5 4.4 12.3 47 56 98 221 Gelled
4.17 4.4 14.3 67 74 120 679 Gelled
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 155

GP Coacervate 4.55 4.2 19.8 298 342 1782 Gelled


4.17 4 20.9 373 402 2606 Gelled
GAlg Coacervate 2.11 4.4 15.6 679 806 1636 3396 Gelled
1.59 4.2 18.4 1245 1480 2485 7404 Gelled
GAlg Coacervate 2.11 4 20 1391 1665 2806 6220 Gelled
1.59 4 18.6 1382 1698 3254 7658 Gelled
Gelatin solution Type A 10 11.5 13.2 14.8 17 20 Unstable Gelled
(285 bloom) 15 31 35 40 52.1 66.6 Unstable Gelled
Gelatin solution Type A 10 5.2 7.3 8 9.2 10.3 11.5 16.2 Unstable
(231 bloom) 15 5.4 15.9 17.8 20.6 24.6 28.9 Unstable Gelled
Sodium alginate solution 2 5.5 3 3.4 3.7 4 4.2 4.8
Gum arabic solution 10 4.2 4.1 4.5 4.9 5.4 5.5 6.3

a
Total system solids at the time of coacervation.
b
Total solids of solution used for capillary viscosity measurement.

155
N47
S46
45
44
43
42
41
40
39
38
37
36
35
34
33
32
31
30
29
28
27
26
25
24
23
22
21
20
19
18
17
16
15
14
13
12
11
10
9
8
7
6
5
4
3
2
1
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 156

156 Chapter 7

1 high viscosity at temperatures ranging from 50°C to 40°C, they do not appear to gel until
2 cooled to 35°C.
3 As the data in Table 7.2 show, complex coacervate viscosity is a strong function of the
4 coacervation system, pH, and temperature. Characterizing the effect of temperature
5 changes on rheology of a coacervate system is an important task, because all gelatin-based
6 complex coacervation encapsulation protocols involve a cooling step that lowers the sys-
7 tem temperature below the gel temperature of the coacervate. In such processes, complex
8 coacervate formation always occurs above the coacervate melt temperature so that the
9 coacervate formed is initially a liquid. It must have a viscosity that is sufficiently low to
10 enable it to engulf dispersed core material droplets or particles, thereby coating them with a
11 thin film of liquid coacervate. Once a coacervate is formed, the two-phase system is cooled
12 below the gel temperature of the coacervate, thereby setting the gel structure of the coacer-
13 vate. This transforms the thin liquid film that surrounds the small droplets of core material
14 into a thin gel coating. Viscosity of a coacervate above its melt temperature and changes in
15 its rheology as cooling occurs have a major impact on the success of a complex coacerva-
16 tion encapsulation process.
17 The viscosity data in Table 7.2 cover a very large range of values. Although capillary
18 viscometry is appropriate for measuring the viscosity of Newtonian fluids, it has not been
19 determined that all complex coacervates exhibit Newtonian flow behavior, especially at
20 temperatures that approach the gel point. Leuenberger (1991) reported that 10% w/w%
21 solutions of several different gelatin samples at 40°C are linear in the shear rate range of
22 10–350 s–1. Deviations from linearity occur at high shear rates. Because of the relative flu-
23 idity at 50°C of GGA and GP coacervates, it is believed that they exhibit Newtonian behav-
24 ior at this temperature. The long capillary flow times of the GA coacervates at 50°C suggest
25 that such coacervates exhibit non-Newtonian flow behavior. Although additional measure-
26 ments are needed in order to properly characterize the rheological behavior of a range of
27 complex coacervates, the data in Table 7.2 provide a means of comparing the apparent vis-
28 cosity of several coacervates used to form microcapsules. These data illustrate the signifi-
29 cant effect that composition of a complex coacervate has on its rheological properties. This
30 has a profound effect on capsule formation.
31 It is relevant to note that Koh and Tucker (1988a, b) characterized the gelatin–
32 carboxymethylcellulose (CMC) complex coacervate system. They reported characterization
33 data similar to that shown in Tables 7.1 and 7.2 for this system. Although their characteriza-
34 tion data as well as that shown in Tables 7.1 and 7.2 shed valuable insight into the specific
35 complex coacervation systems studied, it must be recognized that such data represent typi-
36 cal properties of a given complex coacervate system. Specific ρ and ε values reported for
37 any complex coacervate system can be difficult to precisely reproduce consistently within
38 the same laboratory by the same person, let alone different laboratories and different per-
39 sons. This problem is caused by the sensitivity of complex coacervation to many factors.
40 Experimental variations can be minimized by establishing standard experimental protocols.
41 For example, all of the data shown in Table 7.1 were obtained by using aqueous solutions
42 polymers from the same lot. Distilled water was the solvent. Very different results would
43 most likely occur if tap water is used, because tap water can contain a variety of salt ions,
44 and salt ions repress complex coacervation. Lot-to-lot variations in properties of either the
45 gelatin or the polyanion used will also affect results obtained. In order to minimize varia-
46S tion in reported results, the polymer solution preparation protocol must be standardized as
47N must the length of solution storage before use.
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 157

Microencapsulation of Flavors by Complex Coacervation 157

In cases where a flavor is being encapsulated by a process based on complex coacerva- 1


tion, water-miscible or partially water-soluble components present in the flavor can affect 2
the coacervation process and nature of the coacervate formed. Seemingly small variations 3
in a coacervate system can have a major effect on complex coacervation results. 4
5
6
Complex Coacervation Encapsulation Processes 7
8
Bungenburg de Jong’s studies of the complex coacervation of gelatin and GA formed the
9
basis for the original GGA complex coacervation encapsulation procedure reported by
10
Green and Schleicher (1957). Figure 7.1 is a flow diagram of their process. The first step is
11
to emulsify the material being encapsulated in a warm (40–60°C) aqueous gelatin solution.
12
This material is commonly called the core material. It is typically a water-immiscible oil,
13
but could be a water-insoluble solid. Oil emulsification typically is carried out in a warm
14
8–11 w/w% gelatin solution because such concentrated solutions increase the ease of emul-
15
sification to a desired drop size.
16
The second step is to add GA and dilution water to the system followed by adjustment of
17
the pH to a value at which sufficient complex coacervate phase to encapsulate the dispersed
18
oil droplets is formed. As noted in Table 7.1, this pH typically falls in the range of 4.0–4.4,
19
although higher or lower pH values may be needed for a specific core material. Dilution
20
water is added to the system at this point in order to lower the total polymer solids content
21
from the 8 to 11 w/v% used in the emulsification step to the 2.83–3.96 w/v% at which com-
22
plex coacervation occurs (see Table 7.1).
23
The third step is to cool the system below the gel point of the coacervate, thereby caus-
24
ing the coacervate to gel. In order to improve capsule shell stability, capsules with shells
25
26
27
28
29
Water- Aqueous
Adjust pH
30
gum arabic
immiscible
oil
solution (e.g.,4.0–4.6) 31
(40–60°C)
32
33
34
Cool Harvest
35
Mixer Oil-in-water
emulsion
Mixer (to gel
coacervate)
Crosslink microcapsules 36
37
38
39
Aqueous
40
gelatin Water 41
solution (40–60°C)
(40–60°C) 42
43
44
45
Figure 7.1. Flow diagram of gelatin gum arabic (GGA) complex coacervation encapsulation S46
procedure (from Green and Schleicher, 1957, with permission). N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 158

158 Chapter 7

1 formed by the complex coacervation of gelatin are typically chemically cross-linked with
2 glutaraldehyde (glut) before they are isolated.
3 Today, complex coacervation encapsulation protocols still follow the basic Green and
4 Schleicher (1957) procedure shown in Figure 7.1. Specific concentrations and types of the
5 polymers involved may vary, but the three-step protocol of emulsion formation, coacervate
6 formation, and coacervate gelation is still used. Numerous variations of this process have
7 appeared since 1957, because many polyanions other than GA have been shown to interact
8 with gelatin to produce complex coacervates suitable for microcapsule formation. This
9 has led to the development of a broad family of complex coacervation encapsulation
10 procedures. Specific coacervation procedures acceptable for the formation of flavor-loaded
11 microcapsules are those in which gelatin interacts with food-grade polyanions such as GA,
12 sodium alginate, carrageenan, pectin, CMC, gellan, and sodium polyphosphate. Combina-
13 tions of these polyanions can also be used.
14 Significantly, the data in Table 7.1 show that properties of the coacervate phase formed in
15 each protocol differs in some manner. Even if the difference is small, considerable time may
16 be required to experimentally define conditions required for suitable capsule formation.
17 Table 7.3 is a list of a number of complex coacervation encapsulation protocols reported
18 by various workers in journals or patents. It is not an exhaustive list, but the references cited
19 provide information about a number of gelatin-based complex coacervation systems that
20 are candidates for flavor oil encapsulation. All the protocols are based on materials that the
21 author regards as suitable for encapsulating food flavors.
22 It is interesting that no two protocols disclosed in Table 7.3 are similar. They all have an
23 emulsification, coacervation, and cooling step, but the conditions under which these are
24 carried out are not standardized; that is, a standard protocol is not followed. Some workers
25 use Type A gelatin, while others use Type B. Gelatin bloom strength varies. Reported coac-
26 ervation pH values fall between 3.0 and 5.5. Some workers adjust pH with acetic acid,
27 while others use HCl.
28 Gelatin involved carries a positive charge in the 3.0–5.5 pH range and serves as the
29 polycation in all but one of the complex coacervation encapsulation systems cited in Table
30 7.3. The sole case where this may not be the case is the coacervation of Type B gelatin with
31 chitosan at pH 5.25–5.5 (Remunan-Lopez and Bodmeier, 1996). If this system is a complex
32
33
34 Table 7.3. Complex coacervation encapsulation protocols based on food-grade shell materials
35
36 Gelatin Polyanion Coacervation (pH) References
37 Type B, 225 bloom CMC 3.0–4.0 (0.1 M HCl) Koh and Tucker (1988a, b)
38 Type B, 225 bloom Acacia 3.9 (1 M HAc) Jegat and Taverdet (2000)
39 Type A Acacia 4.2 (glacial, acetic acid) Mayya et al. (2003)
40 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)
41
Type B, 175 and Chitosan glutamate 5.25–5.50 (0.5 M HCl) Remunan-Lopez and
42 225 bloom Bodmeier (1996)
43 Type A, 175 bloom Gellan 3.5–5.50 (0.5 M HCl) Chilvers and Morris (1995)
44 Type A Sodium alginate 3.5–4.5 (0.5 N HCl) Joseph and Venkataran (1995)
45 Type A, 275 bloom Sodium pyrophosphate 4.5 (10% acetic acid) Yan (2005)
Fish gelatin CMC/gum arabic NA Soper (1997)
46S
Type A Gum arabic 3.9–4.7 (10% acetic acid) Saeki and Hosoi (1984)
47N
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 159

Microencapsulation of Flavors by Complex Coacervation 159

coacervation system, the gelatin must act as the polyanion, while chitosan is the polycation. 1
At pH 5.25–5.5, such protocols should be limited to Type B gelatins. The pI of Type A gela- 2
tin is typically 8–9, so its complex coacervation with chitosan should be limited to pH 3
values above this. 4
Several of the studies referenced in Table 7.3 explore how various parameters affect 5
microcapsule formation by complex coacervation. For example, Jegat and Taverdet (2000) 6
found that the relationship of stirring rate and size of capsules produced by gelatin-GA 7
coacervation agrees well with predictions of the inertial breakup theory. Mayya et al. 8
(2003) reported that addition of low concentrations of sodium dodecyl sulfate (SDS) to the 9
aqueous phase during emulsification promotes GGA coacervate capsule shell formation on 10
dispersed paraffin oil droplets. The SDS concentration used was well below its CMC. These 11
workers suggest that SDS causes deposition of a two-layer shell. Duquemin and Nixon 12
(1985) examined the effect of SDS and cetrimide on complex coacervate formation and 13
encapsulation by complex coacervation. Three SDS concentrations were used: 0.07, 0.2, 14
and 0.35 w/v%. The 0.07 w/v% SDS solution was below the CMC of SDS. Although the 15
weight of coacervate obtained at pH 4.35 and 40°C decreased linearly with increasing SDS 16
concentration, 0.07 w/v% SDS caused very little reduction. Significantly, Duquemin and 17
Nixon (1986) reported that 0.07 w/v% SDS caused a major reduction in the amount of core 18
material (phenobarbitone) encapsulated. 19
The author has historically avoided the addition of surfactants to a complex coacerva- 20
tion system. I am concerned that surfactants will have a negative effect on capsule quality. 21
In my experience, nonionic surfactants have consistently had a negative effect on capsule 22
quality; this is consistent with the observations of Luzzu and Gerraughty (1964). Neverthe- 23
less, the positive results with an anionic surfactant reported by Mayya et al. (2003) coupled 24
with the positive result with a cationic surfactant (cetrimide) reported by Duquemin and 25
Nixon (1985) indicate that further studies of how ionically charged surfactants affect com- 26
plex capsule formation are warranted. 27
Although it is not complex coacervation, the report by Vinietsky and Magdassi (1997) 28
that soybean oil droplets are encapsulated by an SDS–Type A Gelatin complex is interest- 29
ing. Encapsulation occurred at pH 4 and an SDS concentration of 1.5–2.0 mM. This SDS 30
concentration range is below the CMC of SDS. The gelatin concentration after SDS addi- 31
tion was 0.3 mM. 32
Yan (2005) claims the formation of a microcapsule structure in which an agglomeration 33
of primary microcapsules is encapsulated by an outer shell. Each individual primary micro- 34
capsule in the agglomeration is claimed to have a primary shell and this agglomeration is 35
encapsulated by an outer shell. Yan (2005) described the formation of this capsule structure 36
by a GP complex coacervation procedure in which the aqueous phase contained 0.5% 37
sodium ascorbate. The first step is to prepare an 8.33 w/w% gelatin and 0.5% sodium 38
ascorbate solution in water at 50°C. A fish oil concentrate is emulsified in this solution 39
under high shear. The resulting oil-in-water emulsion has oil droplets with an average size 40
of 1 µm. After it is formed, the emulsion is diluted by addition of a 0.5% aqueous sodium 41
ascorbate solution at 50°C. A 5% sodium polyphosphate plus 0.5% sodium ascorbate solu- 42
tion is then added. After the pH is adjusted to 4.5, the system is cooled, thereby forming 43
coacervates that coats the individual oil droplets, which creates primary microcapsules. 44
The temperature at which this occurs and the rate of cooling to this temperature are not 45
mentioned, but it is noted that the primary microcapsules begin to agglomerate as cooling is S46
carried out to above the gel point of the coacervate. Upon further cooling of the system, N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 160

160 Chapter 7

1 additional coacervate forms and coats the agglomerates of primary microcapsules, thereby
2 creating an agglomerate of primary microcapsules with an outer shell and an average size
3 of 50 µm. Once the system is cooled to 5°C, glut is added and allowed to react for 12 h with
4 the suspended capsules in the system under continuous stirring for 12 h. The microcapsule
5 suspension is subsequently washed with water and spray dried to give a free-flow powder.
6 Two interesting complex coacervation encapsulation systems that contain an essentially
7 nonionic polymer have been reported. Although such polymers are not believed to be
8 directly involved in complex coacervate formation, they undoubtedly affect it in some man-
9 ner. One system was reported by Jizomoto (1984). He reported that the addition of small
10 amounts of a nonionic polymer like polyethylene oxide or poly (ethylene glycol) expanded
11 the pH range over which a GGA complex formed. By using this approach, it was possible
12 to prepare GGA capsules loaded with paraffin oil at pH 6.5. The pH range over which GGA
13 coacervates were formed was 2–9. This type of system offers a possible method of encap-
14 sulating active agents sensitive to the acidic conditions characteristic of a typical GGA
15 encapsulation system. The author views it as a process that combines complex coacervation
16 with polymer–polymer incompatibility.
17 The second procedure was reported by Xing et al. (1973). They prepared capsaicin-
18 loaded GGA microcapsules in the presence of low concentrations of hydroxylethyl cellu-
19 lose (HEC), poly (vinyl alcohol) (PVA), and poly (vinyl pyrrolidone) (PVP). It was noted
20 that the presence of HEC yielded GGA microcapsules with a better morphology and geom-
21 etry than capsules prepared in the presence of PVA or PVP. These polymers were classified
22 by the authors as surfactants, but it is possible that differences in their polymer–polymer
23 incompatibility behavior could contribute to the observed results even at the low concentra-
24 tion used.
25
26
Cross-Linking of Gelatin-Based Coacervate Capsule Shells
27
28 When initially formed, gelatin-based complex coacervate capsule shells are highly water
29 swollen and melt if reheated. They also dissolve in warm aqueous media, thereby releasing
30 their core. This latter property is highly desirable in many food applications, but it also
31 poses problems because the isolation of discrete gelatin-based coacervate capsules that
32 have not been cross-linked in some way is difficult. The shell of such capsules is highly
33 water swollen and melts if subjected to relatively low levels of thermal energy. Extractive
34 drying with a water-miscible solvent at low temperatures is one option, but extraction of
35 core material by the extractive drying solvent must be minimized.
36 In any case, drying capsules that have not been cross-linked in some manner is an issue.
37 For this reason, it is common practice to treat gelatin coacervate shells in some way in order
38 to stabilize them, so that they can be dried using conventional spray-drying techniques and
39 fluidized bed units. Historically, this has been done by using aldehydes or tannins to cross-
40 link gelatin-based capsule shells. Treatment with an aldehyde has been the most common
41 approach taken. Formaldehyde and glut are two aldehydes cited in many publications.
42 However, glut, a five-carbon-chain dialdehyde, is used by most commercial capsule
43 producers.
44 Glut effectively cross-links gelatin-based complex coacervates and insolubilizes them
45 under conditions that rapidly and completely dissolve untreated capsules (1973). Glut
46S uptake at 4°C by acid or alkaline precursor gelatin gels ranges from 0.9 to 1.4 mM/g gela-
47N tin. Initial solids content of these gels varied from 1.4 to 5.5 wt%, significantly lower than
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 161

Microencapsulation of Flavors by Complex Coacervation 161

the 12–17 wt% initial solids content of the GGA gels treated with glut. GGA gels formed 1
from both types of gelatins had a glut uptake of 0.54–0.65 mM/g gelatin when the reaction 2
was carried out at 4°C. Glut uptake by GGA gels formed with acid precursor gelatin essen- 3
tially doubled when the reaction temperature was increased to 28°C. This increase was 4
attributed to temperature-dependent changes in the gel structure of the coacervate (1973). 5
Since a 10 wt% GA solution did not react with a significant amount of glut, glut consump- 6
tion by GGA coacervate gels is attributed to reaction with gelatin. 7
The amount of glut reacting with the gelatin in a GGA gel at acid pH generally does not 8
exceed the titratable amino content of the gelatin. Thus, glut produces a lightly cross-linked 9
gel structure. Such cross-linked gels are largely insoluble in water, but retain an ability to 10
swell in water. They are also able to absorb moisture at a relative humidity (RH) above 11
70%. Thus, the shell of glut-treated GGA and other complex coacervate capsules remains 12
sensitive to moisture. At high RH, the amount of moisture absorbed by such shells is suffi- 13
cient to plasticize them and thereby reduce their barrier properties significantly. For this 14
reason, glut-treated complex coacervate microcapsules loaded with volatile flavors are typ- 15
ically unstable at high RH. This should also be true for such capsules loaded with oxygen- 16
sensitive flavors stored in air at high RH. 17
Although glut-treated complex coacervate capsules have been approved for specific fla- 18
vor uses, the safety of capsules cross-linked with aldehydes such as glut has always been 19
open to question. For this reason, interest in an alternate ways to stabilize complex coacer- 20
vate capsule shells has existed for some time. The goal is to produce stabilized capsules that 21
are broadly accepted as food grade. One of the first alternate approaches involved posttreat- 22
ing complex coacervate capsules with aqueous tannic acid solutions. Tannic acid rapidly 23
and dramatically shrinks coacervate capsule shells, thereby reducing the water content of 24
the capsule shells and greatly increasing their ease of capsule isolation and drying. How- 25
ever, the interaction of tannic acid with gelatin is intense and rapid. This makes it difficult 26
to achieve precise control of the treatment process required in order to obtain reproducible 27
results. When capsules with a continuous core/shell structure and high oil loading are 28
treated, the effective degree of cross-linking achieved can be so intense that the capsules 29
crack and break open upon drying. Lot-to-lot variations in tannic acid properties are 30
another issue. Nevertheless, various workers continue to explore the use of tannins and nat- 31
ural phenolic compounds as cross-linking agents for coacervate gels. 32
Xing et al. (2004) reported that they successfully treated GGA capsules with tannins. 33
The GGA capsules were prepared in the presence of HEC, PVA, or PVP and subsequently 34
immersed for 10 h in pH 8–9 aqueous media that contained 2.4 w/v% tannins. The capsules 35
so treated contained upon drying 19% core material, much lower than the core content of 36
continuous core/shell capsules typically produced by complex coacervation. Further, the 37
core material was sonicated in a GA/HEC solution for 30 min. before gelatin was added to 38
the system and coacervation induced. This suggests that the capsules isolated had a multi- 39
core structure and not a continuous shell/core structure. Stresses that tannins induce when 40
they rapidly shrink a water-swollen capsule shell may have less effect on capsule stability 41
when the core material is distributed in small droplet form throughout the final dry particle. 42
Strauss and Gibson (2004) reported that treating gelatin and pectin-gelatin coacervate 43
gels with plant phenolics increased their mechanical strength and thermal stability as well 44
as reduced swelling. They interpreted their results as being consistent with a picture of 45
polyphenols reacting under oxidizing conditions with gelatin side chains to form covalent S46
cross-links. It was noted that coffee, grape juice, and various other plant materials contain N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 162

162 Chapter 7

1 enough phenolics to be effective cross-linking materials, so that isolation of the active com-
2 ponents was not necessary. The effect of plant phenolics on capsules with a gelatin-based
3 coacervate shell was not reported. The effect of lot-to-lot variability of the natural product
4 solutions on degree of effective cross-linking achieved was also not discussed.
5 Another non-aldehyde route to chemical cross-linking gelatin-based complex coacer-
6 vate capsule shells involves using transglutaminase (TG), an enzyme produced by micro-
7 bial fermentation and sold in the United States by Ajinomoto Food Ingredients, Paramus,
8 NJ. Dickenson reviewed the use of covalent cross-linking enzymes to introduce cross-links
9 as a tool for controlling the rheology and stability of protein-based foods (Dickinson,
10 1997). He noted that in 1997 TG, an extracellular product, was the only commercially
11 available cross-linking enzyme, although another enzyme, lysyl oxidase, should be of
12 interest to food technologists. TG is commercially available, because it is readily isolated
13 from the broth of fermented Streptoverticillium mobaraense. Ajinomoto literature stresses
14 that its TG functions without calcium. This is important, because many common food
15 proteins such as casein tend to precipitate at relatively low calcium ion concentrations
16 (Dickinson, 1997).
17 TG introduces cross-links between protein molecules, because it catalyzes the acyl
18 transfer reaction between the γ-carboxyamide group of a glutamine residue of a protein and
19 the ε-amino group of a lysine residue of a protein (Folk and Finlayson, 1977). The two
20 protein molecules may be different. Various Ajinomoto data sheets specify the range of pH
21 and temperature conditions under which Activa®, the trademarked name for Ajinomoto’s
22 commercial TG, will function. The optimal temperature range is 50–55°C, while the opti-
23 mal pH range is 6–7. The enzyme functions outside these ranges, although at a reduced
24 activity level. Since uncross-linked gelatin-based coacervate gels typically melt above
25 35–40°C and swell significantly at neutral pH values, TG –induced cross-linking of such
26 gels will not be carried out under optimal conditions.
27 Cho et al. (2003) used TG to crosslink capsules loaded with fish oil that were formed
28 by a double emulsion process. The capsule shell was isolated soy protein (ISP). The fish
29 oil was first emulsified in a 10% ISP aqueous solution that contained 0.025% TG. This
30 emulsion was then dispersed in corn oil that contained 3% Span 80. The resulting double
31 emulsion was warmed to 37°C and held at this temperature for 4 h. During this time, the
32 protein solution was converted to a gel.
33 Soper and Thomas (2000, 2001) disclosed a process in which TG was used to cross-link
34 the shell of capsules formed by complex coacervation. The aqueous capsule system
35 described as the example in their patents was formed by complex coacervation of gelatin by
36 a CMC–GA mixture. The aqueous capsule system produced was cooled to 5–10°C at
37 which temperature its pH was adjusted to 7.0. TG was slowly added to the system and
38 allowed to react with the capsules for 16 h at 10°C. The TG was then inactivated by lower-
39 ing the system’s pH to 2.75 with concentrated citric acid. No specific properties of the cap-
40 sules cross-linked by TG were presented.
41
42
Complex Coacervation Encapsulation Technology Issues
43
44 Complex coacervation encapsulation procedures have existed for 50 years and continue to
45 be used to produce large amounts of microcapsules for various commercial applications.
46S Nevertheless, such procedures have a number of issues. Consistent quality control of com-
47N plex coacervation encapsulation processes can be a challenging task, because final capsule
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 163

Microencapsulation of Flavors by Complex Coacervation 163

properties are very sensitive to small changes in the many variables associated with complex 1
coacervation. Since complex coacervation is a polymer phase-separation process, repro- 2
ducibility is very dependent on solution properties of the polymers involved. Lot-to-lot 3
variations in properties of the polymers used will impact process control. The degree of 4
phase separation that occurs in a specific complex coacervate encapsulation system is influ- 5
enced by the polymers involved, their MW, ionic charge density, and concentration. Varia- 6
tions in these properties affect polymer phase-separation behavior. The source, type of 7
gelatin used, acid or alkaline precursor, and bloom strength are variables that must be con- 8
trolled. Since gelatin is hydrolytically unstable, thermal conditions throughout the encapsu- 9
lation protocol must be controlled. The ratio of polymers present in the complex coacervate 10
system is another factor. System pH, temperature, and salt ion content also influence the 11
phenomenon and must be controlled. Other factors that may affect coacervation include 12
the type of acid used to adjust system pH as well as the presence of surfactants in the sys- 13
tem. Soper et al. (2000) also describe a number of factors that affect flavor encapsulation by 14
complex coacervation. 15
The core material being encapsulated is another process variable. This is particularly 16
true when the core material is a complex mixture of many components of differing polar- 17
ity. Flavors are a specific example of such types of core materials. In order to illustrate 18
this point, Table 7.4 lists experimentally determined compositions of a sample of 19
orange, lemon, and mint oils (Arneodo et al., 1988/1989). Although these flavor oils are 20
composed of a mixture of hydrocarbons, ketones, aldehydes, alcohols, esters, and other 21
unidentified components, the dominant component is limonene. The data in Table 7.5 22
(Arneodo et al., 1988/1989) establish that some of the minor components present in orange 23
and lemon oil partition into the aqueous phase. Limonene glycol, a limonene-degradation 24
product that impacts citrus oil quality, was present in the aqueous phase equilibrated with 25
both oils. 26
27
Table 7.4. Composition of orange oil, lemon oil, and mint oil flavors 28
29
Component Orange oil Lemon oil Mint oil 30
Hydrocarbons (limonene, others) 96 73 0.9 31
Ketones 0.7 21.8 33.5 32
Aldehydes 2 1.8 60.4 33
Alcohols 0.47 1.2 60.4
Esters 0.05 0.9 2.9
34
35
36
Table 7.5. Composition of orange oil, lemon oil, and mint oil that partition at 30°C and 50°C 37
(component concentration in aqueous phase, ppm)
38
Orange oil Lemon oil Mint oil 39
Component 30°C 50°C 30°C 50°C 30°C 50°C 40
41
Hydrocarbons 2
(limonene, others)
42
Ketones 185 185 43
Aldehydes 12 9 44
Alcohols 6 8 36 31 212 245 45
Esters 8 7 S46
Limonene glycol 10 12 8 5
N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 164

164 Chapter 7

1 Even though limonene is the major component of orange and lemon oils, the interfacial
2 behavior of both oils is dominated by polar components present in small amounts. The
3 interfacial tension of D-limonene against water at 50°C declines from 35 to 23 mJ/m2 over
4 5 h (Arneodo et al., 1988). These interfacial tension values are much greater than those
5 obtained with orange and lemon oils against water at 50°C (46). In contrast, the initial
6 interfacial tension value of 1-octanol against water at 50°C is 8 mJ/m2 and it declines to
7 2 mJ/m2 after 5 h. The interfacial tension of decanal against water at 50°C declines over 5 h
8 from an initial value of 10–6 mJ/m2. Although both of these compounds are present in
9 orange and lemon oils in small amounts, they have an affinity for the oil–water interface
10 and undoubtedly impact the interfacial activity of both oils.
11 Interfacial tension values of GA coacervate and supernatant phases measured at 50°C
12 against D-limonene decay from an initial value of 14–10 mJ/m2 after 5 h (Arneodo et al.,
13 1988). Since the coacervate phase is a much more concentrated polymer solution, it is sur-
14 prising that the two interfacial tension decay curves overlap as well as they do. The slope of
15 the interfacial tension decay curve decreases significantly at an interface age of 1 h after
16 which linear slow decay continues up to at least 5 h.
17 Interfacial tension values of orange and lemon oils measured against water at 25°C,
18 30°C, or 50°C decline significantly as the interface ages over a 5–10 h period (Arneodo
19 et al., 1988). Initial interfacial tension values are 4–8 mJ/m2. Values after interfacial aging
20 range from 6 mJ/m2 to a value too low to be measured by the Wilhelmy plate method.
21 The rate of interfacial tension decline is reduced but not eliminated when the system tem-
22 perature was lowered to 1.2°C. The interfacial tension behavior of both oils against the
23 supernatant phases isolated from GGA, GP, and GAlg coacervate systems was similar to
24 that observed with water. This was not surprising, because the supernatant phases are dilute
25 polymer solutions. However, it is interesting that the interfacial tension of GGA, GP, and
26 GAlg coacervate phases against orange and lemon oils is similar to that observed with
27 water. The primary difference with the coacervate phases is that the rate of interfacial ten-
28 sion decline was generally faster at the same temperature than that observed with water
29 and an interfacial tension too low to measure by the Wilhelmy plate method was observed
30 more often.
31 Interfacial tension aging at an oil/aqueous phase interface is an indication of interface
32 instability. The precise cause of this instability is difficult to define because it can be caused
33 by many factors. Furthermore, a very small amount of interfacially active material can have
34 a major effect on interfacial tension. For flavor encapsulation procedures, the possibility
35 that interfacial aging is due to one or more chemical reaction(s) at the flavor oil/water inter-
36 face is a concern. Candidate reactions in a gelatin-based complex coacervation encapsula-
37 tion system include oxidation, hydrolysis, and aldehyde condensation with any free
38 primary amino groups in the coacervate. However, interfacial aging could also be caused
39 by slow physical adsorption of interfacial active agents at the interface and slow rearrange-
40 ments of the molecules in the adsorbed layer. Although polymer solutions frequently show
41 this type of behavior, the author favors interfacial chemical reactions as the primary cause
42 of interfacial aging. This point of view is based on several experimental observations. First,
43 dispersed precipitate particles or a continuous intact film often formed at citrus oil/water
44 interfaces aged in several hours. When such systems were agitated, the aqueous phase
45 became cloudy. A second observation is the significant interfacial tension aging that both
46S oils experience against highly purified water. Gelatin and other polymers that could con-
47N tribute to prolonged interfacial aging and interfacial film formation are not initially present
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 165

Microencapsulation of Flavors by Complex Coacervation 165

in such systems. Finally, interfacial aging can be markedly reduced by reducing the temper- 1
ature of the system to 2°C. 2
Gelatin has been the dominant polycation in complex coacervation encapsulation proto- 3
cols since 1957, because it is readily available at reasonable cost and forms a gel structure 4
upon cooling, thereby setting an embryonic capsule shell. Nevertheless, the use of gelatin 5
creates a number of issues. For example, the appearance of mad cow disease raised serious 6
concerns about the safety of bovine gelatin for human consumption. This issue prompted 7
global gelatin manufacturers to support extensive studies of the possible transmission of 8
prions via gelatin consumption. These studies have concluded that commercially available 9
food grades of gelatin are not a potential source of mad cow disease transmission. Never- 10
theless, residual concern about its safety remains; if not for mad cow disease, other diseases 11
may appear in the future. Web sites posted by the Gelatin Manufacturers of Europe (GME) 12
or Gelatin Manufacturers of Asia Pacific (GMAP) provide a means of monitoring the regu- 13
latory status of various types of gelatin. 14
Another gelatin issue is the reality that food products must increasingly meet global 15
dietary requirements. This has promoted a search for materials that can replace beef and 16
pork gelatins. Fish skin gelatin is one candidate. Soper (2001) disclosed the formation of 17
capsules by coacervation of warm water fish gelatin with a CMC/GA mixture. Fish skin 18
gelatin is currently much more expensive than beef or pork gelatin but can be used. 19
Residual concern about the safety of gelatin has sparked interest in producing microcap- 20
sules by complex coacervation of other proteins. Weinbreck et al. (2003) disclose an encap- 21
sulation procedure that is based on the complex coacervation of whey protein. Although a 22
range of polyelectrolytes is claimed to be suitable for the coacervation process, GA is cited 23
as the preferred one. The preferred coacervation pH range is 2.5–4.5. The capsules pro- 24
duced can be hardened by treatment with an aldehyde or enzyme. A claimed feature of their 25
process is that it can be carried out at or below room temperatures. 26
27
28
Solvent Exchange: A Unique Property of Complex Coacervate
29
Microcapsules
30
An interesting feature of gelatin-based complex coacervate microcapsules is their ability to 31
undergo solvent exchange; that is, a water-immiscible liquid originally encapsulated within 32
a complex coacervate shell can be exchanged with a second, chemically different liquid 33
that has finite water miscibility. The exchange process occurs by diffusion through intact 34
capsule shells and broadens the range of core materials that can be incorporated in a com- 35
plex coacervate capsule. Because of solvent exchange, capsules formed by complex coac- 36
ervation can be loaded with liquids and flavors that cannot be encapsulated at the time of 37
capsule formation. 38
Figure 7.2 is a simplified flow diagram of the Brynko and Olderman solvent exchange 39
procedure (Brynko and Olderman, 1970). The first step is to prepare complex coacervate 40
microcapsules loaded with an oil that is water immiscible. For food applications, the oil 41
will be edible. Capsules to be subjected to solvent exchange must have a water-swollen 42
shell. It may be chemically cross-linked or uncross-linked. Once such capsules are avail- 43
able, they are subjected to the solvent exchange treatment. In one case, Brynko and Older- 44
man (1970) dispersed a water-wet filter cake of oil-loaded capsules in a concentrated 45
(80 w/w%) aqueous sorbitol solution for a defined time (e.g., 30 min). The purpose of this S46
treatment was to introduce into the capsule shell a finite amount of sorbitol, a compound N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 166

166 Chapter 7

1
Oil-loaded capsules
2
Shell: Water-swollen
3 Gelatin-based
4
5 Solvent exchange liquid with
finite H2O and core miscibility
6
7 Agitation for several hours
8 (exchange by diffusion)
9 Anhydrous water-miscible
10 solvent (e.g., ethanol)
11 Brief agitation
12 (extraction of water)
13
14
Dry capsules loaded with
15 Isolation and drying exchanged liquid
16
17 Figure 7.2. Simplified flow diagram of solvent exchange coacervation procedure (from Brynko
18 and Olderman, 1970, with permission).
19
20
21 designed to plasticize the capsule shell. After excess sorbitol solution was removed from
22 the capsule slurry, typically by filtration, a finite volume of ethanol was added to the sys-
23 tem. Food-flavoring agents may be dissolved in the ethanol. After a finite diffusion or sol-
24 vent exchange time (e.g., 30 min), excess ethanol solution may be removed from the system
25 by decantation or filtration and replaced with a fresh ethanol solution. This cycle is
26 repeated until the desired extent of solvent exchange is achieved. Brynko and Olderman
27 (1970) completed their solvent exchange process by immersing the capsules in excess
28 anhydrous ethanol free of solute. The objective is to remove the last remaining traces of
29 water from the capsule shell, thereby sealing them. After the capsules are dried, they form a
30 free-flow powder.
31 Brynko and Olderman (1970) teach that the shell of a complex coacervate capsule must
32 be water-swollen in order to successfully undergo solvent exchange. The water may be
33 present either because the capsule was never dried after formation or because water was
34 added after the capsule had been dried. Water is essential. It causes complex coacervate
35 shells to swell and become porous to liquids that have finite water miscibility. This enables
36 the diffusion of such liquids into the interior of capsules. Since solvent exchange is a diffu-
37 sion process, a finite time is required. Solvent exchange is a diffusion-controlled equilibra-
38 tion or partitioning process, so complete removal of the oil originally carried by a capsule is
39 difficult to achieve. A small but finite amount of oil is typically left in a capsule after sol-
40 vent exchange is deemed complete. In some cases, a water-swollen coacervate capsule
41 shell acts as a semi-permeable membrane that allows diffusion of the solvent exchange liq-
42 uid into the capsule, but not diffusion of the oil out. The resulting increase in liquid content
43 of the capsule may be so significant that the capsules visibly increase in diameter.
44 Although water must be present in the shell of a complex coacervate when solvent
45 exchange is carried out, its presence in the capsule shell after completion of the solvent
46S exchange process is detrimental to prolonged storage stability. Brynko and Olderman
47N (1970) correlated retention of a solvent exchange with its dielectric constant. They report
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 167

Microencapsulation of Flavors by Complex Coacervation 167

that single liquids with finite water miscibility and a dielectric constant below 20 yield 1
stable capsule powders, whereas liquids with a dielectric constant above 20 do not. Since 2
the dielectric constant of ethanol, a desirable exchange solvent, is 24.3, ethanol-loaded cap- 3
sules prepared by solvent exchange are unstable. The lower the dielectric constant of the 4
exchange liquid, the more stable the capsules. Mixtures of dielectric solvent liquids with 5
low dielectric constants can be used as long as the dielectric constant of the mixture is <20. 6
Thus, stable capsules that contain a finite concentration of ethanol can be prepared as long 7
as the other solvents present in the capsule have a low dielectric constant and the solvent 8
mixture has a dielectric constant below 20, preferably below 14. 9
The instability of capsules loaded with a high dielectric constant solvent is attributed to 10
their ability to readily absorb water from the atmosphere in which the capsules are stored. 11
The absorbed water plasticizes the capsule shell, thereby facilitating rapid solvent release. 12
Brynko and Olderman (1970) note that capsules isolated after solvent exchange with butyl 13
acetate contain 0.01% water. These have excellent storage stability, because the dielectric 14
constant of butyl acetate is 5 and butyl acetate does not promote water absorption from the 15
atmosphere under typical conditions (e.g., RH <70%). Although Soper et al. (2000a, b) also 16
disclose a solvent exchange procedure for preformed capsules. In one example, capsules 17
loaded with purified vegetable oil were formed by the complex coacervation of gelatin with 18
a mixture of GA and sodium CMC. Both chemically cross-linked and uncross-linked cap- 19
sules were used. As noted by Brynko and Olderman (1970), Soper et al. (2000a, b) require 20
the shell of capsules that are candidates for solvent exchange to be water swollen. Thus, 21
capsules that were dried after formation were placed for 5 min in a flavor-water mixture. 22
They were subsequently transferred to a closed plastic container and incubated for 24 h 23
before use. The inventors note that the shell of capsules subjected to their solvent exchange 24
procedure can be treated in order to prevent removal of flavor from microcapsules and 25
water removal from the shells was specifically cited. The importance of residual water to 26
dry capsule stability was not referenced, but it is reasonable to suggest that high humidity 27
will affect storage stability of capsules subjected to solvent exchange, as disclosed by 28
Soper et al. (2000) process. Soper et al. (2000, 2001) do not discuss the need for the solvent 29
exchange system to remain in one phase throughout the solvent exchange process nor the 30
effect of the exchange solvent’s dielectric constant on solvent exchange. 31
Soper et al. (2000) report that capsules with water-swollen shells can absorb flavors 32
from the gas phase. They placed oil-loaded capsules having a water-swollen shell in a con- 33
tainer and purged them with a loaded gaseous phase that contained an aroma. After a finite 34
purging time, 0.5–5 h depending on the aroma, the aroma absorbed by the capsules could 35
be detected. A variety of applications were cited, including the separation and concentra- 36
tion of volatile hydrophilic aroma components from hydrophobic components. 37
38
39
Summary
40
Complex coacervation encapsulation technology is versatile and adaptable. Capsules pro- 41
duced by this technology have many interesting features, which continue to attract interest 42
in them. Soper (1995) discusses applications of flavors encapsulated by complex coacerva- 43
tion. Such capsules are prime candidates for flavor oil encapsulation. They offer food tech- 44
nologists a high degree of versatility and flexibility. Microcapsules with sizes ranging from 45
a few microns to over a millimeter in diameter can be produced. Such capsules typically S46
carry a flavor payload of 60–90 wt%, although lower and higher payloads can be produced. N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 168

168 Chapter 7

1 The capsules can be supplied as an aqueous slurry or dry powder. Drying is typically done
2 in a fluidized bed or spray drier. Capsules supplied as aqueous slurries are able to withstand
3 finite handling or processing stresses, because water-swollen complex coacervate shells are
4 rubbery and resist failure. Such encapsulation protocols are not free of limitations or issues
5 including: component partitioning during capsule manufacture, moisture sensitivity during
6 storage, and reaction of aldehyde components of flavors with gelatin-containing shells on
7 storage. Nevertheless, the technology continues to diversify and expand. Because many of
8 its characteristic properties remain uncharacterized and unexploited, it is a fruitful area for
9 further study.
10
11
12 References
13 Arneodo, C., Baszkin, A., Benoit, J.-P. and Thies, C. 1988/1989. Interfacial studies of essential oil-water systems.
14 Colloids and Surfaces 34: 159–169.
15 Arneodo, C., Baszkin, A., Benoit, J.-P. and Thies, C. 1988a. Interfacial tension behavior of citrus oil components,
16 Absts. 6th International Conference on Surface and Colloid Science, Hakone, Japan, p. 106.
17 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,
18 eds., American Chemical Society, Washington, DC, Chapter 15.
19 Brenner, J. 1983. The essence of spray dried flavors: the state of the art. Perfumer and Flavorist 8: 40–44.
20 Brynko, C. and Olderman, G.M. 1970. Replacement of capsule contents by diffusion, US Patent 3,516,943 (June
21 23, 1970).
22 Bungenberg de Jong, H.G. 1949. Complex colloidal systems, In Colloid Science, Vol. II, H.R. Kruyt, ed., Elsevier,
Chapter 10.
23 Burgess, D.J. 1990. Practical analysis of complex coacervate systems. J. Colloid and Interface Sci. 140: 227–238.
24 Chilvers, G.R. and Morris, V.J. 1987. Coacervation of gelatin-gellan gum mixtures and their use of microencapsu-
25 lation. Carbohydrate Polymers 7: 111–120.
26 Cho, Y.-H., Shim, H.K. and Park, J. 2003. Encapsulation of fish oil by an enzymatic gelation process using transg-
27 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-
28 vates. Proceed. Intern. Symp. Control. Rel. Bioact. Mater. 16: 279–280.
29 Duquemin, S.-J. and Nixon, J.R. 1985. The effect of sodium lauryl sulphate, cetrimide and polysorbate 20 surfac-
30 tants on complex coacervate volume and droplet size. J. Pharm. Pharmacol. 37: 698–702.
31 Duquemin, S.-J. and Nixon, J.R. 1986. The effect of surfactants on the microencapsulation and release of pheno-
32 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.
33 Trends in Food Science and Technology 8: 34–339.
34 Folk, J.E. and Finlayson, J.S. 1977. The ε-(γ-glutamyl)lysine crosslink and the catalytic role of transglutaminase.
35 Adv. Protein. Chem. 31: 1–133.
36 Goubet, I., Le Quere, J.L. and Voilley, A.J. 1998. Retention of aroma compounds by carbohydrates: influ-
37 ence of their physicochemical characteristics and their physical state. A review. J. Agric. Food Chem. 46:
1981–1990.
38 Green, B.K. and Schleicher, L. 1957. Oil-containing microscopic capsules and method for making them, US
39 Patent 2,800,457, July 23.
40 Jegat, C., Taverdet, J.L. 2000. Stirring speed influence study on the microencapsulation process and on the drug
41 release from microcapsules. Polymer Bulletin 44, 345–351.
42 Jizomoto, H. 1984. Phase separation induced in gelatin-base coacervation systems by addition of water-soluble
nonionic polymers I: Microencapsulation. J. Pharm Sci. 73: 879–882.
43 Kerkof, P.J. and Thijssen, H.A. 1974. Retention of aroma components in extractive drying of aqueous carbohy-
44 drate solutions. J. Food Technol. 9: 415–423.
45 Kim, J.-C., Song, M.-E., Lee, E.-J., Park, S.-K., Rang, M.-J. and Ahn, H.-J. 2001. Preparation and characterization
46S of triclosan-containing microcapsules by complex coacervation. J. Dispersion Science and Technology 22:
47N 591–596.
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 169

Microencapsulation of Flavors by Complex Coacervation 169

Koh, G.-L. and Tucker, I.G. 1988a. Characterization of sodium carboxymethylcellulose-gelatin complex coacer- 1
vation by viscosity, turbidity and coacervate wet weight and volume measurements. J. Pharm. Pharmacol. 40: 2
233–236.
Koh, G.-L. and Tucker, I.G. 1988b. Characterization of sodium carboxymethylcellulose-gelatin complex coacer-
3
vation by chemical analysis of the coacervate and equilibrium fluid phases. J. Pharm. Pharmacol. 40: 309–312. 4
Leuenberger, B.H. 1991. Investigation of viscosity and gelatin properties of different mammalian and fish 5
gelatins. Food Hydrocolloids 5: 353–361. 6
Liu, X.-D., Atarashi, T., Furuta, T., Yoshii, H., Aishima, S., Ohkawara, M. and Linko, P. 2001. Microencapsulation 7
of 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 coacervate
8
capsules. J. Pharm Sci. 53: 429–431. 9
Mayya, K.S., Bhattacharyya, A. and Argillier, J.-F. 2003. Micro-encapsulation by complex coacervation: influence 10
of surfactant. Polym. Int. 52: 644–647. 11
McMullen, J.N., Newton, D.W. and Becker, C.H. 1984. Pectin-gelatin complex coacervates II: effect of microen- 12
capsulated sulfamerazine on size, morphology, recovery, and extraction of water-dispersible microglobules.
J. Pharm. Sci. 73: 1799–1803.
13
Menting, L.C. and Hoogstad, B. 1967. Volatiles retention during the drying of aqueous carbohydrate solution. 14
J. Food Sci. 32: 87–90. 15
Re, M.I. 1998. Microencapsulation by spray drying. Drying Technology 16: 1195–1236. 16
Reineccius, G. 2004. The spray drying of food flavors. Drying Technology 22: 1289–1324. 17
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.
18
Rulkans, W.H. and Thijssen, H.A. 1978. Retention of organic volatiles in spray-drying aqueous carbohydrate 19
solution. J. Food Technol. 7: 95. 20
Saeki, K. and Hosoi, N. 1984. Microencapsulation by a complex coacervation process using acid-precursor 21
gelatin. Appl. Biochem. Biotechnol. 10: 251–254. 22
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.
23
Soper, J.C. 1995. Utilization of coacervated flavors, In Encapsulation and Controlled Release of Food Ingredients, 24
S.J. Risch and G.A. Reineccius, eds., American Chemical Society, Washington, DC. 25
Soper, J.C. 1997. Method of encapsulating food or flavor particles using warm water fish gelatin and capsules pro- 26
duced therefrom. US Patent 5,603,952 (February 18, 1997). 27
Soper, J.C. and Thomas, M.T. 2000. Enzymatically protein-encapsulating oil particles by complex coacervation.
US Patent 6,039,901 (March 21, 2000).
28
Soper, J.C. and Thomas, M.T. 2001. Enzymatically protein-encapsulating oil particles by complex coacervation. 29
US Patent 6,325,951 (December 4, 2001). 30
Soper, J.C., Kim, Y.D. and Thomas, M.T. 2000a. Method of encapsulating flavors and fragrances by controlled 31
water transport into microcapsules. US Patent 6045835 (April 4, 2000). 32
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).
33
Strauss, G. and Gibson, S.M. 2004. Plant phenolics as cross-linkers of gelatin gels and gelatin-based coacervates 34
for use as food ingredients. Food Hydrocolloids 18: 81–89. 35
Thies, C. 1973. The reaction of gelatin-gum Arabic coacervate gels with glutaraldehyde. J. Colloid and Interface 36
Sci. 44: 133–141. 37
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.
38
Thijssen, H.A.C. 1975. In Freeze Drying and Advanced Food Technology, S.A. Goldblith, L. Rey and W.W. 39
Rothmayer, eds., Academic Press, London. 40
Veis, A. 1970. Phase equilibria in systems of interacting polyelectrolytes, In Biological Polyelectrolytes, A. Veis, 41
ed., Marcel Dekker, NY. 42
Veis, A. and Aranyi, C. 1960. Phase separation in polyelectrolye systems. I. Complex coacervates of gelatin 64:
1203–1210.
43
Venkataram, J.S. 1995. Indomethacon sustained release from alginate-gelatin or pectin-gelatin coacervates. Int. J. 44
Pharm. 126: 161–168. 45
Vinietsky, Y. and Magdassi, S. 1997. Microencapsulation by surfactant-gelatin insoluble complex: effect of pH S46
and surfactant concentration. J. Colloid and Interface Sci. 189: 83–91. N47
Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 170

170 Chapter 7

1 Weinbreck, F., De Kruif, C. and Schrooyen, P. 2003. Complex coacervates containing whey proteins. WO
2 03/106014 A1 (December 24, 2003).
Xing, F., Cheng, G., Yang, B. and Ma, L. 2004. Microencapsulation of capsaicin by the complex coacervation of
3 gelatin, acacia and tannins. J. Applied Poly. Sci. 91: 2669–2675.
4 Yan, N. 2005. Encapsulated agglomeration of microcapsules and method for the preparation thereof. US Patent
5 6974592 (December 13, 2005).
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S
47N
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 171

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

8 Confectionery Products as Delivery Systems 1


2
for Flavors, Health, and Oral-Care Actives 3
4
5
Jamileh M. Lakkis 6
7
8
9
10
11
Introduction
12
Despite consumers apprehension about sugar-based products, the confectionery market is 13
currently experiencing record growth. Euromonitor International has estimated the global 14
confectionery market at more than $142 billion in 2005. This surge is due to two main fac- 15
tors: first, the availability of sugar-free versions of traditional sugared products and second, 16
the new trend in formulating confectionery products with functional actives that can deliver 17
unique health benefits. 18
The last two decades have witnessed an intense effort to shift the market positioning of 19
confectionery products from just pleasantly tasting sweet snacks to a platform for delivering 20
nutraceuticals, breath fresheners, nicotine, antimicrobial and dental health agents as well as 21
drug actives. The latter category—including analgesics, insulin, antibiotics, and other phar- 22
maceutical ingredients, although outside the scope of this book—will be referred to help 23
explain the mechanisms of delivery and absorption. 24
Functional confections are currently enjoying an unprecedented acceptability from con- 25
sumers trying to increase their intake of functional and health-promoting ingredients such 26
as vitamins, minerals, herbal extracts, etc. in a familiar food format, which does not signal ill- 27
ness and can be consumed discreetly. Successful examples of this category include Viactive® 28
from McNeil, a calcium- and vitamin-containing chewy candy formulated for women, Orbit® 29
chewing gum from Wrigley’s that claims teeth-cleaning benefits, mentholated lozenges such 30
as Vicks® and Robitussin® that claim throat relief or nasal decongestion, Listerine Pocket- 31
paks® from Pfizer, and Nicorette®, smoke-cessation chewing gum from GlaxoSmithkline. 32
The first patent on functional confectionery products was granted to W.F. Semple in the 33
nineteenth century for developing a dentifrice in the form of a chewing gum (Semple, 1869). 34
However, the first commercial product claiming delivery of functional ingredients was a sal- 35
icylic acid-containing chewing gum, Aspergum®, which was marketed in the United States 36
in 1924 and is still available today. A breakthrough in utilizing chewing gums as delivery 37
systems was documented in the clinical finding that smokers may be able to give up smoking 38
by self-titrating the amount of nicotine they absorb (Fernö, 1973; Mulry, 1988; Silagy et al., 39
2002). This finding was the basis for the development and marketing of nicotine-containing 40
chewing gums, where subjects can chew the gum to release and absorb the needed amounts 41
of nicotine (Benowitz et al., 1987; Mulry, 1988). 42
Despite these advances, the challenge for true commercial success of functional confec- 43
tions lies mainly in the inability of some formats to deliver therapeutic levels of health 44
actives and the harsh conditions in the stomach that can sometimes degrade the active before 45
it had the chance to reach its target site such as lower intestines or the blood stream. Unlike S46
flavor delivery, where the only requirement is dissolution of the active from the dosage into N47

171
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 172

172 Chapter 8

1 the saliva and its extended release for a desired period of time, delivering therapeutic actives
2 requires more elaborate capsule or delivery system design and an understanding of the phys-
3 iology of absorption across membranes. Actives incorporated into a confectionery product
4 must be transported from the dosage carrier to specialized tissues and epithelia and eventu-
5 ally to the target site in the blood stream or the cytoplasm of a particular cell group.
6 Two types of oral delivery routes can be distinguished; these are local (target release site
7 is the mouth or throat areas) and systemic (blood stream or specific organ or cell). In design-
8 ing delivery systems, it is imperative to take into consideration not only the physiology and
9 organizational structure of the oral cavity but also the physicochemical properties of the
10 delivery system including dose concentration, format, residence time in the mouth, etc.
11
12
Physiology and Organization of the Oral Area
13
14 Organs that constitute the oral area include the mouth, tongue, and esophagus (Figure 8.1).
15 Within these organs, several regions can be differentiated that are critical for permeability
16 and absorption (Squier et al., 1976). The mouth extends from the lips to the oropharynx at
17 the rear; its temperature and humidity vary greatly during normal activities such as drinking
18 and eating, thus impacting the active’s dissolution and its absorption. The oral cavity can be
19 divided into two main regions (Figure 8.2), namely:
20
21 1. The oral cavity proper consisting of the tongue, hard and soft palates, and floor of the
22 mouth.
23 2. The outer vestibule consisting of cheeks (buccal mucosa), maxillary (upper jaw), and
24 mandibular (lower jaw) areas.
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46S
47N Figure 8.1. Views of the oral cavity and pharynx.
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 173

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 173

In humans, the tongue is essential for several processes including moving the food bolus 1
around in the mouth, chewing, speech, sucking, and swallowing. The latter is achieved by 2
virtue of the negative pressure created within the oral cavity. The tongue consists of a mass 3
of interwoven, striated muscles interspersed with glands and fat and covered with mucus 4
membrane tissues that are responsible for secreting small amounts of mucus. The tongue 5
surface contains papillae, which are sensitive to food flavors along with several ridges that 6
help grip the food article while the tongue agitates it during chewing. 7
The tongue is a highly sensitive well-coordinated organ that occupies the middle of the 8
mouth; therefore any device placed in the oral cavity should take this into consideration. 9
The sublingual area moves extensively during eating, drinking, and speaking, thus impact- 10
ing the residence time of food bolus or any delivery device placed in the oral cavity (Collins 11
and Dawes, 1987). The inferior portion of the tongue (under surface leading from the tip of 12
the tongue to the floor of the mouth) contains mucus membranes and is smooth and purple 13
in color due to the many blood vessels present. The root contains bundles of nerves, arter- 14
ies, and muscles that branch to the other regions. Nerves from the tongue receive chemical 15
stimulation from food in solution which gives the sensation of taste. 16
The esophagus is a muscular tube that connects the pharynx to the stomach. It is approxi- 17
mately 25-cm long and about 2-cm in diameter. Similar to the buccal area, the esophagus is 18
lined with stratified squamous epithelium lining whereas the very remote portion (toward the 19
stomach) is lined with columnar epithelia, which are highly specialized for absorption. The 20
main role of the esophagus is to move ingested materials from the mouth area to the stomach 21
and lower gastrointestinal tract (GIT). The esophageal epithelial area is non-keratinized and 22
23
24
25
26
Upper lip
27
Underside
28
of tongue 29
Alveolar mucosa 30
Hard palate
31
Gingiva
32
Soft palate 33
34
Cheek Floor of mouth
35
36
37
38
Tongue
Lower lip 39
40
Masticatory mucosa
41
42
Lining mucosa 43
Specialized mucosa 44
45
Figure 8.2. Anatomical location and extent of masticatory, lining and specialized mucosa in the S46
oral cavity (Squier and Kremer, 2001 with permission). N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 174

174 Chapter 8

1 is lined with mucus-secreting glands that help keep the esophagus moist and protect it from
2 gastric acidity. Typical food transit time in the esophagus is very short (10–14 seconds).
3 One peripheral system, the trigeminal nerve, is responsible for specialized sensations
4 and constitutes an important part of the oral cavity. Its function resembles that of the spinal
5 nerves, which are responsible for the sensation in the rest of the body. The trigeminal nerve
6 is a cranial nerve comprised of three major branches: the ophthalmic nerve, the maxillary
7 nerve, and the mandibular nerve. The sensory function of the trigeminal nerve is to provide
8 conscious awareness of the face and mouth. The maxillary nerve carries sensory informa-
9 tion mainly from the cheek, upper lip, upper teeth and gum, palate and roof of the pharynx.
10 Mandibular nerve carries sensory information from the lower lip, lower teeth and gum, and
11 floor of the mouth. The mandibular nerve carries touch/position and pain/temperature sen-
12 sations from the mouth but not taste sensations. Unlike touch/position input that takes place
13 immediately, pain/temperature sensation experiences a perceptible delay due to the
14 unmyelinated slow-conducting nerve fibers. This type of sensation is mostly caused by a
15 specific group of chemicals commonly referred to as “sensates” and which include sub-
16 stances that induce cooling, warming, tingle, and similar effects. Sensates have been used
17 in confectionery formulations to provide perception of refreshment (cooling, tingle) or
18 soothing (warming) and calming sensations.
19
20
Permeability and Barrier Functions of the Oral Cavity
21
22 Permeability and barrier selectivity of the oral cavity are complex phenomena. A better appre-
23 ciation for these functions can be gained by understanding the structure and critical functions
24 of tissues, salivary glands and their secretions as well as their interactions (Rojanasakul et al.,
25 1992). Table 8.1 shows variations in thickness of the oral mucosa in various regions of the
26 human oral cavity. However, mucosal thickness does not explain variations in permeability in
27 various regions of oral cavity.
28
29
Physiological and Structural Basis of Transport Routes
30
(Plasma and Epithelial Membranes)
31
32 Plasma Membranes
33
Plasma membranes retain the contents of the cell and act as permeability barriers. They
34
allow only certain substances to enter or leave the cell, though the rate of entry is strictly
35
controlled. Hydrophobic materials enter the cells easily due to the presence of a lipoidal
36
layer at the cell surface, commonly known as the bilayered lipid membrane, with bands
37
38
Table 8.1. Thickness of various regions
39
of the human oral cavity (Robinson,
40 2000 with permission)
41
42 Region (microns) Thickness
43 Skin 100
44 Hard palate 250
45 Attached gingival 200
46S Buccal mucosa 200–600
Floor of mouth 100–200
47N
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 175

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 175

approximately 3 nm in width and an overall thickness of between 8 and 12 nm (Curatolo, 1


1987). Plasma membranes are highly organized structures, where proteins in specific con- 2
formations act as structural elements, transport nutrients, and sample the cell environment. 3
Lipid-soluble substances tend to diffuse along the plasma membranes of the cells while 4
water can flow through transcellular routes by virtue of the small polar channels through 5
these membranes. 6
7
8
Epithelial Membranes
9
Most internal and external body surfaces are covered with epithelia, which contain a layer of 10
basal lamina and structural collagen underlying layers of epithelial cells. There are several 11
morphologically distinct epithelial types, namely, simple squamous (line blood vessels), 12
simple columnar (line stomach and small intestines), and stratified squamous epithelium 13
(line mouth and esophagus). 14
The epithelium has a vertical dimension of 600 microns through the epithelial ridges 15
and 250 microns through the areas overlying the connective tissue papillae. The buccal 16
epithelium possesses some net charge, hence its permeability and selective ion transport. 17
Assessing the role of epithelial layers can better be understood by differentiating between 18
two criteria, namely permeability and permselectivity. The former refers to permeation 19
magnitude (as quantified by electrical resistance), while the latter describes its qualitative 20
ability to show preference for cations or anions or within a series of cations and anions 21
(Fromter and Diamond, 1972; MacKnight and Leader, 1983). 22
Epithelia of the epidermis, hard palate, and gingivae are keratinized and are known to 23
be not very permeable to water. Earlier studies showed that these keratinized epithelia con- 24
tain neutral lipids such as acylceramides and ceramides, which have been associated with a 25
barrier function (Wertz and Downing, 1983). Epithelia of the soft palate, sublingual, and buc- 26
cal area as well as those located in the floor of the mouth are nonkeratinized and have shown 27
significant permeability to water presumably due to the absence of acylceramides (Squier and 28
Hall, 1985). 29
30
31
Oral Mucosa
32
The oral mucosa represents one type of epithelial membranes that secretes mucus 33
(Figure 8.3). Similar to the skin and intestinal mucosa, oral mucosa mainly protects the oral 34
cavity from harmful substances as well as facilitates absorption of chemical entities. The oral 35
mucosa plays a protective role during mastication, which involves compression and shear 36
forces. Areas such as the hard palate and attached gingivae have a textured surface to resist 37
abrasion and are tightly bound to the underlying bone to resist shear forces. The cheek 38
mucosa is elastic to allow for distension. Like the skin, the human oral mucosa consists of 39
stratified squamous epithelia. However, unlike the skin, it is always maintained moist 40
because of the presence of numerous salivary glands and does not show the presence of 41
keratin. These dissimilarities make the oral mucosa more permeable than the skin (Chen 42
and Squier, 1984; Gandhi and Robinson, 1988; Squier and Wertz, 1996). 43
The mucosa of the human mouth is permeable to various vitamins such as thiamine, 44
ascorbic acid and nicotinic acid. Several investigations have shown that the absorption 45
characteristics of the oral mucosa were broadly similar to those of the small intestine of a S46
rat (Evered and Mallett, 1983; Evered et al., 1980). N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 176

176 Chapter 8

1
2
3
4
5 Epithelium
6
7
8
9
10 Lamina
11
12
13
14
15
16 Submucosa
17
18
19
20
21
Figure 8.3. Structure of the oral mucosa (Harris and Robinson, 1992 with permission).
22
23
24
25
Saliva
26
27 Saliva is a mucus, viscous, colorless fluid that originates in the buccal and sublingual glands.
28 It is a unique fluid that plays a significant role in controlling absorption and bioavailability of
29 ingested actives both as an enhancer and as a barrier to permeability. Saliva forms a thin film
30 (0.07–0.10 mm) of hypotonic nature (110–220 milliosmoles/lit) that lubricates and moistens
31 the inside of the mouth. Saliva is believed to play a significant role in repairing injuries and
32 tears in the oral area due to the abundance of hyaluronic acid molecules. pH of human saliva
33 ranges from 7.4 to 6.2 depending on its flow rate (low to high flow rates). Certain foods such
34 as carbohydrates, due to bacterial action, can reduce saliva pH to 3–4.
35 Saliva is primarily composed of water, mucus, proteins, glycoproteins, mineral salts, and
36 amylases. The composition of the saliva depends on the rate at which different cell types con-
37 tribute to the final secretion: mucus secretion (due to the glycoprotein and mucin) and watery
38 secretion (containing salivary amylase). The major ions are sodium, potassium, chloride, and
39 bicarbonates (Weatherell et al., 1994). In the ducts of the salivary glands, sodium and chlo-
40 ride are reabsorbed but potassium and bicarbonates are secreted, thus, the electrolyte balance
41 is altered depending on the rate of salivary flow. Other salivary enzymes include ptyalin,
42 lingual amylases and so on (Chauncey et al., 1957; Lindqvist and Augustinsson, 1975;
43 Tan, 1976).
44 In order to be absorbed orally, the active must first dissolve in the saliva. Extremely
45 hydrophobic materials do not dissolve well and are likely to be swallowed intact unless a
46S specialized delivery system is used to present them to the mucosa. Saliva containing dis-
47N solved actives is constantly being swallowed, thus competing with buccal absorption.
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 177

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 177

Keratinization 1
2
Barrier function of the surface layers of the buccal epithelium depends on the intercellular 3
lipid composition. Epithelia that contain polar lipids notably cholesterol sulfate and gluco- 4
syl ceramides are considerably more permeable to water than keratinized epithelia. Intra- 5
cellular lamellae composed of chemically nonreactive lipids have been identified in 6
the human buccal mucosa and may be relevant to drug permeability. There are intercellular 7
barriers in the superficial layers of both keratinized and nonkeratinized oral epithelia 8
that can limit the penetration of substances traversing the tissue by this route, espe- 9
cially polar molecules and electrolytes. Substances with a preferential solubility are 10
more likely to pass along membranes and these may be limited by the formation of a ker- 11
atin layer. 12
13
14
Membrane Coating Granules 15
Membrane coating granules (MCGs) are spherical or oval organelles of about 100–300 nm 16
in diameter found in many stratified epithelia and are believed to form major permeability 17
barriers. MCGs appear to play a major filtration barrier role in the kidneys (Kanwar et al., 18
1980) by delaying or preventing the movement of large molecules such as proteins. They 19
have also been found in both keratinized (gingivae) and nonkeratinized (buccal) epithelia 20
(Hayward, 1979). 21
These granules contain glycoproteins, formed by covalent linkage between glyco- 22
saminoglycans, mucopolysaccharide, and proteins. The glycosaminoglycans are high- 23
molecular weight linear molecules with complex sequence. MCGs are also negatively 24
charged molecules (abundant in sulfate and carboxyl groups). The glycosaminoglycan 25
molecule occupies a much larger volume than other molecules with comparable size. These 26
characteristics make the glycosaminoglycan molecule an effective diffusional barrier in 27
particular against electrolytes and water in extracellular fluids. 28
29
30
31
Polarity
32
Permeability routes across the oral mucosa can be classified into nonpolar and polar: (i) the 33
non-polar route involves lipid elements of the mucosa, which partition the active 34
into the lipid bilayer of the plasma membrane or into the lipid of the intercellular matrix; 35
and (ii) the polar route involves passage of hydrophilic materials through aqueous pores 36
in the plasma membranes of individual epithelial cells or ionic channels in the intercel- 37
lular spaces of the epithelium. Whether a given nonelectrolyte will pass rapidly across 38
the oral mucosa is determined by its partitioning between lipid and aqueous phases 39
(Schanker, 1964). Substances with high lipid solubility will be transported across the lipid- 40
rich plasma membranes of the epithelial cells while water-soluble substances will pass 41
through the intercellular spaces. 42
An alternative classification involves passage through intercellular spaces between cells 43
(i.e. the paracellular route) or transport into and across the cells (i.e. the transcellular route). 44
The latter involves partitioning, cellular channel diffusion, and carrier-mediated transport 45
(Blanchette et al., 2004). The paracellular route represents diffusive convective transport S46
occurring through the intercellular space (Figure 8.4). N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 178

178 Chapter 8

1
2
3
4
5
6
7
8
9
10
11
12 Figure 8.4. The four mechanisms of transport across a cell monolayer (Blanchette et al., 2004
13 with permission).
14
15
16 pH
17
18 The buccal epithelium has an isoelectric point (pH at which potential is zero) of 2.6.
19 At neutral pH, the buccal epithelial membrane is negatively charged, relatively imperme-
20 able to anions and therefore functions as an ion-exchange surface for cations. At acidic pH
21 values (i.e. below the isoelectric point), the membrane carries a net positive charge and
22 becomes relatively impermeable to cations and functions as an anion exchanger.
23 Although diffusion potential experiments have shown a higher relative permeability of
24 potassium cation (K) over the chloride anion (Cl), information on the absolute permeabil-
25 ities of these ions is lacking (Kaber, 1974; Lesch et al., 1989). As ionic strength increases,
26 resistance decreases due to increased electrostatic shielding and therefore lower electrostatic
27 potential barrier to permeation of ions and a reduction in membrane resistance.
28
29
Transport Mechanisms across Membranes
30
31 Drugs and active components, except when given intravenously, must be transported across
32 several biological barriers before reaching general circulation. Four transport mechanisms
33 are known, namely: simple (passive) diffusion, facilitated diffusion, active transport, and
34 pinocytosis. It is generally believed that most substances passing across the oral mucosa
35 move by simple Fickian diffusion (Siegel et al., 1971). Only qualitative evidence of facili-
36 tated diffusion for small substances has been reported (Siegel, 1984). The oral mucosa
37 employs an active uptake mechanism for a very few number of small molecules such as
38 monosaccharides (Manning and Evered, 1976). In buccal epithelia, passive diffusion is,
39 likely, the most frequent mechanism.
40
41 1. Passive diffusion is the transport across the cell membrane wherein the driving force for
42 the movement is the concentration gradient of the solute. In orally administered actives,
43 this absorption occurs in the small intestines.
44 2. Facilitated diffusion can best be described by the movement of molecules from a higher
45 concentration to a lower one as a result of their random motion. Depending on the physi-
46S cal or chemical properties of the active, diffusion across biological membranes can take
47N place through a lipid phase or along aqueous channels. In either situation, provided that
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 179

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 179

an adequate concentration of the active is applied to one side of a membrane and there is 1
sufficiently rapid removal of it from the other side, then a steady state is reached in which 2
the rate of diffusion is directly proportional to the concentration of the active (this is 3
known as Fick’s law). 4
3. Active transport involves the movement of molecules against a concentration gradient or 5
of ions against an electrochemical gradient and requires the expenditure of metabolic 6
energy. Some sugars and amino acids are transported across intestinal epithelia but are 7
unlikely to take place across skin or oral mucosa (Kaaber, 1973). 8
4. Endocytosis is a process by which a large number of different cell types are capable of 9
taking up solid particles (phagocytosis) or fluids (pinocytosis) from their external envi- 10
ronment by engulfing the material in membranous vesicles. While cells of the oral epithe- 11
lium are capable of taking up material by endocytosis, particularly in the basal and 12
prickle layers, it does not seem a likely transport mechanism across an entire stratified 13
epithelium (Berridge and Oschman, 1972). 14
15
16
Effect of Dosage Position in the Mouth
17
Within the oral cavity, delivery of drugs can be classified into four categories: 18
19
1. Sublingual delivery, in which the dosage form is placed on the floor of the mouth under 20
the tongue. 21
2. Buccal delivery, in which the formulation is positioned against the mucus membranes 22
lining the cheeks. 23
3. Local oropharyngeal delivery, where the delivery vehicle is positioned to treat the mouth 24
and throat. 25
4. Periodontal delivery to treat below the gum margin. 26
27
It has been suggested that drug absorption through the sublingual mucosa is more effective 28
than through the buccal mucosa, even though both these regions are nonkeratinized. The 29
sublingual epithelium is, however, thinner and immersed in saliva, both of which aid absorp- 30
tion (Altman et al., 1960). 31
When fluoride tablets were placed in the lower mandibular sulcus, fluoride concentra- 32
tions were found to increase significantly in the region of the tablet, but there was no appre- 33
ciable increase in salivary levels. In addition, relatively small amounts of fluoride had 34
migrated to the opposite side of the mouth suggesting that the lower mandibular sulci are 35
quite isolated from the remainder of the mouth. However, when the tablet was placed in the 36
upper sulcus, the fluoride migrated some distance from the site of administration (Weath- 37
erell et al., 1984). Glucose was also found to behave in a similar fashion (Weatherell et al., 38
1989). It may be concluded that the site-specific differences are due to saliva movement and 39
dilution of the test substance rather than the nature of the substance. Thickness of the sali- 40
vary film will vary from place to place depending upon the proximity to the ducts of the 41
major and minor salivary glands, separation of mucosal layers during speaking and mouth 42
breathing. 43
Weatherell et al. (1989) reported that glucose retention in the oral cavity was least under 44
the tongue presumably due to (i) dilution and flushing by saliva, (ii) mechanical action 45
of the tongue, and (iii) tendency for some glucose to disappear by absorption through the S46
sublingual mucosa or in certain other areas by metabolism in the plaque. Despite the N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 180

180 Chapter 8

1 widespread use of the buccal absorption test, which expresses results as average “buccal
2 permeability” through the whole oral mucosa, recent efforts have focused on differentiat-
3 ing permeability between structurally different regions of the oral mucosa (Beckett and
4 Moffat, 1969; Tucker, 1988).
5
6
Advantages of the Oral Route for Drug Delivery
7
8 Most research has focused on the absorption and bioavailability of actives in the GIT
9 epithelial area of the stomach and lower intestines. Orally administered actives, however,
10 and their subsequent transport across the oral cavity are less understood. Transport of
11 actives across the oral route, nevertheless, has many advantages including:
12
13 1. Rapid action or onset of actives: The oral cavity is very rich in blood vessels (Table 8.2).
14 Blood supply from the buccal mucosa, unlike the rest of the gastrointestinal tract, does
15 not drain into the hepatic portal vein, since these peripheral areas are not specialized for
16 nutrients absorption. Buccal dose forms have often been found to show the same
17 bioavailability as intravenous formulations, without the need for aseptic preparations.
18 2. Bioavailability: Absorption of drugs via the oral route can avoid first-pass organs such as
19 the intestine, liver, and lung (Pang, 2003).
20 3. Actives can be incorporated into consumer-friendly formats (confectionery products),
21 which may help in masking the taste of some objectionable actives.
22
23
Disadvantages of Oral Route Delivery
24
25 Despite the role of oral mucosa in transporting nutrients and actives, several structural prob-
26 lems hinder the delivery of active substances across the oral mucosa.
27
28 1. Compared to the intestinal lining, the oral cavity occupies a very small surface area
29 (2–5 cm2).
30 2. Buccal cavity, like the entire alimentary canal, is a lipoidal barrier to the passage of
31 active substances. Active transport, pinocytosis, and passage through aqueous pores play
32 only insignificant roles in moving actives across the oral mucosa; hence the majority of
33 absorption is passive and only lipophilic molecules are well absorbed. Polar actives, that
34 is, those ionized at the pH of the mouth (6.2–7.4), are poorly absorbed.
35 3. Little intercellular absorption is possible across the cuboid squamous epithelium of the
36 oral cavity. However, some amino acids such as glutamic acid and lysine and some
37
38
39 Table 8.2. Blood flow (ml/min/100 cc) in various regions of the
oral mucosa of the rhesus monkey (Veillard et al., 1987 with
40 permission)
41
42 Region of oral mucosa Blood flow (ml/min/100 cc)
43 Buccal tissue 2.4
44 Sublingual floor of mouth 0.97
45 Sublingual ventral tongue 1.17
46S Gingival tissue 1.47
Palatal tissue 0.89
47N
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 181

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 181

vitamins such as L-ascorbic acid, nicotinic acid, and thiamine are transported via a carrier- 1
mediated process. 2
4. Dose form must be kept in place while absorption is occurring since excessive salivary 3
flow may wash the substances away. 4
5
6
Dosage Formulation: Physicochemical Properties of the Active and Dosage 7
A further issue affecting the absorption of orally administered actives is their physicochem- 8
ical properties. Most of these actives are presented in the form of tablets or capsules so in 9
order to be absorbed, the carriers have to be disintegrated or dissolved. A variety of factors 10
affect the dissolution rate and therefore the availability of the actives for absorption. Prod- 11
uct characteristics include format (tablet, lozenge, chewing gum, edible strips, capsule), 12
particle-size distribution of the active, dosage porosity, and presence/absence of coatings. 13
14
15
Chewing Gums 16
Compared to other confectionery formats, chewing gums provide the most hospitable envi- 17
ronment for encapsulated and unencapsulated ingredients due to the mild preparation 18
conditions, mainly the absence of heat stress or excess moisture. In addition, the physico- 19
chemical properties of gum base can be used effectively to delay/sustain the release of 20
actives. There is no monograph about chewing gum in any pharmacopoeia, but it is described 21
in guidelines for pharmaceutical dosage forms issued by the Commission of the European 22
Communities (1991) as a “solid preparation with a basis consisting of gum which should be 23
chewed and not swallowed, providing a slow steady release of the medicine contained.” 24
Controlling the release of flavors and active ingredients from chewing gums can best be 25
accomplished by a thorough understanding of the complex chemistry of gum bases and 26
their binding affinity to those ingredients. 27
28
29
Typical Chewing Gum Composition and Manufacturing 30
31
Chewing Gum Composition
32
Chewing gum preparations involve gum base and nonbase components (flavor, sweeteners, 33
color, etc.). Optional ingredients include vitamins, cooling and warming agents, menthol, 34
and other active ingredients. A typical chewing gum formulation is shown in Table 8.3. 35
36
Table 8.3. Typical composition of chewing gum formulation 37
38
Component Sugared Sugar-free 39
Gum base 18% 22–30% 40
Glucose syrup 45° 19% — 41
Mannitol 0–5% 42
Powdered sugar (to make 100%) —
Liquid sorbitol 70% 15–22%
43
Flavor 1.0% 1–1.5% 44
Color 0.1% 0.1% 45
Salvage 5% 5% S46
Glycerin 2% 1–6% N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 182

182 Chapter 8

1 Gum base formulations are usually held as trade secrets by confectionery manufacturers;
2 due to the competitive nature of the chewing gum business, most chewing gum companies
3 have established their own gum base manufacturing factories. Several categories of gum
4 bases can be distinguished, depending on the ultimate application. Gum bases are primarily
5 made up of hydrophilic and hydrophobic polymers (styrene-butadiene elastomers, polyviny-
6 lacetates (PVA), waxes, elastomer plasticizers, waxes, fats, oils, softeners, emulsifiers,
7 fillers, texturizers (talc, calcium carbonate), hydrogenated soybean oil, sugar, glycerin, fla-
8 vors, color, antioxidants, and other minor ingredients. Gum bases are notorious for their
9 affinity to most flavors, thus complicating their release from the chewing gum matrix.
10 A new category of ingredients referred to as sensates has recently been introduced
11 into chewing gums and other confectionery products to provide unique trigeminal sensa-
12 tion of cooling, warming, tingle, etc. When combined with flavors, cooling agents such as
13 N-ethyl-p-menthane-3-carboxamide, N,2,3-trimethyl-2-isopropyl-butanamide, menthyl
14 glutarate, menthyl lactate, isopulegol, menthone glyceryl ketal, and others have been found
15 to enhance the pleasant perception of flavors and breath freshening (Johnson et al., 2004;
16 Wolf et al., 2005). Similarly, warming/heating agents such as capsicum oleoresin, cinnamic
17 aldehyde, pepper oleoresin, gingerol, shoagol, etc. are often used to provide unique warm-
18 ing sensation in the mouth and the trigeminal area.
19
20
Chewing Gum Manufacture
21
22 Gum base is softened or melted (50–70°C) and placed in a kettle/mixer fitted with z-shaped
23 blades for 10–30 minutes. Powdered sweeteners, syrups, active ingredients are added fol-
24 lowing accurate time schedule. Late in the mixing procedure, flavors and cooling/warming
25 agents are then added and the mixture is cooled to 35–45°C, rolled onto plates, scored into
26 strips, and cut into pieces to produce sticks or tablets. Recently, extruders have been intro-
27 duced for manufacturing chewing gums due to the efficiency, process flexibility, and cost-
28 effectiveness of such units.
29 Coating is an essential step in finishing pellet gums and where flavors, colors,
30 and actives can be added. Sugar syrup, gum arabic, starches, and other binders are applied
31 to the surface of gum pellets placed in rotating basket-type mixing/coating units. Tum-
32 bling continues until sufficient amounts of coating material are applied followed by gentle
33 polishing to provide smooth surface free of imperfections. A variety of ingredients
34 such as waxes, shellac, talc, and emulsifiers can be used in chewing gum polishing
35 applications.
36 Coated chewing gums are hardened in the 8 weeks following preparation; the coating
37 sugars and polyols (sorbitol and xylitol) develop crystals that provide hardness and
38 crunchy texture. Crystallization, however, creates a porous coating structure with multiple
39 microchannels. The latter can allow migration of moisture and oxygen, thus exposing labile
40 actives to possible losses or degradation reactions.
41
42
Chewing Gums for Delivering Flavors and Nonmedicated Actives
43
44 Significant advances in delivering unique flavors and sensations via chewing gums have
45 been documented in the patent literature. Most of these patents have been filed in the last
46S decade by gum manufacturers and flavor houses who have been keen on adapting their deliv-
47N ery systems technologies to this highly profitable sector of the confectionery business.
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 183

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 183

Loss of flavor, either due to degradation from the product or due to its tight binding to 1
the gum base, remains the most challenging concern in chewing gum manufacture. Several 2
approaches have surfaced recently for addressing this problem either directly by (i) acceler- 3
ating flavor release (burst effect) or indirectly (ii) by enhancing flavor perception via 4
extending the release of physiological cooling agents or sweeteners throughout the prod- 5
uct’s normal chewing time. Encapsulated flavors prepared by coacervation, particle coat- 6
ing, entrapping into liposomes or amorphous matrices can be simply applied to the outer 7
coating of chewing gums. For burst-release effect, it is advisable to entrap flavor compo- 8
nents into a water-soluble matrix so that flavor release can take place instantly as the prod- 9
uct is placed in the mouth (Clark and Shen, 2004). The following citations represent a 10
group of controlled-release applications of chewing gums designed for local delivery of 11
flavors, sweeteners, breath-fresheners, etc. 12
A two-step process for controlling the release of flavors from a chewing gum system 13
was devised by Merritt et al. (1985), where the flavors were incorporated into an emulsion 14
via a hydrophilic matrix. The latter is further dried and ground to appropriate particle size 15
followed by coating with a water-impermeable substance (matrix-reservoir combination 16
systems). Song and Courtwright (1992) patented a method for manufacturing sustained 17
flavor releasing structures. The inventive process comprised of blending the flavors and 18
binding material such as amorphous silicon dioxide hydrates and further coating the com- 19
positions with a barrier material such as polyvinylpyrrolidone (PVP). The amount of flavor 20
released was claimed to be about 20% during 20 min. of chewing compared to 35% in a 21
conventional chewing gum. Sustained flavor release was claimed for a process whereby the 22
flavor is partitioned into a water-soluble phase of the gum for immediate release, while the 23
delayed effect was provided by the other flavor portion embedded into the water-insoluble 24
fraction such as polyethylene or polypropylene (Rutherford et al., 1992). Song and Copper 25
(1992) disclosed an innovative approach to controlled release via a fiber structure using 26
melt-spinning technique, which was followed by stretching via applying a draw or a 27
stretching force. The flavor droplets exposed along the sides of the fiber can be released as 28
the solvent infuses into the fiber creating channel-like structures. The length of these chan- 29
nels gradually increases as the active agent directly in contact with the solvent is dissolved. 30
The fiber structures can be incorporated directly into a chewing gum where the pressure 31
generated from chewing will flatten, stretch, and deform the fibers exposing new surface 32
areas of active to the solvent. 33
Wolf et al. (2005) patented a composition for extending the perception of breath- 34
freshening in a chewing gum by encapsulating physiological cooling compounds into the 35
gum matrix. Monitoring breath-freshening using trained sensory panel showed a signifi- 36
cant increase in perceived breath freshening intensity compared to unencapsulated control 37
(Figure 8.5). 38
McGrew et al. (2006) formulated a chewing gum containing metal salt, which is 39
claimed to reduce/eliminate oral malodors associated with bad breath. The controlled 40
release of Zn lactate and Cu gluconate claimed to provide breath-freshening benefits by 41
binding to volatile sulfur compounds generated in the GIT that are commonly associated 42
with bad breath. 43
Manufacturing good-tasting sugar-free chewing gums can be a challenging task because 44
of several factors, for instance incompatibility of artificial sweeteners with other compo- 45
nents of the chewing gum system. Aspartame (APM) is a methyl ester of L-aspartyl-L- S46
phenylalanine dipeptide molecule, which exhibits about 180 times the sweetening ability of N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 184

184 Chapter 8

1 10

Perceived breath freshness


2 9
3 8
Encapsulated
7 cooling agents
4
(intensity)
6
5 5
6 4
7 3
8 2 Control
1
9 0
10 0 5 10 15 20
11 Time (min.)
12
13 Figure 8.5. Perceived breath freshening intensity of chewing gum containing encapsulated
14 cooling agents and control (reproduced from Wolf et al., 2005).
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30 Figure 8.6. Effect of acids and gelatin on aspartame (APM) retention (reproduced from Bunzeck
31 and Urnezis, 1993).
32
33
34 sugar on an equal weight basis. APM can be destabilized and its sweetening power can be
35 significantly reduced in the presence of aldehyde-based flavors such as cinnamon resulting
36 in chewing gums with unacceptable taste, color, and texture. Similarly, APM can degrade
37 quickly in chewing gums containing sodium pyrophosphate. The latter is added to chewing
38 gum systems to provide teeth remineralization benefits.
39 Bunczek and Urnezis (1993) patented a method for stabilizing APM in cinnamon-
40 flavored chewing gums by mixing an aqueous solution of APM with hydrochloric acid and
41 a thickener (gelatin) followed by air-drying and grinding and further incorporation into a
42 chewing gum formula (Figure 8.6). Stability of acid-treated APM with or without gelatin
43 coating was found to be superior to the native untreated sweetener. Acesulfame-K (Ace-K)
44 is another sweetener that can be encapsulated to extend its release throughout chewing.
45 Broderick and Record (1992) developed water-insoluble porous beads that comprised a
46S copolymer of divinylbenzene and styrene impregnated with Ace-K. The beads were further
47N coated with hydroxypropylmethylcellulsoe (HPMC) prior to incorporation into a chewing
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 185

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 185

gum system. Release of Ace-K and spearmint was found to be about 40% after 5 min. of 1
chewing compared to 80% from control. 2
Other interesting controlled release systems have been documented in the patent litera- 3
ture such as those proposed by Johnson and Yatka (2000), Ream et al. (2003), Savage et al. 4
(2002), Sharma and Yang (1986), Song et al. (1992) and many others. 5
6
7
Chewing Gum for Delivering Caffeine 8
Caffeine is a well-known stimulant, which is used to alleviate the effects of sleep depriva- 9
tion and combat headache and fatigue. The caffeine molecule is completely metabolized by 10
the liver, its rate of inactivation is unaffected by the delivery to the liver and can only be 11
modified by a change in hepatic enzyme activity. Incorporating caffeine into beverages and 12
other food products has been very challenging due to many constraints—mainly its aque- 13
ous insolubility (2.1%), objectionable bitterness as well as delayed stimulant activity. 14
Syed et al. (2005) studied the pharmacokinetics of three doses (50, 100 and 200 mg) of 15
caffeine delivered via Stay Alert® chewing gum and proposed a dose-proportionate linear 16
increase in plasma caffeine levels. Their study showed that delivering caffeine via chewing 17
gums is an effective and convenient means of maintaining desirable levels of alertness and 18
performance in sleep-deprived individuals. The same chewing gum (Stay Alert®) was also 19
used earlier by another research group (Kamimori et al., 2002) to compare the bioavailabil- 20
ity of caffeine delivered via a chewing gum and a capsule. Mean plasma Tmax for individu- 21
als who chewed the gum was found to be in the range 44.2–80.4 min. compared to 22
84.0–120.0 min. for the capsule group, indicating an early onset of pharmacological effect 23
and a faster rate of absorption of the caffeine molecule via the buccal mucosa. 24
Enhancing buccal absorption of caffeine was attempted both in vivo and in vitro. 25
Donbrow and Freidman (1974) investigated the release properties of caffeine using a diffu- 26
sion cell and concluded that diffusion of caffeine via an ethyl cellulose cell was time- 27
dependent, that is, the diffusion follows a zero-order mechanism. Increasing membrane 28
hydrophilicity by incorporating 40% polyethylene glycol (PEG) demonstrated the possibil- 29
ity of enhancing permeability of the caffeine molecule (Table 8.4). An in vitro study by 30
Nicolazzo et al. (2003) showed that pretreating porcine buccal mucosa with different levels 31
of sodium dodecyl sulfate (SDS) (0.05, 0.1, and 1%) significantly enhanced caffeine flux 32
by a factor of 1.57, 1.63, and 1.81% respectively. 33
Gudas et al. (2000) developed a chewing gum containing slow-releasing caffeine profile 34
by pre-encapsulating the active (50–100 mg caffeine) into a water-soluble matrix. Con- 35
trolled release of small amounts of caffeine over a longer period of time was designed to 36
reduce the impact on taste. Results from the corresponding clinical trials conducted using 37
6 subjects showed enhanced absorption rate constant (Ka ) when caffeine was administered 38
through the chewing gum due to high buccal absorption rate and subsequent fast delivery 39
into the systemic circulation. A similar change in the onset of dynamic response was 40
noted, for example alertness and performance when caffeine was incorporated at 50–500 41
milligrams levels. Plasma caffeine concentration was also found to be significantly greater 42
for gum than caffeinated cola or other beverages within the first 10–30 min. after caffeine 43
intake, that is, faster uptake by the body. 44
Ream et al. (2001) formulated a chewing gum for delivering caffeine by layering the 45
active onto the outer shell of the pellet coating. Their study demonstrated significant levels S46
of buccal absorption presumably due to pressure development in the buccal cavity, a result N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 186

186 Chapter 8

1 Table 8.4. Transfer rate of caffeine as a function of film thickness, caffeine concentration and
2 PEG concentration in film (Donbrow and Freidman, 1974 with permission)
3 Rate of transfer reciprocal Rate of transfer/film thickness
4 Film thickness (cm  104) (mol s1  109) (mol cm s1  107)
5 26.8 400 1.07
6 32.7 3.32 1.08
7 48.0 2.20 1.06
8 Concentration of caffeine Rate of transfer/caffeine
(mol  108 l 1) concentration (l s1  107)
9 1.03 0.52 0.505
10 2.06 1.06 0.509
11 4.12 2.10 0.509
12 Concentration of PEG Rate of transfer/PEG concen-
13 in film (% w/w) tration (mol s1  109)
0 0.123 —
14 10 1.15 0.115
15 20 2.06 0.103
16 40 4.45 0.111
17 50 5.40 0.108
18
19
20 of continued chewing, which may have forced the permeation of the released caffeine
21 molecules through the mucosa.
22
23
Chewing Gums for Delivering Vitamins
24
25 The release of ascorbic acid from chewing gum formulations was investigated by several
26 groups. Ascorbic acid mixed with hydrophobic components has been shown to exhibit a
27 slower but complete release of the drug within 15 min., compared to mixing with hydrophilic
28 ones (Odumusu and Wilson, 1977; Sadoogh-Abasian and Evered, 1979). A slightly faster
29 release of L-() ascorbic acid was observed in vivo compared to its in vitro release in a masti-
30 cation machine; however, a good correlation was observed between the in vivo and in vitro
31 release patterns within the first 5 min. of mastication. Andersen (2004) incorporated vitamin C
32 into a chewing gum formulation at two different gum base levels (30 and 45%) and showed a
33 very high level of vitamin recovery especially in the 30% gum base formulation.
34 Sadoogh-Abasian and Evered (1979) and Stevenson (1974) independently studied the
35 transport of ascorbic acid across the human mucosal membranes and showed that its absorp-
36 tion is Na ion-dependent. Calcium ions were also found to increase ascorbic acid absorption
37 presumably due to a secondary effect of Na ion fluxes. Buccal mucosa was found to be
38 permeable to dehydroascorbic acid and D-isoascorbic acid. The presence of D-glucose and
39 3-O-methyl-D-glucose increased the absorption of ascorbic acid but D-fructose had little
40 effect and D-mannitol had no effect.
41 The impact of ascorbic acid solubility and its ionization behavior on the vitamin buccal
42 permeability were also studied by changing pH of the medium stepwise from 3.4 to 9.0.
43 Increasing pH resulted in a gradual decrease in buccal absorption of the vitamin. Vitamin C is
44 only 13.7% ionized at pH 3.4 but almost fully ionized at pH 9.0, thus indicating passive diffu-
45 sion of the molecule (Odumusu and Wilson, 1977). The process was reported to be not stere-
46S ospecific since both the natural form L-ascorbic acid and the unnatural D-isomer were
47N transferred across buccal mucosa at similar rates. Glucose was also found to enhance transfer
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 187

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 187

of ascorbic acid across buccal membranes, though the effect may be due to glucose acting 1
as an energy metabolite (Stevenson, 1974). 2
Nicotinic acid and nicotinamide displayed similar buccal absorption rates. The latter 3
were also consistent with those determined across intestinal mucosa (Evered et al., 1980). 4
Despite the differences in their ionization behavior at pH 6.0 (nicotinic acid is 6% union- 5
ized while nicotinamide is almost fully ionized), both forms of the vitamin are soluble in 6
water and poorly soluble in lipids. Isonicotinic acid and nicotinic acid have identical 7
molecular weights and very similar pKa values (4.84 and 4.77, respectively), yet the former 8
showed a lower rate of absorption suggesting a carrier-mediated transport system (i.e. facil- 9
itated diffusion). These studies revealed for the first time that human buccal mucosa is 10
permeable to the water-soluble vitamin, thiamine, as with ascorbic acid (Sadoogh-Abasian 11
and Evered, 1979), nicotinic acid and nicotinamide (Evered et al., 1980) showing some 12
similarities to absorption from mammalian small intestine (Evered et al., 1980). 13
14
15
Chewing Gums for Delivering Antimicrobial Agents
16
Epigallocatechin gallate (ECGC) has been studied for its potential benefits in reducing the 17
risk of heart disease and cancer as well as weight loss. However, recent information from 18
the Tearrow Co. suggests that tea extracts of Camellia sinesis incorporated into chewing 19
gum can permeate the mucosal barriers to help in treating gingivitis and eliminating 20
microbial growth in the oral area (Gelski, 2006). 21
Miconazole is a well-known oral care antimicrobial active. Attempts to incorporate 22
miconazole directly into chewing gums were not very successful due to its strong binding to 23
the gum base. To promote its release, different solid dispersions of miconazole in PEG 6000, 24
PVP 40,000 and xylitol were tested. Dissolution rate data showed that dispersions of 25
miconazole:PEG 6000 (1:4) had the highest level of release from a chewing gum (15-times 26
compared to pure miconazole) due to enhanced aqueous solubility of the active. Addition of 27
lecithin to the miconazole–PEG chewing gum formulation was found to enhance the release 28
rate as well as the time of release both in vitro and in vivo. Lecithin may have improved 29
miconazole solubility by virtue of its ability to form liposomes in aqueous media (Pedersen 30
and Rassig, 1990). In vitro data using a mastication device of Christup and Møller (1986) 31
correlated well with the in vivo data derived from six healthy volunteers. Release of micona- 32
zole was also shown to be significantly facilitated by the addition of Panodan 165 (acidic 33
surfactant) to a chewing gum formulation. High surface activity of Panodan 165 as well as 34
its low pH may have increased the solubility of miconazole and/or enhanced saliva absorp- 35
tion into the chewing gum during mastication. 36
Using a panel of five subjects, Witzel et al. (1980) investigated the in vivo release of nys- 37
tatin, a slightly soluble antifungal agent from chewing gum. Coating the antifungal agent 38
with gum arabic resulted in 24% release of the nystatin compared to only 4% from uncoated 39
nystatin. Despite the enhanced solubility of nystatin using enhancers such as Cremophor 40
RH40, Tween 60 (nonionic surfactants) and Panodan AB 90, in vitro release rate was too fast 41
to provide any significant antifungal effect (up to 99% in the first 10 min. of administration). 42
Lombardy et al. (2001) disclosed an oral hygiene plaque-disrupting chewing gum compris- 43
ing a core containing encapsulated sodium bicarbonate, which is surrounded by a coating 44
that contains an encapsulated edible acid. Upon chewing and subsequent effervescence 45
development, the formed foam penetrates between the teeth and gum crevices to loosen S46
plaque build up. N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 188

188 Chapter 8

1 Chewing Gums as Delivery Systems for Oral Health


2
3 Tooth decay is mainly caused by Streptococcus mutans metabolizing fermentable carbo-
4 hydrates leading to a drop in pH in the tooth and plaque microenvironment and to gradual
5 dissolution of the hydroxyapatite [calcium phosphate hydroxide, Ca10 (PO4)6(OH)2], the pri-
6 mary component of tooth enamel. Natural remineralization process involves, in part, the
7 flow of saliva (saturated with calcium and phosphate) that raises the pH so that the calcium
8 and phosphate ions can precipitate to replace the dissolved hydroxyapatite. However, this
9 process is very inefficient in most individuals.
10 Sugar-free chewing gums have been promoted for their effectiveness in preventing dental
11 caries. Based on clinical trials, it was suggested that mastication of xylitol-based chewing
12 gum may reduce dental caries in children and young adults better than any other sugar-free
13 chewing gum. This improvement has been associated with reduced levels of Streptococcus
14 mutans and Lactobacilli in saliva along with a reduction in plaque build up at neutral pH
15 (Assev and Rølla, 1986; Wennerholm and Emilson, 1989). However, S. mutans may develop
16 resistance to xylitol after few months of chewing.
17 The effect of chewing nonmedicated chewing gums on plaque pH, saliva flow rates and
18 the incidence of dental caries have been the topic of many studies. Different brands of sugar-
19 free chewing gums claim to stimulate saliva flow rate compared to that of unstimulated
20 saliva flow rate. Peak salivary flow rates are known to develop within the first minute
21 of chewing.
22 Leach et al. (1989) presented evidence for the remineralization of artificial caries-like
23 lesions in human teeth enamel in situ following mastication of a sorbitol-containing chewing
24 gum. Winston and Usen (2002) patented a confectionery composition containing soluble
25 phosphate and calcium salts that are claimed to help remineralize teeth surface lesions and
26 exposed dentin tubules. The composition was described to be applicable to hard candies,
27 chewing gums, lozenges as well as other formats.
28 Patients with xerostomia (dry mouth) most often show elevated risk of caries. Sugar-free
29 chewing gums have been recommended to patients who still have some capacity to secrete
30 saliva (Jenkins and Edgar, 1989). The saliva produced during mastication can alleviate the
31 risk of caries by reducing plaque pH generally seen in response to a sucrose challenge. To fur-
32 ther increase caries prophylactic activity of chewing gum, addition of an acid-neutralizing
33 agent (e.g. carbamide) may be appropriate.
34
35
Chewing Gum for Delivering Acetylsalicylic Acid
36
37 The high clearance drug salicylamide has been used as a model substance in chewing gum
38 experiments (Christup et al., 1988b). The bioavailability of acetylsalicylic acid from Asper-
39 gum® has been compared to the bioavailability of acetylsalicylic acid from pre-oral tablets.
40 The rate of absorption was shown to be faster from chewing gum than from the tablets and it
41 was concluded that chewing gum might provide a faster relief of pain (Woodford and Lesko,
42 1981). These results were speculated to be due to a reduction in drug metabolism in the GIT
43 and the liver.
44 Christup and co-workers (1990, 1988) studied the in vitro and in vivo release of different
45 salicylamide chewing gum formulations and showed higher release from the formulation
46S containing less gum base. Micronized salicylamide in a chewing gum showed greater
47N release from hydrophilic formulations compared to their hydrophobic counterparts when the
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 189

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 189

gum was chewed for 30 min. (Christup et al., 1988). The release of coarse salicylamide 1
particles from a chewing gum composition was reported to be limited, but was doubled 2
when the active was micronized. 3
These studies suggest that drug release from chewing gum can be modified through for- 4
mulation or manufacturing processes either by the addition of substances to the gum base 5
which exhibit different lipophilic or hydrophilic characteristics or by modifying the physical 6
characteristics of the incorporated drug. 7
8
9
Comparison of Delivery Profiles between Chewing Gum and Lozenge
10
Christup et al. (1990) used gamma-scintigraphy to examine how effectively drugs were 11
released in vivo from chewing gum, their distribution profile once released and the length of 12
time drug remained in the oral cavity following release. Release profile from chewing gum 13
was compared to those observed following the administration of lozenges and sublingual 14
tablets. Vitamin C was found to be absorbed better from a chewing gum than from a tablet 15
(Christup et al., 1988) 16
Non-absorbable, water-soluble compound Tc E-HIDA (N-(N-(2,6-dimethylphenyl)) car- 17
bamoylmethyl iminoacetic acid) used as a model active showed complete release after 18
10 min. of chewing at a rate of 1 chew/sec. Activity (counts) vs. time (min) profiles of the oral 19
cavity and the stomach of six subjects following administration of the 3 different dosage 20
forms (chewing gum, lozenge and sublingual tablet), did not show any difference in the 21
distribution of Tc-HIDA within the oral cavity, glottis or upper oesophagus. However, 22
Tc E-HIDA released from sublingual tablets remained for the longest time while Tc E-HIDA 23
released from lozenges remained for the shortest period in the oral cavity (Rassing, 1994). 24
25
26
Lozenges (Hard Boiled Confections)
27
Lozenges have long been used as vehicles for delivering medicaments to alleviate cold 28
symptoms such as decongestion, to soothe sore throats and clear nasal passages. Such 29
medicaments include analgesics, antitussives, expectorants, cooling, warming, numbing 30
and tingle agents. 31
Lozenges are essentially hard-boiled candies that can be formulated in sugared and 32
sugar-free versions. Lozenge manufacture involves heating a glucose/sucrose mixture to 33
evaporate water and transform the matrix from a crystalline to an amorphous glassy phase. 34
Lozenges can be manufactured using batch or continuous processes. Essential additives 35
include acids, flavors and colors while therapeutic additives include actives such as menthol, 36
benzocaine, dextromethorphan, pectin, vitamins or other nutrients. Lozenge manufacture 37
exposes these actives to harsh moisture and heat environments and often results in partial or 38
total degradation of heat-labile components. 39
40
41
Lozenges for Delivering Flavors and Sensates
42
Release of actives from a lozenge is activated by sucking and gradual dissolution of the 43
sugar (or sugarless) matrix. Menthol released from lozenges, for example, can bind to 44
thermo-receptors located within the free nerve endings of the trigeminal and nasal cavities. 45
The resulting cooling sensations are presumed to provide analgesic effects via modulating S46
the sensitivity of cutaneous pain fibers. N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 190

190 Chapter 8

1 Due to their water-based nature, lozenges can be most effective in providing “burst
2 release” of flavors, sensates or other actives. Their hydrophilicity makes them ideal perme-
3 ability enhancers for hydrophobic actives (e.g. cooling compounds) across membranes of
4 the oral area. Incorporating encapsulated particulates into lozenge formulations results
5 most often in premature destruction of the capsule and the release of entrapped sub-
6 stances during processing and before consumption. Micelles and emulsion-based carrier
7 systems are, generally speaking, better suited for lozenge formulations.
8 An effective “burst” release approach was developed by Clark and Shen (2004) whereby
9 encapsulated particulates or powders were layered onto the lozenge outer surface to
10 provide quick dissolving matrix to liberate the entrapped flavor components. Rivier (2005)
11 patented a lozenge-based delivery system to provide “burst” release of a solid active nes-
12 tled into the center of an oval-shaped lozenge. Due to the inherently thin walls at the ends of
13 the larger diameter of the oval piece, “burst release” is activated by sucking the lozenge
14 and creating channels for quick diffusion of the active. A wide range of actives can be
15 incorporated into this lozenge design including flavors and sensates for refreshment.
16 A unique type of actives that can be dosed into the center of the oval piece is polyols.
17 By virtue of their negative heat of dissolution in particular xylitol and eryhthritol,
18 their release and high solubility in the saliva can provide a refreshing moist cooling
19 sensation. Therapeutic actives such as analgesics can be also employed to provide
20 quick relief.
21
22
Lozenges for Delivering Throat Relief Actives
23
24 Despite these advantages, sugar- or polyol-based lozenges do not adequately provide
25 long-lasting solutions to problems unique to the mouth and the esophageal area due to the
26 quick dissolution, short residence time and mode of lozenge consumption, that is, moving
27 around in the mouth and saliva stimulation that can be secreted and swallowed. A recent
28 trend in lozenge formulations involves incorporating high molecular weight polymers with
29 mucoadhesive properties into sugared/sugar-free formulations. This practice is supported by
30 the USP monograph permitting the use of mucoadhesive materials (referred to as demul-
31 cents) for providing relief from mucus irritation, pain and discomfort associated with laryn-
32 gopharyngitis (sore throat) and other upper respiratory tract infections. Examples of these
33 demulcents include gelatin, pectin, celluloses, and alginates. Other types of nonpolymer-
34 based mucoadhesive agents include titanium and silicon dioxide (Dobrozsi, 2003) and lipid
35 vesicles (Bealin-Kelley et al., 2002).
36 Lozenges formulated with demulcents, however, are bland tasting and are often
37 perceived by consumers as nonefficacious. A new generation of lozenges has been formul-
38 ated with sensates (cooling or warming compounds) to provide an additional sensory
39 cue of soothing. Coincidentally, it was discovered that combining demulcents with sen-
40 sates can extend the perceived soothing warmth from the mouth to the throat (Figure 8.7),
41 an attribute that is highly desirable by consumers (Bealin-Kelley et al., 2002; Lakkis,
42 2006).
43
44
Lozenges as Delivery Systems for Dry Mouth Relief
45
46S Lozenges have also been formulated to provide relief from dry mouth symptoms referred to
47N as xerostomia. Wolfson (2002) patented a composition using Heliopsis longipes root for
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 191

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 191

1
time, min
4 2
3
8 3 1
4
5
2
6
1 0.3% pectin 7
7 2
8
0
9
10
0.2% pectin 11
6 3 12
13
14
5 4 15
0.1% pectin
16
Figure 8.7. Effect of pectin level (0.1, 0.2 and 0.3%) on perceived warming in the human throat.
17
Sensory panel ratings scale ranged from 0 (very low warming intensity) to 4 (highest warming 18
intensity). Graph reproduced from Lakkis (2006 with permission). 19
20
21
22
increasing salivation, thus alleviating dry mouth feeling while maintaining oral hygiene. 23
Tutuncu et al. (2003) developed a food acid-containing lozenge, which claimed mouth 24
moistening benefits. Kayane et al. (2003) described a throat care lozenge, which promotes 25
the secretion of mucin to provide bactericidal effect by inhibiting the adhesion of pathogenic 26
bacteria, Pseudomonas aeruginosa, Haemophilus influenzae, or Staphylococcus aureus. 27
Efficacy of the lozenge was confirmed via ELISA testing, which showed increased levels of 28
IgA and lysozyme content in the mucus secretion. 29
30
31
Lozenges as Delivery Systems for Teeth Remineralization Actives
32
Calcium phosphate, the main component of dentin and teeth enamel, while insoluble at 33
neutral saliva pH, is readily soluble in acidic media generated by fermentation of ingested 34
carbohydrates. To alleviate the impact of these events and slow down the develop- 35
ment of caries and lesions, it is desirable to increase the available concentration of calcium 36
and phosphate ions in the oral cavity to speed up the remineralization process. Several 37
inventions and commercially available confectionery products (lozenges and chewing 38
gums) that claim teeth remineralization are commercially available (Chow and Takagi, 39
2001; Kaufmann, 2003; Mazurek et al., 2000; Savage et al., 2002; Winston and Usen, 40
2002). 41
One drawback with using calcium- and phosphate-containing salts and buffers in 42
hard-boiled lozenges is the development of bitterness and grittiness due to complexation 43
of the calcium and phosphate ions in the candy matrix. One approach to overcome these 44
drawbacks is to skillfully incorporate these actives into separate components of the 45
candy matrix so that the actives can be released concurrently in their soluble forms to form S46
the hydroxyapatite in the generated saliva (Lakkis and Wong, 2007). N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 192

192 Chapter 8

1 Bioerodible and Bioadhesive Devices


2
3 This category encompasses a wide range of weak and strong adhering structures that are
4 commercially available in the form of films/strips, patches, tablets or other devices. Weak
5 adhesion is desirable for breath freshening and flavor delivery films while strong adhesion is
6 critical for medicated applications (e.g., antimicrobials and teeth whitening strips).
7 Bioadhesive and bioerodible devices represent an ideal delivery system due to many
8 attributes such as (i) ease of consumption by individuals having difficulties in swallowing,
9 (ii) localization in specified regions of the oral or other GIT sites to improve and enhance
10 the bioavailability of actives, (iii) optimum contact with the absorbing surface to permit
11 modification of tissue permeability, (iv) reduced need for drug overage and (v) avoidance
12 of first-pass metabolism.
13 Mucoadhesion takes place by establishing an adhesive bond between the device and
14 the mucus membranes resulting in a reduced total surface energy of the system because
15 two free surfaces are replaced by one (Anlar et al., 1984; Guo, 1994). Polymers with
16 hydrophilic moieties such as carboxyl and hydroxyl groups can bind to the sialic acid and
17 other oligosaccharide residues in the mucosal membranes. The process takes place in three
18 stages: hydration, interpenetration, and mechanical interlocking between mucus and the
19 polymer. Mucoadhesive strength is affected by various factors such as molecular weight of
20 the polymer, its swelling power, size and configuration of the device, time of contact with the
21 mucus, and the physiological nature of the membrane. Generally, oral bioadhesive and bio-
22 erodible devices can be designed to adhere to the cheek (buccal area), the floor of the mouth
23 (sublingual tissue), gums surrounding the teeth, and the roof of the mouth (palate tissue).
24 Efficacy of mucoadhesive and bioerodible devices is well documented in many pharma-
25 ceutical and consumer health applications. Examples include teeth-whitening, accel-
26 erated healing of inflamed or damaged tissues, prolonged and improved coating and
27 protection of the mouth and esophagus (Barklow et al., 2002; Choi and Kim, 2000), sus-
28 tained release of insulin in the stomach via mucoadhesive microspheres of glizpide, which
29 is a second-generation sulfonylurea used to acutely lower blood glucose levels (Kahn and
30 Shechter, 1991).
31 Confectionery-based devices are available in the form of medicated and nonmedicated
32 films commonly referred to as edible strips. The latter are usually of the size of a postal
33 stamp, which is placed on the tongue to deliver flavors, breath-fresheners, decongestants,
34 etc. Popularity of these films has soared recently with the introduction of Listerine® pocket-
35 paks™ by Pfizer, Eclipse® Flash by Wrigley’s and most recently Theraflu® cough relief
36 strips by Novartis, Inc.
37 A wide variety of water-soluble and/or -insoluble food-grade hydrogels has been used in
38 formulating edible strips. The choice of a suitable composition depends largely on desired
39 functionality and residence time in the oral cavity, its solubility, type of active, and required
40 payload. Typical edible-strips formulations comprise a combination of film-forming poly-
41 mers, fillers, plasticizers, colors, and actives (menthol, flavor, cooling/warming compounds,
42 vitamins, analgesics, etc.). Polymers such as pullulan are favored for their film forming
43 properties and excellent solubility and clean aftertaste (no residual gumminess). Manufac-
44 turing films with pure pullulans, however, has been hindered by its weak mucoadhesive
45 properties and cost. Mucoadhesion of pullulans may be improved by incorporating several
46S additives such as PEO, mono- or oligosaccharides to the strip formulation (Ozaki and
47N
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 193

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 193

Miyake, 1995). Alternative economical film materials have been proposed such as cellu- 1
loses, glucans, native, and modified starches. 2
Edible strips manufacture involves forming a hot, stable aqueous solution of the film 3
polymer and the active(s), casting the solution over a conditioned belt followed by heat 4
drying and cutting into desired strip dimensions. Such conditions may degrade heat and 5
moisture labile actives. Encapsulating actives (e.g., food acids) prior to their incorporation 6
into film formulation may help retard their degradation and help maintain the film integrity 7
(Virgallito and Zhang, 2006). 8
In vivo assessment of release mechanisms from mucoadhesive devices has been hindered 9
by physiological variables such as amounts and physicochemical properties of human saliva 10
and movement of the device in the mouth. Most investigations indicate that the release from 11
mucoadhesive devices takes place via erosion, diffusion, or a combination of both. Critical 12
factors that can have profound impact on release from bioadhesive/bioerodible devices can 13
be summarized as follows: 14
15
1. Film physicochemical properties, mainly active payload, film forming polymer chem- 16
istry, its thickness and solubility (addition of maltodextrin to pullulan films can enhance 17
their dissolution and release of the active). 18
2. Although high viscosity polymers can improve the bioadhesion of films, at very high vis- 19
cosity, nonhomogeneous distribution of the active may result in unpredictable drug 20
release rates (Wong et al., 1999). 21
3. Location of the device in the mouth as well as tongue movements can play a crucial role in 22
the device’s residence time and the active’s release rate/mechanism. De Vries et al. (1991) 23
showed that application of buccal patch to the palate provided longer adhesion than 24
that of the cheek mucosa. Bouckaert et al. (1993) compared the adhesion of miconazole 25
mucoadhesive tablet to the gingival, palatal, and cheek mucosa and concluded that the 26
longest adhesion was in the gingival area while the shortest was at the cheeks. When 27
applied to the cheek, the tablet is lodged very near the parotid duct, thus the adhesion time 28
might be reduced by the salivary flow. Subsequently, the polymer mixture may swell more 29
rapidly and the tablet will become prone to erosion. Despite the fact that less saliva is 30
present in the palatal and gingival mucosa compared to the buccal mucosa, adhesion time 31
for the palate was comparable to that for the cheek and significantly lower than the 32
gingival. 33
4. Excessive matrix swelling, such as the case with native starch, can hinder the controlled 34
release of actives. Tuovinen et al. (2003) using two small model molecules (sotalol, 35
m. wt. 308 and timolol m. wt. 332) and two large model molecules (FITC dextran, 36
m. wt. 4400 and BSA m. wt. 68,000) showed that the small molecules, solatol, and tim- 37
olol were released more rapidly than the FITC dextran and BSA from a native potato 38
starch matrix (PBS buffer pH 7.4 with or without α-amylase) due to excessive starch 39
swelling. Release of the small molecules was continuous whereas the release of macro- 40
molecules showed discontinuities. 41
5. Hydrophobic/hydrophilic nature of the hydrogel and active also can have a significant 42
impact on the release behavior. Tuovinen et al. (2003) showed that the model molecule, 43
sotalol, was released faster than timolol (more hydrophobic) from a starch acetate film 44
(hydrophobic) demonstrating stronger interaction between the matrix and active. 45
Release of timolol and sotalol was faster than the weight loss of the corresponding film S46
N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 194

194 Chapter 8

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17 Figure 8.8. Cross-section of a typical seamless capsule showing solid shell and a liquid center.
18
19
20 presumably due to the hydrophobic nature of the starch acetate film, thus providing
21 evidence for erosion-controlled mechanism.
22
23
Seamless Capsules
24
25 Seamless capsules represent a class of delivery systems that can provide powerful impact,
26 but has not yet realized its full potential. Soft seamless capsules are ideal carriers for liq-
27 uids or a suspension of solids in a liquid. A variety of applications have been documented
28 in the patent literature, including delivery of flavors, menthol, and eucalyptus oil for breath-
29 freshening (Karles et al., 2006; Tanner and Shelley, 1996; Yang, 2005) the pro-vitamin
30 A lycopene (Paetau et al., 1999) and concentrated alcoholic and nonalcoholic beverage
31 concentrates for recreational use (Hutchinson and Garnett, 1999; Sexton and Lakkis,
32 2003).
33 Ideal seamless capsules are 4–8 mm diameter with a thin shell wall of 300–600 microns
34 and maximum core-to-shell ratio of 9:1 w/w. This ratio represents the highest payload of any
35 encapsulation technology known today. Seamless capsule formulations comprise a liquid
36 center and a solid soft shell. The latter can be made of gelatin, agar, alginates, celluloses, or
37 other gelling (moldable) polymers in combination with suitable plasticizers (Figure 8.8).
38 Seamless capsules are manufactured using specialized machines with coaxial multiple
39 nozzles such as the Spherex system (available from Freund Industrial Co. Japan) as well as
40 other suppliers. The outermost shell layer is formed by extrusion of a hot gelatin solution
41 (60°C) from the outer nozzle and the liquid core (immiscible in water) is extruded from an
42 inner nozzle to form a concentric jet. The jet is further injected into a cooled vegetable oil
43 bath (ca.12°C) to harden the shell. Seamless capsules in the form of spheres are formed due
44 to surface tension.
45 One of the critical attributes of seamless capsules is the shell quality, which is expected to
46S be soft and to dissolve readily in the mouth with no residues. For maximum efficacy and
47N clean aftertaste, release of the active from seamless capsules should take place via breaking
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 195

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 195

the sphere without any perceivable swelling. Few challenges, however, can complicate the 1
manufacture of seamless capsules and their functional performance, mainly: 2
3
1. Shell–core interactions: Due to the hydrophilicity of the shell material, this technology 4
can only accommodate hydrophobic cores. Hydrophilic substances can interact with the 5
shell material resulting in film plasticization and in some cases can help promote micro- 6
bial growth, a major concern upon processing and shipping of the capsules. 7
2. Shell thickness: Readily dissolvable capsules require the formation of a thin shell. 8
Shell thickness can affect capsule diameter, that is, for a constant core to shell-mass ratio, 9
shell thickness increases with increased diameter of the capsule. 10
3. Shell (polymer viscosity): Desired film properties can be achieved by maintaining a deli- 11
cate balance among many parameters, mainly between polymer viscosity and shell 12
dissolution in the mouth. Very low viscosity polymers can lead to capsule deforma- 13
tion and crushing while too high viscosity can lead to the formation of satellites extended 14
from the capsule surface. Wonschik et al. (2005) patented a unique shell formula- 15
tion comprising a mixture of high bloom and hydrolyzed gelatin (zero bloom). The high 16
bloom component is claimed to provide solid network, critical for shell processing 17
while the hydrolyzed gelatin occupies spaces in the formed network to provide rapid dis- 18
solution by the saliva. 19
4. Core Physical properties: For effective processing, liquids, solutions, or suspen- 20
sions should flow by gravity at room temperature. In general liquids with a wide range of 21
viscosity from 0.2 to >3000 cp at 25°C can be encapsulated. Also, encapsulated liquids 22
must have a pH from 2.5 to 7.5 beyond which the gelatin shell would deteriorate. 23
A new generation of seamless multilayered capsules has been commercialized recently 24
(Jintan Co., Japan) that claims a multitude of functions. Sunohara et al. (2002) patented a 25
multilayered seamless capsule design where the outer shell can provide flavor or breath- 26
freshening and the inner layers can be swallowed to treat stomach-originated bad breath. 27
28
29
Pressed Tablets 30
Pressed tablets include mouth dissolving, fast-dissolving, rapid-melt, porous, orodis- 31
persible, and melt-in-mouth products. A wide range of tablets and capsules are commonly 32
used for delivering breath freshening (breath mints) and pharmaceutical actives, although 33
the majority of such tablets are designed to be absorbed in the GIT. 34
Pressed tablets are prepared by dry blending the active ingredients with water- 35
disintegratable compressible carbohydrate and a binder and then compressing into a convex- 36
shaped tablet. Strength/compactness of these tablets can be detrimental to the extent of 37
disintegration, dissolution, and absorption of the active. One of the attractive features of 38
pressed tablets is the possibility of incorporating high levels of actives compared to other 39
dosage formats. 40
Haines (2004) investigated the buccal absorption and bioavailability of vitamin B12 from 41
pressed tablets and a nanofluidized B12 suspension (NF®) via spray applicator as an alterna- 42
tive means to deliver this essential nutrient to patients suffering from intestinal disorders such 43
as celiacs, who cannot absorb this vitamin from food sources. Results from that study showed 44
that nanodroplets provided a more effective vehicle for delivering the active molecule across 45
the mucosal barriers at a faster and more even rate than from the tablet or even the non- S46
processed or “normal” vitamin B12 solutions. N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 196

196 Chapter 8

1 Effervescent Tablets
2
3 Effervescent tablets can be designed to provide enhanced release in the mouth as well as
4 the lower GIT; Effervescence is the reaction (in water) of acids and alkalis to produce
5 carbon- dioxide. Typical acids used in effervescent tablets are citric, malic, tartaric, adipic,
6 and fumaric while sodium bicarbonate and potassium carbonate are the most commonly
7 used alkalis.
8 Effervescence can help promote calcium absorption in the stomach as well as sustain
9 biological activity of probiotic bacteria (Lee, 2004). It has been speculated that CO2 pro-
10 duced by effervescent reactions induce some alteration (widening) of paracellular path-
11 ways, a primary route of absorption of hydrophilic actives (Anderson, 1992; Nuernberg
12 and Brune, 1989). Absorption of hydrophobic species can also be enhanced due to the
13 nonpolar CO2 gas molecules partitioning into the cell membrane to create a hydrophobic
14 environment which allows hydrophobic actives to be absorbed (Eichman, 1997). CO2 may
15 also help absorption by reducing the thickness and viscosity of the mucus layer adjacent to
16 the mucosa (Pather et al., 2002).
17
18 Chewable Tablets
19 Most chewable confectionery products are gelatin based. Gelatin is a very reactive surface
20 that interacts with mucin rendering bioavailability very difficult.
21
22 Conclusions
23
24 The oral mucosa responds to the senses of pain, touch, and temperature in addition to its
25 unique sense of taste. Some physiological processes are triggered by sensory input from the
26 mouth such as the initiation of chewing, masticating, swallowing, etc. The most important
27 physiological variable, however, that can markedly affect the release characteristics of an
28 active from a confectionery dosage is whether a person sucks or chews the formulation,
29 since systems designed to be chewed will invariably be sucked and vice versa by some indi-
30 viduals. Chewing gums possess an advantage over other confectionery formats for control-
31 ling the release of drugs such as nicotine, caffeine, or other medicinal substances in that if
32 the gum is swallowed, release of the active in the stomach and lower GIT is extremely low;
33 therefore, reducing potential or toxicity.
34 Drugs and actives ingested via the oral route can be designed for either or both of the
35 following:
36
37 • Local Delivery: Confectionery products have demonstrated their practical effectiveness
38 in delivering flavors, cooling and warming agents, antimicrobials, caries prevention and
39 xerostomia relief agents; and
40 • Systemic Delivery: Nicotine, vitamins, caffeine, salicylic acid are the most common
41 actives that can be embedded in a confectionery matrix and have the potential to be
42 absorbed through the oral mucosa into the circulation, thus giving rise to a systemic effect.
43 Actives absorbed directly via the membranes lining the oral cavity avoid metabolism in
44 the GIT and the first-pass effect of the liver since the oral veins drain directly into the
45 vena cava. Alternatively, actives released from a chewing gum or other confectionery
46S dosage form—but not absorbed through the oral cavity—membranes will be swallowed
47N and enter the stomach in a dissolved or a dispersed form in saliva.
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 197

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 197

References 1
2
Andersen, 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: 3
31–35W. 4
Anlar, S., Capan, Y., Guven, O., Gogus, A., Dlarara, T. and Hincel, A.A. 1984. Formulation and in-vitro in-vivo 5
evaluation of buccoadhesive morphine sulfate tablet. Pharm. Res. 11: 231–236. 6
Assev, S. and Rølla, G. 1986. Sorbitol increases the growth inhibition of xylitol on Strep. mutans OMZ 176. Acta
7
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 8
making the same. US Patent 0131990 A1. 9
Bealin-Kelley, F.J., Hanke, B. and Nienaber, P. 2002. Throat soothing compositions. US Patent 6432441 B1. 10
Beckett, A.H. and Moffat, A.C. 1969. The influence of substitution in phenylacetic acid on their performance in 11
the buccal absorption test. J. Pharm. Pharmacol. 21: 139S–143S.
12
Benowitz, N., Jacob, P. and Savanapridi, C. 1987. Determination of nicotine intake while chewing nicotine
polacrilex gum. Clin. Pharmacol. Ther. 41: 467–473. 13
Berridge, M.J. and Oschman, T.L. 1972. In Transporting Epithelia, Academic Press, New York and London. 14
Blanchette, J., Kavimandan, N. and Peppas, N.A. 2004. Principles of transmucosal delivery of therapeutic agents. 15
Biomed. Pharmacother. 58: 142–151. 16
Bouckaert, S., Lefebvre, R.A. and Remon, J-P. 1993. In vitro/in vivo correlation of the bioadhesive properties of a
17
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 18
chewing gum ingredients and method. US patent 5139787. 19
Bunczek, M.T. and Urnezis, P. 1993. Aspartame stability in chewing gum using an acid gelatin system. US Patent 20
5192561. 21
Channer, K.S. and Roberts, C.J.C. 1985. Effect of delayed esophageal transit on acetominophen absorption. Clin.
22
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, 23
distribution and origin of the whole saliva enzymes. J. Dent. Res. 33: 321–334. 24
Chow, L.C. and Takagi, S. 2001. Calcium phosphate hydroxyapatite precursor and methods for making and using 25
the same. US Patent 6325292 B1. 26
Choi, H-G. and Kim, C-K. 2000. Development of omeprazole buccal adhesive tablets with stability enhancement in
27
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 28
and influence of formulation upon the release rate of a water soluble drug. Arch. Pharm. Chem. Sci. Edu. 14: 30–36. 29
Christup, L.L., Davis, S.S. and Frier, M. 1990. Deposition of a model substance 99mTcE-HIDA, in the oral cavity 30
after administration of lozenges, chewing gum and sublingual tablets. Int. J. Pharm. 66: 169–174. 31
Christup, L.L., Rasmussen, S.N. and Rassing, M.R. 1988a. Chewing gum as a drug delivery system. Proc. 3rd Int.
32
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 33
drug delivery system. III. Bioavailability of salicylamide administered in tablets and chewing gum. Farmaci. Sci. 34
Ed. 16: 6–14. 35
Clark, J.C. and Shen, C.A. 2004. Fast flavor release coating for confectionery. US Patent 0175489 A1. 36
Collins, L.M.C. and Dawes, C. 1987. The surface area of the adult human mouth and thickness of the salivary film
37
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, 38
route of administration, container, closure and administration devices, III/3593/91. 39
Curatolo, W. 1987. The lipoidal permeability barriers of the skin and alimentary tract. Pharm. Res. 4: 271–277. 40
De Vries, M.E, Boddé, H.E. and Junginer, H.E. 1991. In vitro and in vivo adhesion performance of buccal 41
mucoadhesive patches. In thesis: De Vries, M. Buccal drug absorption and development of mucoadhesive poly-
42
mer systems, State University of Leiden, The Netherlands.
Dobrozsi, D.J. 2003. Oral liquid mucoadhesive compositions. US Patent 6638521. 43
Donbrow, M. and Freidman, M. 1974. Permeability of films of ethyl cellulose and PEG to caffeine. Commun. 44
J. Pharm. Pharmac. 26: 148–150. 45
Eichman, J.D. 1997. Mechanistic studies on effervescent permeability enhancement; PhD dissertation, University S46
of Wisconsin-Madison.
N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 198

198 Chapter 8

1 Evered, D.F. and Mallett, C. 1983. Thiamine absorption across human buccal mucosa in vivo. Life Sci. 32:
2 1355–1358.
Evered, D.F, Sadooght-Abasian, F. and Patel, P.D. 1980. Absorption of nicotinic acid and nicotinamide across
3 human buccal mucosa in vivo. Life Sci. 27: 1649–1651.
4 Fernö, O. 1973. A substitute for tobacco smoking. Psychopharmacologia 31: 201–204.
5 Fromter, E. and Diamond, J. 1972. Route of passive ion permeation in epithelia. Nature New Biol. 235: 9–13.
6 Gandhi, R.E. and Robinson, J.R. 1988. Bioadhesion in drug delivery. Ind. J. Pharm. Sci. 50: 145–152.
7 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
8 release of caffeine in chewing gum. US Patent 6165516.
9 Guo, J-H. 1994. Investigating the surface properties and bioadhesion of buccal patches. J. Pharm. Pharmacol. 46:
10 647–650.
11 Haines, H.G. 2004. A pilot study evaluating the bioavailability and absorption rates of two vitamin B12 prepara-
12 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:
13 1–10.
14 Hayward, A.F. 1979. Membrane-coating granules. Int. Rev. Cytol. 59: 97–127.
15 Hutchinson, K.G. and Garnett, K.R. 1999. Method of making water-based beverages. US 5871789.
16 Jacob, J.S., Mathiowitz, E., Nangia, A., Shaked, Ze’ev and Moslemy, P. 2006. Controlled regional oral delivery. US
17 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:
18 786–790.
19 Johnson, S. and Yatka, R.J. 2000. Method of making chewing gum products containing perillratine. US patent
20 6159509.
21 Johnson, S., Sheldon, G. and Greenberg, M.J. 2004. Oral products containing novel flavor composition. US Patent
22 0141927.
Kaaber, S. 1973. Studies on the permeability of human oral mucosa VI. The mucosal transport of water, sodium and
23 potassium under varying osmotic pressure. Acta Odont. Scand. 31: 307–316.
24 Kaaber, S. 1974. The permeability and barrier functions of the oral mucosa with respect to water and electrolytes.
25 Acta Odontol. Scand. 32 (Suppl 66): 26.
26 Kahn, C.R. and Shechter, Y. 1991. “Oral hypoglycemic agents and the pharmacology of the endocrine pancreas”.
27 In Theodor W.R., Alan, S.N., Taylor, P. and Gilman, A.G., (Eds) Goodman and Gilman’s The Pharmacological
Basis of Therapeutics, 8th ed., New York, NY: McGraw Hill: 1484.
28 Kamimori, G.H., Karyekar, C.S., Ottersetter, R., Cox, D.S., Balkin, T.J., Belenky, G.L. and Eddington, N.D. 2002.
29 The rate of absorption and relative bioavailability of caffeine administered in chewing gum versus capsules to
30 normal healthy volunteers. Int. J. Pharm. 234(1–2): 159–167.
31 Kanwar, Y.S., Linker, A. and Farquhar, M.G. 1980. Increased permeability of the glomerular basement
32 membrane to ferritin after removal of glucosaminoglycans (heparin sulfate) by enzyme digestion. J. Cell Biol.
688–693.
33 Karles, G., Garthaffner, M., Juper, R., Kellog, D. and Skinner, I. 2006. Flavor capsule for enhanced flavor delivery
34 in cigarettes. US Patent 082529 A2.
35 Kaufmann, K. 2003. Hard candy with plaque-neutralizing effect comprising alkali metal monophosphate. US
36 Patent 6582731 B1.
37 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-
38 nologies. Paper presented at the Controlled Release Society Meeting, July 25, 2006, Vienna, Austria.
39 Lakkis, J. and Wong, S. 2007. Multilayered confectionery product with controlled release teeth remineralization
40 actives (patent submitted).
41 Leach, S.A., Lee, G.T.R. and Edgar, W.M. 1989. Remineralization of artificial caries-like lesions in human enamel
42 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:
43 40–45.
44 Lesch, C.A., Squier, C.A., Cruchley, A., Williamson, D.M. and Speight, P. 1989. The permeability of human oral
45 mucosa and skin to water. J. Dent. Res. 68: 1345–1349.
46S Lindqvist, L. and Augustinsson, K.B. 1975. Esterases in human saliva. Enzyme 20: 277–291.
47N Lombardy, C.M., Lombardy, D.R. and Liebrecht, J.W. 2001. Effervescent chewing gum. US patent 6235318 B1.
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 199

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 199

Macknight, A.D.L. and Leader, J.P. 1983. Intra- and trans-epithelial analytical techniques. CRC Crit. Rev. Clin. Lab. 1
Sci. 18: 339–395. 2
Manning, A.S. and Evered, D.F. 1976. The absorption of sugars from the human buccal cavity. Clin. Sci. Molec.
Med. 51: 127–132.
3
Mazurek, P.M., Reed, M. and Broderick, K. 2000. Chewing gum with dental benfits including calcium in a food 4
grade acid. WO 00/62762. 5
McGrew, G.N., Maxwell, J.R., Tyrpin, H., Greenberg, M.J. and Record, D.W. 2006. Chewing gum that provides 6
breath freshening characteristics. US patent 7087255 B2. 7
Merritt, C., Wingerd, W.H. and Keller, D.J. 1985. Encapsulated flavorant material, methods for its preparation and
food and other compositions incorporating the same. US Patent 4515769.
8
Mulry, J.T. 1988. Nicotine gum dependency: A positive addiction. Drug Intell. Clin. Pharm. 22: 313–314. 9
Nicolazzo, J.A., Reed, B.L. and Finnin, B. 2003. Assessment of the effects of sodium dodecyl sulfate on the buccal 10
permeability of caffeine and estradiol. J. Pharm. Sci. 93(2): 431–440. 11
Nuernberg, B. and Brune, K. 1989. Buffering the stomach content enhances the absorption of diflunisal in man, 12
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-
13
brane. Int. J. Vitam. Nutr. Res. 47: 135–144. 14
Ozaki, Y. and Miyake, T. 1995. Pullulan binder and its uses. US Patent. 5411945. 15
Paetau, I., Rao, D., Wiley, E.R., Brown, E.D. and Clevidence, B.A. 1999. Carotenoids in human buccal mucosa cells 16
after 4 wk of supplementation with tomato juice or lycopene supplements. Am. J. Clin. Nutr. 70: 490–494. 17
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.
18
Pather, S.I., Robinson, J.R., Eichman, J.D., Khankai, R.K., Hontz, J. and Gupta, S.V., 2002. Effervescent drug 19
delivery systems for oral administration. US Patent 6391335. 20
Pedersen, M. and Rassing, M.R. 1990. Miconazole and miconazole nitrate chewing gum as drug delivery 21
systems—a practical application of solid dispersion technique. Drug Dev. Ind. Pharm. 16(1): 55–74. 22
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
23
20030313 A1. 24
Ream, R.L., Corriveau, C.L., Graff, G. and Matulewicz, L. 2001. Over-coated chewing gum formulations including 25
tabeleted center. US Patent 6322806 B1. 26
Rivier, V. 2005. Confectionery product containing functional ingredients. EP 1280414 B1. 27
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:
28
1029–1034. 29
Rutherford, H.J., Desai, N., McDermott, K. and Wiener, C. 1992. Chewing gum containing compositions for con- 30
trolled release of flavor-bearing substances and process for producing same. EP 0502750 A1. 31
Sadoogh-Abasian, F. and Evered, D.F. 1979. Absorption of vitamin C from the human buccal cavity. Br. J. Nutr. 42: 32
15–20.
Sadoogh-Abasian, F. and Evered, D.F. 1980. Absorption of nicotinic acid and nicotinamide across human buccal
33
mucosa. Life Sci. 27: 1649–1651. 34
Savage, W.D., Schnell, P.G., Aumann, R.A. and Yatka, R.J. 2002. Chewing gum formulations including encapsu- 35
lated aspartame and sodium phosphate. US Patent 0142068 A1. 36
Schanker, L.S. 1964. Physiological transport of drugs. Adv. Drug Res. 1: 71–106. 37
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.
38
Sharma, S.C. and Yang, K.Y. 1986. Chewing gum compositions containing novel sweetener delivery systems and 39
method of preparation. US patent 4597970. 40
Siegel, I.A. 1984. “Permeability of the oral mucosa”. In Meyer, J., Squier, C.A. and Gerson S.J. (Eds) The Structure 41
and Function of Oral Mucosa. Oxford, Pergamon Press. 95–108. 42
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. 274–286
43
Silagy, C., Lancaster, T., Stead, L., Mant, D. and Fowler, G. 2002. Nicotine replacement therapy for smoking cessa- 44
tion. Cochrane Database Syst. Rev. 4: CD000146. 45
Song, J.H. and Copper, C.M. 1992. Gradual release structures for chewing gum. US patent 5165944. S46
Song, J.H. and Courtwright, S.B. 1992. Flavor releasing structures for chewing gum. US Patent 5128155. N47
Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 200

200 Chapter 8

1 Song, J.H., Sundstrom, E., Record, D., Townsend, D., Broderick, K.B. and Schnell, P. 1992. Gum composition
2 containing 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 as
3 related to the thickness of the permeability barrier. J. Invest. Dermatol. 84: 176–179.
4 Squier, C.A., Johson, N.W. and Hopps, R.M. 1976. Human Oral mucosa: Development, Structure and Function.
5 Blackwell Scientific, Oxford.
6 Squier, C.A. and Kremer, M.J. 2001. Biology of oral mucosa and esophagus. J. Natl. Cancer Inst. Monograph 29:
7 7–15.
Squier, C.A. and Wertz, P.W. 1996. Structure and function of the oral mucosa and implications for drug delivery,
8 pp. 1–26, in Oral Mucosal Drug Delivery, Rathbone, M.J. (Ed.), Marcell Dekker, Inc. New York, NY.
9 Stevenson, N.R. 1974. Active transport of L-ascorbic acid in the human ileum. Gastroenterology 67: 952–956.
10 Sunohara, H., Kamaguchi, R., Kagawa, M., Nishikawa, M. and miura, Y. 2002. Multilayered soft capsule for elimi-
11 nating bad breath and process for producing the same. US Patent 6426089 B1.
12 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.
13 Tabak, L.A., Levine, M.J., Mendell, I.D. and Ellison, S.A. 1982. Role of salivary mucins in the protection of the
14 oral cavity. J. Oral Pathol. 11: 1–7.
15 Tan, S.C. 1976. Human saliva esterases. Genetic studies. Human Hered. 26: 207–216.
16 Tanner, K. and Shelley, R.S. 1996. Fill compositions for soft elastic gel capsules. US patent 5569466.
17 Tucker, I.G. 1988. A method to study the kinetics of oral mucosal drug absorption from solutions. J. Pharm.
Pharmacol. 40: 679–683.
18 Tuovinen, L., Peltonen, S. and Järvinen, K. 2003. Drug release from starch-acetate films. J. Control. Release 91:
19 345–354.
20 Tutuncu, N.P., Hartunian, S., Bratton, J.A. and Stanton, K.J. 2003. Confectionery product having a salivation region
21 and an oral comfort region. US Patent 0118628 A1.
22 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.
23 Virgallito, M.T. and Zhang, J. 2006. Edible film containing food acid. US Patent 0035008 A1.
24 Weatherell, J.A., Robinson, C. and Rathbone, M.J. 1994. Site-specific differences in the salivary concentration of
25 substances in the oral cavity—implications for the etiology of oral disease and local drug delivery. Adv. Drug
26 Del. Rev. 13: 23–42.
27 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.
28 Weatherell, J.A., Strong, M., Robinson, C., Nakagi, H. and Ralph, J.P. 1989. Retention of glucose in oral fluid at dif-
29 ferent sites in the mouth. Caries Res. 23: 399–405.
30 Wennerholm, K. and Emilson, C.-G. 1989. Effect of sorbitol- and xylitol-containing chewing gum on salivary
31 microflora, saliva, and oral sugar clearance. Scand. J. Dent. Res. 97: 257–262.
32 Wertz, P.W. and Downing, D.T. 1983. Ceramides of pig epidermis: Structure determination. J. Lipid Res. 24:
759–765.
33 Winston, A. and Usen, N. 2002. Products and methods for the remineralization and prevention of demineralization
34 of teeth. US Patent 6451290 B2.
35 Witzel, F., Wayne, J.P., Warren, C., Donald, A. and Mackay, M. 1980. Chewing gum capable of releasing finely
36 divided water-insoluble materials therefrom. US Patent 4238475.
37 Wolf, F.R., Mcgrew, G.N. and Tyrpin, H.T. 2005. Chewing gum containing controlled release acyclic carboxamides.
US 0082271 A1.
38 Wolfson, P. 2002. Herbal composition for improving oral hygiene, for providing local anesthesia, for use as an oral
39 sensate, flavor enhancer and potentiator, and methods of using same. US patent 0122778 A1.
40 Wong, F.W., Yuen, K.H. and Peh, K.K. 1999. Formulation and evaluation of controlled release Eudragit buccal
41 patches. Int. J. Pharm. 178(1): 11–22.
42 Wonschik, J., Machinek, A. and Koschorreck, C. 2005. Coated spherical: Seamless filled capsules. WO
2005/077521 A1.
43 Woodford, W. and Lesko, L.J. 1981. Relative bioavailability of aspirin gum. J. Pharm. Sci. 70: 1341–1343.
44 Yang, J. H. 2005. Gelatin soft capsule having the properties of removal of oral smell and cleaning of oral cavity. US
45 Patent 6852309 B2.
46S
47N
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 201

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

9 Innovative Applications 1
2
of Microencapsulation in Food Packaging 3
4
5
Murat Ozdemir and Tugba Cevik 6
7
8
9
10
11
12
Introduction 13
The use of proper packaging materials and methods to minimize food losses to provide safe 14
and wholesome food products has always been the focus of food packaging. In addition, 15
consumer demands for better-quality, fresh-like and convenient products have been intensi- 16
fying in the last two decades. A wide variety of packaging materials and technologies have 17
been developed to meet these consumer requirements and to limit package-related environ- 18
mental pollution and disposal problems (Ozdemir and Floros, 2004). Despite these advances 19
and availability of unique materials such as plastics that can be specifically designed to delay 20
adverse effects of the environment on food products and to extend their shelf-life, novel 21
approaches to the development of packaging materials containing microencapsulated active 22
particles have emerged recently. 23
Encapsulation is a technique by which a material or a mixture of several materials can be 24
coated or entrapped in another material. The development of a successful microencapsu- 25
lated product primarily depends on: 26
27
1. selecting an appropriate shell formulation, usually GRAS (generally recognized as safe) 28
materials that are approved by the Food and Drug Administration (FDA) or other inter- 29
national health authorities; 30
2. selecting an appropriate process to provide the desired functionality, stability, and 31
release mechanism; 32
3. economic feasibility of large-scale production including capital, operating costs, and 33
other miscellaneous expenses. 34
35
An appropriate shell formulation must stabilize the core material, must not react with or 36
deteriorate the active agent, yet releases it under specific conditions based on the product 37
application. Polysaccharides, proteins, waxes, fatty acids, gums and their derivatives are 38
common shell materials that are approved for food use. 39
Microencapsulation of food ingredients can be achieved by either physical or chemical 40
methods. Physical methods include extrusion, fluidized bed, spinning disc, and spray drying. 41
Chemical methods include coacervation, gelation, phase separation, and molecular inclu- 42
sion. Microcapsules can be produced by depositing a thin polymer coating on small solid 43
particles or liquid droplets, or by the process of dispersion of solids in liquids. The core 44
material or the active agent may be released by friction, pressure, diffusion through the poly- 45
mer wall, dissolution of the polymer wall coating, or by biodegradation. S46
N47

201
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 202

202 Chapter 9

1 Microencapsulated Actives for Packaging Applications


2
3 Microencapsulation can most often extend the shelf-life of foods while improving their
4 nutritional quality, appearance, and in various instances inhibiting the growth of pathogenic
5 and spoilage microorganisms, thus ensuring food safety. Important examples of microen-
6 capsulation in food packaging include incorporation of antimicrobial agents, insect and/or
7 rodent repellents, scented fragrance-inserts and flavor-releasing systems, pigments, inks,
8 and time–temperature indicators.
9
10 Antimicrobial Food Packaging Materials
11
12 Traditional food protection techniques include curing, smoking, or pickling which were
13 primarily effective in changing the moisture content or water activity of the foods. In recent
14 years, more sophisticated preservation methods have been developed to extend shelf-life of
15 foods. Figure 9.1 shows an example of novel microcapsules that can deliver preservatives
16 from plastic films or edible coatings that are currently available. Changing lifestyles and
17 the limited time available for food preparation require an increasing variety of high-quality,
18 safe, nutritious, and convenient food products today.
19 Allyl isothiocyanate is an effective inhibitor against various pathogens, particularly
20 Escherichia coli O157:H7. Consumption of undercooked ground beef has been identified
21
22
Active agent released
23 Plastic film Food
from the microcapsule
24
25
26 Core containing
27 active agent
28
29
30
31 Microcapsule
32
33 (a)
34
35 Plastic film Edible coating Food Active agent released
36 from the microcapsule
37
38
Core containing
39 active agent
40
41
42
43 Microcapsule
44
45
(b)
46S
47N 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 203

Innovative Applications of Microencapsulation in Food Packaging 203

as one of the main causes of E. coli O157:H7 outbreaks in North America (Waters et al., 1
1994). Chacon et al. (2006) microencapsulated allyl isothiocyanate in gum acacia and corn 2
oil prior to incorporating the preparation into aseptically treated chopped beef that was 3
inoculated with a known concentration of E. coli O157:H7. The system was packed under 4
nitrogen and stored under refrigeration (4°C). After 18 days, the chopped beef was found to 5
be free of E. coli O157:H7. Similarly, Klein et al. (2004) described a method for microen- 6
capsulating actives such as antibacterial and antifungal agents into a variety of polymeric 7
food packaging materials such as polyethylene, polypropylene, polyester, polycarbonate, 8
polyvinyl chloride, and polyvinylidene chloride. These films were described as especially 9
useful for controlling bacteria and fungi on food surfaces. Agar diffusion tests showed 10
that microencapsulated antibacterial agents were effective against Staphylococcus aureus, 11
E. coli, Pseudomonas aerugenosa, and Streptococcus spp. Thomas et al. (2005a, b) paten- 12
ted a method in which an antimicrobial agent was microencapsulated prior to being inte- 13
grated into the package structure via extrusion. The method was claimed to be effective in 14
providing thermal stability during package processing and manufacturing, but readily 15
released the active agent upon contact with moisture. 16
Avery Dennison Corp. (USA) developed an antimicrobial active label that releases trace 17
amounts of chlorine dioxide (ClO2) gas from the label. Chlorine dioxide is a broad spec- 18
trum antimicrobial agent effective against both bacteria and fungi. It can also be used to 19
eliminate odors and retard microbial growth on perishable food products, thus extending 20
their shelf-life. Laboratory tests showed that the inclusion of one small antimicrobial label 21
on the inside of rigid plastic packaging can significantly extend the shelf-life of fresh 22
berries. The time release delivery of the chlorine dioxide is moisture activated. The main 23
advantage of this system is that it does not require direct contact with the food. 24
A promising application of controlled release is in antimicrobial agents incorporated 25
into chewing gums for reducing the growth of microorganisms in the mouth and thereby 26
retarding tooth decay. Barabolak et al. (2005) produced a chewing gum with controlled- 27
release properties in which the antimicrobial agent (chlorhexidine digluconate) was encap- 28
sulated via film coating. Particles containing the encapsulated antimicrobial agent were 29
claimed to be adaptable to produce fast or delayed release when the gum is chewed. 30
Microencapsulating properties of whey proteins have been investigated extensively in 31
recent years (Ozdemir and Floros, 2001; Rosenberg, 1997). Whey protein concentrate and 32
whey protein isolate have been shown to exhibit excellent microencapsulating properties 33
for both volatile and non-volatile core materials (Young et al., 1993a; Lee and Rosenberg, 34
2000). The high microencapsulation yield of whey proteins is presumed to be a result of 35
their efficient emulsifying capacity, especially in the presence of carbohydrates (Young 36
et al., 1993b). Films and coatings from whey not only degrade more readily than polymeric 37
materials, but also could supplement the nutritional value and improve the sensory 38
attributes of coated foods. Ozdemir and co-workers (Ozdemir, 1999; Ozdemir and Floros, 39
2001) developed active antimicrobial films made of whey protein isolate, sorbitol, 40
beeswax, and potassium sorbate. The mechanism and release profile of potassium sorbate 41
in these films were found to follow non-Fickian diffusion model. A mathematical model 42
derived from Fick’s second law of diffusion with a time-dependent diffusion coefficient was 43
used to analyze potassium sorbate diffusion. Subsequent analysis showed that diffusion 44
coefficients of potassium sorbate in whey protein films were ten-fold higher than those in 45
edible wheat gluten and low density polyethylene (LDPE) films, and ten-fold lower than S46
those in intermediate moisture foods. N47
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 204

204 Chapter 9

1 In a recent study, Ozdemir and Floros (2003) investigated the effect of different con-
2 stituents (levels of whey protein, plasticizer, wax, lipid, and antimicrobial agent as encapsu-
3 lant) on the diffusivity of potassium sorbate using mixture response surface methodology.
4 Their study showed that increasing whey protein concentration in the film decreased
5 potassium sorbate diffusivity. Sorbate diffusion increased with increasing sorbitol concen-
6 tration but decreased with increased concentration of beeswax in the film. A rise in the ini-
7 tial active (potassium sorbate) concentration in the film resulted in higher diffusion
8 coefficients. Strong interactions were observed between beeswax and potassium sorbate,
9 and whey protein and beeswax.
10
11
12 Insect and/or Rodent Repellent Food Packages
13 Insect infestation of produce and food products results in spoilage and subsequent eco-
14 nomic losses. Controlling insect infestation has generally been achieved by fumigation
15 with methyl bromide. Methyl bromide is a toxic substance that can adversely affect the
16 human central nervous and respiratory systems if present at high concentrations. Methyl
17 bromide is also known to be a major contributor to the depletion of Earth’s ozone layer. One
18 way to overcome the disadvantages of methyl bromide is to find less toxic and less harmful
19 insect repellents and to incorporate them into packaging materials to form packages with
20 controlled-release properties.
21 Microencapsulation of pesticides, herbicides, and other pest control agents has been an
22 active area of development. Pest control agents are currently microencapsulated to prolong
23 their activity while reducing mammalian toxicity, volatilization losses, phytotoxicity, and
24 environmental degradation. Spector (1981) introduced a low-cost, self-stick tab in which an
25 active agent-saturated pad can be enveloped within a perforated sac which can then be
26 adhered to a desired site to release the active over an extended period of time. The active agent
27 can be an insect or animal repellent or a fragrance (Figure 9.2), and the system can be stuck
28 directly onto food packages or boxes to prevent their infestation by insects and animals.
29 The protection of agricultural products with biopesticides has been promoted recently as
30 a means of reducing the adverse effects of chemical insecticides (Marrone, 1999). Develop-
31 ment cost, time and ease of registration, and potential growing markets make biopesticides
32 popular over chemical pesticides (Brar et al., 2006). A number of biopesticides (bacteria,
33
34 Core containing
35 repellent material
36
37
38
39
40 Microcapsule wall
41
42
43
44 (a) (b)
45 Figure 9.2. Description of the action of a system composed of microcapsules containing
46S a repellent material: (a) microcapsule core containing a repellent, and (b) diffusion and
47N evaporation of the repellent material through the wall.
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 205

Innovative Applications of Microencapsulation in Food Packaging 205

fungi, virus, pheromones, plant extracts) have been already in use to control various types of 1
insects responsible for the destruction of agricultural crops. Bacillus thuringiensis-based 2
biopesticides are especially important and are estimated to constitute almost 97% of the 3
world biopesticide market (Cannon, 1993). 4
A biological pesticide is effective only if it has a potential major impact on the target 5
pest, market size, cost- effective and is capable of overcoming a number of technological 6
challenges such as fermentation, formulation and delivery systems. Biopesticides have 7
been encapsulated in a coating made of gelatin, starch, cellulose, or other polymers, and 8
even microbial cells (Barnes and Cummings, 1987a, b; Barnes and Edwards, 1989). Bok 9
et al. (1993) showed that encapsulated microbial pesticides possess excellent adhesiveness; 10
therefore, they can be applied directly to the soil or the plant. When the encapsulated 11
biopesticide is embedded into a plastic film, the film can be applied near the roots or cuts of 12
the crops to protect them against pathogens upon storage or during transportation. 13
Another recent advance in encapsulation is the production of hydrocapsules that are 14
water-based shellcores, consisting of a polymer membrane surrounding a liquid center. 15
These shells can be produced using UV radiation-initiated free-radical copolymerization of 16
functionalized prepolymers (silicones, urethanes, epoxys, polyesters, etc.) and/or vinyl 17
monomers such as acrylates for better dispersion and UV radiation protection (Lechelt- 18
Kunze et al., 2000; Toreki et al., 2004). 19
El-Rehim et al. (2005) formulated a polyacrylamide/polyethylene oxide hydrogel to 20
encapsulate and cross-link bioactives such as Atrazine. The active was incorporated into 21
the hydrogel matrix via electron beam irradiation process. Results showed that copolymer 22
blend composition, its gel content, and irradiation dose greatly affected the Atrazine release 23
rate. In addition, Atrazine release rate was found to decrease with increasing pH but 24
increased at high temperatures. 25
Packaged products are also susceptible to infestation by many insects and mites which 26
are capable of perforating the packaging material or use existing holes or openings in the 27
food package for penetration. Rieth et al. (1986) encapsulated 2-heptanone, an insect repel- 28
lent for bees and other insects, in a polyvinyl chloride–polyvinyl acetate plastic. Jones and 29
Hill (1982) added naphthalene flakes and citronella oil in solid form to synthetic resins 30
such as polyethylene, polypropylene, and polystyrene to form insect- and animal-repellent 31
plastic films. The resultant films were shown to have lower tensile and tear strengths than 32
films made without the actives (insect and animal repellent additives). 33
Atkinson (1991) described a microencapsulation process for manufacturing animal- 34
repellent plastic films where terpenes were incorporated into linear low-density polyethyl- 35
ene (LLDPE) melt via extrusion. Radwan and Allin (1997) developed a controlled release 36
insect repellent device that was described as useful for foods, tobacco, and other consum- 37
able items with the active being an essential oil. Navarro et al. (2005) produced a pest- 38
impervious packaging material by combining ar-turmerone, sesquiterpene alcohols, and/or 39
turmeric oleoresin solid residue. These materials were incorporated into plastics, adhe- 40
sives, or printing inks via microencapsulation. 41
42
43
Scented Fragrance-Inserts and Flavor-Releasing Systems
44
Food packaging materials, particularly plastics, may interact with food flavors, resulting in 45
loss of flavors, known as flavor scalping, therefore the need to replace these lost flavor S46
constituents. Although the use of high-barrier plastics holds food flavors in the package, N47
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 206

206 Chapter 9

1 additional flavor-releasing systems may be necessary in some instances, particularly when


2 heat seal layers of a package have high affinity to flavors. In addition, consumers always
3 like to smell pleasing flavors when they first open a food package (Brody, 1992). Flavor-
4 releasing systems are highly popular in the beverage industry, microwavable foods, and
5 coffee makers. The beverage industry, particularly the soft drink segment, is highly com-
6 petitive. Manufacturers take great care and make substantial efforts to formulate their prod-
7 ucts in such a way to differentiate them from their competitors’ and to make consumption
8 of the food product more enjoyable for the consumers. In the soft drink industry, despite the
9 high contribution of taste to the overall soft drink experience, its aroma especially when the
10 bottle is first opened is equally important. Ashcraft and Wong (1993) invented a package
11 that releases a burst of flavor when the package is opened. The novel flavor-burst structure
12 comprised a multilayer film with a flavor-carrier layer disposed between the barrier layers.
13 Due to their chemical incompatibility, the flavor agent desorbs from the carrier when one of
14 the barrier layers is removed from the carrier. Sun et al. (2000) developed a flavored poly-
15 ethylene terephthalate (PET) packaging system where the aroma is released once the bottle
16 cap was opened. Unfortunately, typical microencapsulated materials do not adhere well to
17 PET; thus the surface of PET bottles must be modified before the microencapsulated mate-
18 rial is applied to the bottles. A successful way to resolve this problem is by treating the
19 surface with a primer that enables the microencapsulated material to adhere to PET or to
20 roughen the surface using laser etching.
21 The use of overwraps on packages can improve the appearance and maintain the quality of
22 materials within the package. The use of a tear strip having the ability to release fragrance at
23 the time of opening of the package can add further benefit to the overwrap; for example,
24 release of a controlled fragrance can give the impression to the user that the ingredients of the
25 package are fresh. Fraser (1988) described a package that releases a fragrant liquid from
26 microcapsules when a tear strip is removed from the package. In this system, the separation of
27 multilayer sheet materials ruptures the microcapsules fitted to the intermediate adhesive lay-
28 ers and subsequently releases the entrapped fragrance. The described packages can be made
29 of paper, cardboards, polymeric materials, coated paper, foil, composite structures, metal-
30 lized paper, and so on. In a similar application, Sprinkel and Newsome (1988) microencapsu-
31 lated an aromatic substance in a cigarette package–overwrap where the aromatic substance is
32 released upon pulling the tear strip. This mechanism was described as useful for releasing
33 aroma of freshness or for adding flavorings to the cigarettes in the package.
34 CSP Technologies (US) developed an aroma emitting and aroma absorbing package in
35 which the active agent was encapsulated within three component plastic system. Disperse
36 Technologies (UK) combined a patented thin film–encapsulating technology with ultravio-
37 let curing technology to produce films and coatings that have controlled-release properties
38 (Wheeler, 2001). These films and coatings were claimed to possess a very long-lasting
39 impact for up to 6 months. Arcade Marketing (based in the United States) commercialized
40 the MicroFragrance label for foods, especially to promote the sales of low calorie cereal
41 bars. MicroFragrance label is printed onto a clear film so that it does not wear down or
42 blend with another smell such as paper.
43 Driscoll Labels (US) customized long-lasting, scratch-and-sniff labels for the fragrance
44 and food industries. This technology allows the consumer to perceive the aroma of the food
45 without opening the package. Scentisphere developed a printable, scented ink known as
46S rub-and-sniff ink that can be printed directly onto packaging materials. The rub-and-sniff
47N ink is claimed to have advantages over the traditional scratch-and-sniff labels since the
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 207

Innovative Applications of Microencapsulation in Food Packaging 207

Encapsulated flavors 1
released into the 2
package contents 3
4
5
6
7
8
9
10
11
12
13
Encapsulated
flavors released
Package 14
into the air 15
16
Figure 9.3. Release of microencapsulated flavors from flavor-incorporated plastics. 17
18
19
scented inks can be added to the ink using standard printers without any interruption in the 20
printing process or the inconvenience of having the ink drying quickly. 21
The US-based company ScentSational Technologies™ is a pioneering company in micro- 22
encapsulated packaging. A schematic of their CompelAroma® flagship technology for 23
encapsulating flavors into plastic-based packaging films is described in Figure 9.3. Using 24
this technology, food grade aromas/flavors can be embedded directly into plastic films dur- 25
ing manufacturing so that they form an integral part of the package. This technology is 26
claimed to be applicable to most existing manufacturing methods, including blow molding, 27
injection molding, thermoforming, and extrusion, and in gaskets and liners. Com- 28
pelAroma® technology can be used in containers, trays, cups, closures, bottles, and flexible 29
packages. The first commercial application of CompelAroma® technology was in Aquaes- 30
cents® refillable water bottles marketed by NutriSystem™ (USA). 31
Microencapsulated flavors and aromas have also been adapted for microwave and frozen 32
foods packaging containers. Yeo et al. (2005) encapsulated flavor oil in complex coacer- 33
vates using gelatin and gum Arabic. The resultant microcapsules were incorporated into 34
packaging films used for frozen or baked foods such as breads, pastries, pizzas, and cookies 35
to improve their appeal and release the flavor oil of interest during heating. 36
37
38
Microencapsulated Pigments
39
Coloring agents containing natural or synthetic substances are commonly used as additives 40
in the manufacture of food products. Commercially available coloring agents can contain 41
synthetic substances including dyes or azodyes or natural pigments. It is a well-known 42
problem in the food industry that coloring agents tend to migrate within the food product or 43
into the environment of the product. This problem is particularly troublesome if it occurs in 44
food products that comprise multiple, separated compartments or layers where the coloring 45
agent is not added to all of such compartments. One typical class of such a compartmental- S46
ized or layered food product is cakes and other desserts, which comprises at least one layer N47
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 208

208 Chapter 9

1 of fruit filling, e.g., in the form of jelly, to which a coloring agent is added, and one or more
2 layers of other ingredients also having an aqueous phase to which the coloring agent is not
3 added. It is evident that migration of coloring agent into the non-colored layers results in a
4 highly unacceptable appearance of these layered products. Another important product in
5 which the migration of coloring agent is not desirable is surimi. Surimi is a stabilized,
6 myofibrillar protein system prepared from fish mince that has been washed with water and
7 blended with cryoprotectants (Park and Morrissey, 2000). Surimi color is a very important
8 attribute to the product’s overall quality since it is commercially graded based on its color.
9 Surimi loses its color even if it is stored at freezing temperature. Floros et al. (1997) dis-
10 cussed the use of coloring agent–containing film in surimi systems as an alternative to tra-
11 ditional direct color addition and to avoid migration of coloring agent from the film to the
12 product. Shahidi and Pegg (1995) described a process in which the coloring agent was
13 encapsulated within a mixture of carbohydrate-based wall material, a binding agent, and a
14 reducing or sequestering agent to improve color stability of surimi as well as other meat
15 products. The encapsulated pigment was reported to be effective by imparting the typical
16 cured color to frankfurters even after 18 months of storage. Popplewell and Porzio (2001)
17 encapsulated various coloring agents in partially hydrogenated vegetable oil as a means of
18 incorporating them into edible coating for snacks, chicken legs, fish, and similar products.
19 In human nutrition, astaxanthin (reddish-orange pigment) has been gaining widespread
20 popularity as a dietary supplement due to its powerful antioxidant properties. As most
21 carotenoids, astaxanthin is a highly unsaturated molecule and thus can easily be degraded
22 by thermal or oxidative processes during the manufacture and storage of foods. This degra-
23 dation can cause the loss of their nutritive and biological value as well as production of
24 undesirable flavor or aroma compounds. Due to their intrinsic high instability, these com-
25 pounds are not usually handled in their crystalline form, but rather as stabilized emulsions
26 or microcapsules. Higuera-Ciapara et al. (2002) microencapsulated synthetic astaxanthin
27 in a chitosan matrix cross-linked with glutaraldehyde using the method of multiple emul-
28 sion/solvent evaporation. A powdered product was obtained containing microcapsules with
29 a diameter of 5–50 µ. Stability of the pigment in the microcapsules was studied under stor-
30 age at 25°C, 35°C, and 45°C for 8 weeks by measuring isomerization and loss of concen-
31 tration of the pigment. Results showed that the microencapsulated pigment did not undergo
32 isomerization or other chemical degradation under the investigated storage conditions.
33
34
Microencapsulated-Inks and Time–Temperature Indicators
35
36 Sakojiri and Takahashi (1990) developed a multicolor imaging material that comprised a
37 substrate and a photosensitive layer. The latter consisted of a heat-meltable microcapsule
38 layer and a color forming layer comprising a diazonium compound and a basic substance. The
39 imaging material can be coated onto paper and polymeric films, and the final product can be
40 used as a food packaging material. Tajiri et al. (1992) formulated microcapsules containing
41 ink for flexographic applications in which encapsulation of the ink ensures its adhesion and
42 flowability. Resins used for this application consist of methacrylate or acrylate of molecular
43 weight of 3000 up to 50,000 g/mol. The microcapsule containing ink compositions for flexo-
44 graphic printing were described to be particularly useful for perfume ink compositions.
45 Chul et al. (2005) prepared an encapsulated color electronic ink by in situ polymeriza-
46S tion utilizing urea/melanine and formaldehyde resin as wall materials. The electrophoretic
47N
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 209

Innovative Applications of Microencapsulation in Food Packaging 209

medium was made of two different types of white and colored pigment particles, bearing 1
charges of opposite polarity, in a colorless dielectric fluid. Optical contrast was achieved by 2
moving charged particles separately to the opposite electrode. White or colored particles 3
held electrostatically to the common electrode depending on the electric field and the 4
particle charge. The white particles modified with polymethylmethacrylate copolymer and 5
the colored ones (magenta, yellow, and cyan) modified with wax material were found to 6
have superior affinity for the suspending medium. 7
More sophisticated applications of microencapsulated inks and coloring agents can be 8
found in the area of time–temperature indicator applications. Because temperature abuse is 9
common during storage, transportation, and handling, these indicators are designed to 10
monitor temperature abuses in the shelf-life of food products. Temperature abuse does not 11
only cause quality and nutritional losses, but also may lead to food poisoning and food 12
losses (Ozdemir and Floros, 2004). In these systems, polymers that contain irreversible 13
thermochromic dyes change color in response to exposure to predetermined temperature 14
over time. These indicators can be formed into labels that can irreversibly change color and 15
warn the consumer when a product has been exposed to a temperature/time abuse. Success- 16
ful time–temperature indicators must satisfy basic requirements to be effective as monitor- 17
ing devices (Selman, 1995): 18
19
1. They must be easily activated and sensitive. 20
2. They must provide high degree of accuracy and precision. 21
3. They should have tamper-proof and should not be removed from the package. 22
4. Response should be irreversible, reproducible, and should correlate with food quality 23
changes. 24
5. Response should be easily readable and not be confusing. 25
6. Physical and chemical characteristics of time–temperature indicators should be determined. 26
27
Recently, Avery Dennison Corp. (US) introduced a new time–temperature indicator in 28
which TT Sensor™ is employed. The TT Sensor™ consists of two labels: an indicator label 29
and a transparent activator label. The activator label is applied to the indicator label and then 30
immediately dispensed onto the package. Once the time–temperature indicator is activated, 31
the indicator label immediately and irreversibly changes color. Activated labels function in 32
the temperature range from –18°C to 60°C. These indicators are claimed to be simple, reli- 33
able, and cost-effective for monitoring time–temperature abuse that fresh foods are normally 34
exposed to. In addition, TT Sensor™ labels do not need to be refrigerated prior to applica- 35
tion. Another time–temperature indicator that uses microencapsulation technology is called 36
Thermax™. By measuring the change in temperature that is reflected in irreversible color 37
changes, the latter indicator shows whether a food product was exposed to extreme tem- 38
peratures, and this indicator is tamperproof. Similarly, Prusik et al. (2000) patented a 39
time–temperature indicator label to measure the length of time to which a food product is 40
exposed under temperature abuse conditions. The label contained a microencapsulated heat- 41
fusible substance, which can melt and flow when a food product is exposed to temperatures 42
above a predetermined level. The indicator can be activated by light finger pressure or 43
preferably by appropriate automated mechanical means to rupture the capsule containing the 44
heat-fusible substance. A distinct color would develop upon contact of the dye precursor and 45
dye activator due to the migration of the heat-fusible substance. S46
N47
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 210

210 Chapter 9

1 Future Perspective
2
3 Microencapsulation has a promising future in food packaging. Attempts at microencapsu-
4 lating flavors, antimicrobials, fragrances, coloring materials, and printing inks into food
5 packaging materials will continue to increase in the next decade. There are significant
6 opportunities in microencapsulation for food packaging, such as self adjusting sell-by-date
7 that senses when the consumer opens a packaged food and flashes if the food in the package
8 is spoiled or poses a health risk for the consumer. Another potential commercial application
9 of microencapsulation in food packaging is package indicators that inform consumers if
10 the package is disposable or not. This is especially useful for the elderly and children. The
11 next step in encapsulation is the development of smart microcapsules that embody multiple
12 micro-compartments, each one dynamically interacting with the other, depending on the
13 changing environmental conditions. This is especially important to form packages that
14 have “self-pasteurizing” or “self-sterilizing” capabilities. In these systems, microencapsu-
15 lated active agents within a polymer matrix will be released in a controlled manner depend-
16 ing on the characteristics of the food such as microbial load, pH, and water as well as the
17 changing environmental conditions such as temperature, relative humidity, and so on to
18 achieve a homogeneous pasteurization or sterilization within the package.
19
20 References
21
22 Ashcraft, C. R. and Wong, M. M. L. 1993. Flavor burst structure and method of making the same. US Patent No.
5,249,676. USA.
23 Barabolak, R. M., Zibell, S. E., Witkewitz, D. L., and Greenberg, M. J. 2005. Method of controlling release of
24 antimicrobial agents in chewing gum. US Patent No. 6,955,827. USA.
25 Barnes, A. C. and Cummings, S. G. 1987a. Cellular encapsulation of biological pesticides. US Patent No.
26 4,695,462. USA.
27 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.
28 Barnes, A. C. and Edwards, D. L. 1989. Cellular encapsulation of biologicals for animal and human use. US Patent
29 No. 4,861,595. USA.
30 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
31 coated microbial pesticides and pesticides produced therefrom. US Patent No. 5,273,749, USA.
32 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.
33 Brody, A. L. 1992. Flavor, flavor everywhere-but in packaging? Cereal Foods World, 37(11): 834–835.
34 Cannon, R. J. C. 1993. Prospects and progress for Bacillus thuringiensis based pesticides. Pesticide Science, 37:
35 331–335.
36 Chacon, P. A., Buffo, R. A., and Holley, R. A. 2006. Inhibitory effects of microencapsulated allyl isothiocyanate
37 (AIT) against Escherichia coli O157:H7 in refrigerated, nitrogen packed, finely chopped beef. International
Journal of Food Microbiology, 107(3): 231–237.
38 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.
39 B., and Kyung, S. S. 2005. Microcapsules as an electronic ink to fabricate color electrophoretic displays. Syn-
40 thetic Metals, 151(3): 181–185.
41 El-Rehim, H. A. A., Hegazy, E. A., and El-Mohdy, H. L. A. 2005. Properties of polyacrylamide-based hydrogels
42 prepared by electron beam irradiation for possible use as bioactive controlled delivery matrices. Journal of
Applied Polymer Science, 98(3): 1262–1270.
43 Floros, J. D., Dock, L. L., and Han, J. H. 1997. Active packaging technologies and applications. Food, Cosmetics
44 and Drug Packaging, 20: 10–17.
45 Fraser, A. D. 1988. Package opening system. US Patent No. 4,720,423. USA.
46S Higuera-Ciapara, I., Felix-Valenzuela, L., Goycoolea, F. M., and Argüelles-Monal, W. 2002. Microencapsulation
47N of astaxanthin in a chitosan matrix. Carbohydrate Polymers, 56(1): 41–45.
Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 211

Innovative Applications of Microencapsulation in Food Packaging 211

Jones, L. R. and Hill, J. L. 1982. Composition for pest repellent receptacle. US Patent No. 4,320,112. USA. 1
Klein, R. B., Selph, J. L., Partridge, J. J., and Reinhard, J. 2004. Compounds and methods for controlling fungi, 2
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
3
embedded in hydrogels, a process for the embedding thereof, and its use as artificial seed. US Patent No. 4
6,164,012. USA. 5
Lee, S. J. and Rosenberg, M. 2000. Whey protein-based microcapsules prepared by double emulsification and heat 6
gelation. Lebensmittel-Wissenschaft und Technologie, 33: 80–88. 7
Marrone, P. G. 1999. Microbial pesticides and natural products as alternatives. Outlook on Agriculture, 28(3):
149–154.
8
Navarro, S., Finkelman, S., Zehavi, D., Dias, R., Angel, S., Mansur, F., and Rindner, M. 2005. Pest-impervious 9
packaging material and pest-control composition. US Patent Application No. 20,050,208,157. USA. 10
Ozdemir, M. 1999. Antimicrobial Releasing Edible Whey Protein Films and Coatings. Ph.D. Dissertation. Purdue 11
University, West Lafayette, IN. 12
Ozdemir, M. and Floros, J. D. 2001. Analysis and modeling of potassium sorbate diffusion through edible whey
protein films. Journal of Food Engineering, 47(2): 149–155.
13
Ozdemir, M. and Floros, J. D. 2003. Film composition effects on diffusion of potassium sorbate through whey pro- 14
tein films. Journal of Food Science, 68: 511–516. 15
Ozdemir, M. and Floros, J. D. 2004. Active food packaging technologies. Critical Reviews in Food Science and 16
Nutrition, 44(3): 185–193. 17
Popplewell, L. M. and Porzio, M. A. 2001. Fat-coated encapsulation compositions and method for preparing the
same. US Patent No. 6,245,366. USA.
18
Prusik, T., Arnold, R. M., and Fields, S. C. 2000. Time-temperature indicator device and method of manufacture. 19
US Patent No. 6,042,264. USA. 20
Radwan, M. N. and Allin, G. P. 1997. Controlled-release insect repellent device. US Patent No. 5,688,509. USA. 21
Rieth, J. P., Wilson, W. T., and Levin, M. D., 1986. Repelling honeybees from insecticide-treated flowers with 22
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
23
No. 5,601,760. USA. 24
Sakojiri, H. and Takahashi, H. 1990. Multicolor imaging material. US Patent No. 4,916,042. USA. 25
Selman, J. D. 1995. Time-temperature indicators. In: Active Food Packaging. pp. 215–237. M. L. Rooney (Ed.). 26
Blackie Academic and Professional, London. 27
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.
28
Sprinkel, Jr. F. M. and Newsome, R. W. 1988. Package with means for releasing aromatic substance on opening. 29
US Patent No. 4,717,017. USA. 30
Sun, R., Quintus-Bosz, H., Given, P., Pineiro, R., and Morrison, A. 2000. Aroma release bottle and cap. US Patent 31
No. 6,102,224. USA. 32
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.
33
Thomas, T. R., Belias, W. P., Chen, P. N., and Kolovich, N. A. 2005a. Packages with active agents. US Patent 34
Application No. 20,050,220,375. USA. 35
Thomas, T. R., Long, S. P., Belias, W. P., and Kolovich, N. A. 2005b. Packages with active agents. US Patent 36
Application No. 20,050,220,374. USA. 37
Toreki, W., Manukian, A., and Strohschein, R. 2004. Hydrocapsules and method of preparation thereof. US Patent
No. 6,780,507. USA.
38
Waters, J. R., Sharp, J. C., and Dev, V. J. 1994. Infection caused by Escherichia coli O157:H7 in Alberta, Canada, 39
and in Scotland: A five-year review, 1987–1991. Clinical Infectious Diseases, 19: 834–843. 40
Wheeler, D. A. 2001. Surface coatings. US Patent No. 6,312,760. USA. 41
Yeo, Y., Bellas, E., Firestone, W., Langer, R., and Kohane, D. S. 2005. Complex coacervates for thermally 42
sensitive controlled release of flavor compounds. Journal of Agricultural and Food Chemistry, 53(19):
7518–7525.
43
Young, S. L., Sarada, X., and Rosenberg, M. 1993a. Microencapsulating properties of whey proteins. 1. Microen- 44
capsulation of anhydrous milk fat. Journal of Dairy Science, 76: 2868–2877. 45
Young, S. L., Sarada, X., and Rosenberg, M. 1993b. Microencapsulating properties of whey proteins. 2. Combina- S46
tion of whey proteins with carbohydrates. Journal of Dairy Science, 76: 2878–2885. N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 213

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

10 Marketing Perspective of Encapsulation 1


2
Technologies in Food Applications 3
4
5
Kathy Brownlie 6
7
8
9
10
11
Introduction
12
From being the enabling technology behind carbonless paper and “scratch and sniff ” 13
fragrance sampling, microencapsulation technologies have found a broad range of indus- 14
trial applications in markets as diverse as pesticides and cosmetics. 15
Microencapsulation has also found widespread use in the food and beverage industry. 16
Flavorings that last longer, both in the mouth and on the shelf, fresher tasting flavorings 17
which are clearly distinguishable from each other, innovative new flavor combinations, fla- 18
vors which are released at the ideal time to provide maximum impact for the consumer, and 19
even products which initially taste of one thing, but then develop a totally different flavor as 20
you chew, all of these things are being achieved in food products through the innovative use 21
of microencapsulation techniques. 22
Flavor microencapsulation, for example, entraps tiny volumes of the flavoring substance 23
in a protective layer of another material, creating particles which are only in the micron or 24
even nano size range, but which can have a huge impact on the flavor profile of the final 25
product they are used in. This ability to isolate tiny amounts of a substance within a protec- 26
tive wall or matrix has played a crucial role in the development of novel groundbreaking 27
products, such as the temperature-regulating clothing sold by Outlast and Frisby Technolo- 28
gies and the electronic displays being developed by E Ink. 29
The combination of a constant flow of innovative new techniques with wider application 30
areas and an increased desire for product differentiation will continue to drive growth in the 31
use of microencapsulation technologies in various markets. But, technology providers must 32
choose their end application market with care to ensure sustained growth. Growth rates in 33
the end-use markets range from negative figures to as much as 30 percent. 34
Several potentially high-volume applications of the technology are still in development, 35
and the companies involved have strong intellectual property positions. In other markets, 36
the end-product manufacturers perform most of the microencapsulation themselves and 37
allow fewer opportunities for companies outside the industry. However, in areas such as the 38
cosmetics and in some food ingredient markets, most of the microencapsulation is per- 39
formed by smaller technology providers. 40
From a performance standpoint, encapsulation of active pharmaceutical ingredients 41
enables the formulator to design a release profile most appropriate for the drug. This 42
includes fast-dissolve formulas, extended release, targeted release, and delayed release. 43
Microencapsulation technologies also enable taste masking of bitter compounds in chew- 44
able tablets, as well as oral dosages that are taken without water. 45
In the pharmaceutical consumer product industries, oral drug delivery continues to S46
dominate the microencapsulated drug market. Although some of the microencapsulation N47

213
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 214

214 Chapter 10

1 technologies have existed for decades, the current trend is to explore drug delivery for
2 increasingly potent or insoluble new compounds or to add value to existing compounds.
3
4 Key Challenges
5
6 Identification of New Applications
7 Although the number of markets in which microencapsulation technology has been applied
8 is already large, the general feeling is that the surface of possible applications has only been
9 scratched. Companies offering this technology have to continue to invest in research and
10 monitor markets and emerging technologies where microencapsulation could add value to
11 products or solve particular problems. For companies possessing in-house microencapsula-
12 tion capabilities, internal communication between research departments is crucial in order
13 to fully utilize these resources. For companies offering microencapsulation services, this
14 can be a much harder task. It is important for such companies to develop a widely recog-
15 nized reputation as experts in this field, so that industry turns to them when projects which
16 might benefit from this expertise arise. Developing close relationships with R&D depart-
17 ments at various companies will be vital.
18
19
20 Raising Awareness of Technique Potential
21 Closely related to the identification of new applications for the technology is the education
22 of as wide a customer base as possible of the potential of microencapsulation technologies.
23 The primary approach for achieving this within the industry appears to be simply talking to
24 people about the possibilities and gaining a reputation as an effective and innovative
25 provider of such technology. This can be a laborious and slow process, so companies may
26 need to be creative in the ways in which they market their capabilities in this field.
27
28
29 Extra Costs Associated with the Use of Microencapsulation
30 The design of a microencapsulation process for a substance, the equipment needed to apply
31 it on an industrial scale, and subsequent production all add to the cost of products using
32 microencapsulated products. These issues tend to limit the use of microencapsulation tech-
33 nology to few markets: (i) those with higher value products where the cost added by
34 microencapsulation would have less impact, (ii) to those markets where microencapsula-
35 tion is absolutely necessary, or (iii) to a few markets where economies of scale can be
36 applied, thus reducing the importance of the fixed costs associated with microencapsula-
37 tion. In other markets, microencapsulation must be seen to provide a very clear added value
38 to the customer, so that they will be willing to pay a higher price. Although this might be
39 clear to the company doing the encapsulation, it is often not easy to sell this to clients and
40 may take a lot of persuasion and increased sales costs in the process.
41
42
High Pace of Technical Innovation
43
44 Although many microencapsulation techniques have been around for a number of years
45 now, it is still a very active research area, in which there is a constant stream of new,
46S improved techniques and consequently patent applications. In order for companies to offer
47N the latest, and best, solutions to industry problems using microencapsulation, and to be able
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 215

Marketing Perspective of Encapsulation Technologies 215

to react to new market opportunities, they need to keep track of new technical innovations 1
and to develop their own techniques as quickly as possible. 2
3
4
Client Communication
5
Whether the client be another company or simply another department in the same company, 6
effective communication during a project to make use of microencapsulation technologies 7
is vital to the project’s success. This can often be no easy matter due to the complex scien- 8
tific nature of many such projects. In addition, a high level of trust needs to be built between 9
the parties concerned, with what may be quite sensitive projects due to the high degree of 10
innovation and possible high returns. 11
Realistic project goals need to be set, including highlighting technical limitations. 12
A thorough initial screening of the technical possibilities is also vital. Care needs to be 13
paid to communication between technical and marketing departments, so that the resulting 14
products are deemed marketable as well as technically innovative. 15
16
17
Scale-up of Processes to Manufacture High-Volume Products
18
The scale-up of any chemical process to an industrial level always presents new challenges. 19
The physical properties of bulk reactions differ markedly from those present in a laboratory 20
scale reaction, adding a level of complexity, which needs to be taken into account during 21
the design of the process. These scale-up challenges can be significant even for the simplest 22
chemical processes; for microencapsulation, it is even more challenging due to the fact that 23
most laboratory scale reactions are not completely understood. The commonly used state- 24
ment that microencapsulation is as much an art as a science bears witness to the problems 25
that are faced in scale-up, the major problem being the reproducibility of reactions to pro- 26
duce a consistently good-quality encapsulation of the substance. 27
28
29
Technique Differentiation
30
Although most microencapsulation techniques can be related to a relatively small number 31
of basic principles, variations on these have resulted in a quite bewildering array of differ- 32
ent techniques being available on the market. Whichever industry a company is targeting, 33
a company with a particular proprietary technique faces the hard challenge of selling the 34
advantages of its own technique over others on the market. The best way to do this is to 35
identify the key defining characteristics of the technology, breaking this down to a few eas- 36
ily recognizable advantages, and putting these to potential clients in a strong and clear mes- 37
sage. Of course, different strategies will need to be employed for the different industries to 38
which the technique might be applicable. Certain factors may be particularly of interest in 39
one market, and these should be identified and highlighted. The further sections of this 40
chapter should provide a clear indication of what these might be in the markets covered. 41
42
43
Identification of High-Volume End Uses
44
Many microencapsulation applications are conducted on relatively small scales of perhaps 45
a few tonnes or less. Whilst these may be of a high value, the high cost of development S46
ultimately reduces the profits available from such applications. It has been mentioned time N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 216

216 Chapter 10

1 and again to Frost and Sullivan during the course of the research that many companies are
2 looking for the next high-volume industrial use of microencapsulation technology, which
3 might rival the volumes used in the carbonless paper industry. The most likely candidates
4 for this would appear to be microencapsulated phase change materials or possibly elec-
5 tronic ink applications if both of these markets expand to their full potential.
6
7
Identification of Technology Potential by Market Participants
8
9 Many companies have developed microencapsulation techniques or brought in technology
10 for their own in-house applications. Often, this has been as far as those companies have
11 taken the process. Without continued R&D in the area, they may find that the technique
12 they are applying is no longer the most suitable or has become uncompetitive compared to
13 microencapsulated products produced by other companies.
14 As microencapsulation in many ways is still very high tech, quite expensive, and often
15 requires a lot of experience to perform, the ability to achieve is still of high value and may
16 be more marketable than some companies realize. It may be that they do not want to go to
17 the trouble or expense of outsourcing the technology. Such companies may also represent a
18 business opportunity for specialist microencapsulators who might be able to demonstrate
19 the cost benefits of a more modern technology, which could be licensed out.
20
21
Overview—Microencapsulated Food Ingredients
22
23 Microencapsulation of food ingredients is not a new concept. Earlier efforts to encapsulate
24 flavors were based on spray drying using acacia gum as the coating materials. However, the
25 ever-increasing complexity of food products is continuing to drive research into novel and
26 different encapsulation techniques and processes. In particular, the rapid growth in functional
27 food seen toward the end of the 1990s is continuing apace, and it is the unstable or unpalatable
28 nature of many of the active ingredients used in these products, which will continue to open
29 up new opportunities for the use of microencapsulation technologies in the food industry.
30 Microencapsulation of food ingredients is performed for a wide number of reasons,
31 including improved substance stability, taste masking, ease of handling, and controlled
32 release. Today, a wide range of food ingredients are microencapsulated using one technique
33 or another. Table 10.1 lists examples of microencapsulated taste-masking solutions cur-
34 rently on the market.
35 This chapter by no means provides an exhaustive reference to all the areas in the food
36 industry to which microencapsulation is being applied, but will attempt to provide a flavor
37 of a few of the most interesting areas, where high-value microencapsulation technologies
38 are being applied to bring improvements to the food products we eat.
39
40
Reasons for Encapsulation
41
42 As has been discussed by other authors in this book, the main reasons for the microencap-
43 sulation of food ingredients can be outlined as follows:
44
45 1. Protection—This is surely the primary reason for the microencapsulation of food and
46S feed ingredients. Protection can be achieved from a wide variety of influences that
47N might cause an ingredient to lose its functionality; this might be simply physical
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 217

Marketing Perspective of Encapsulation Technologies 217

Table 10.1. Microencapsulated food ingredient market: Examples of companies offering 1


microencapsulation taste-masking solutions on the market (source: Frost and Sullivan) 2
Company Product/brand Specific application 3
4
Bio Dar Encapsulates Protected by masking bitterness Chewing gum applications
5
of herbal flavors
Coating Place Barrier coatings Tailor-made masking 6
Colorcon Opadry® Taste masking, pharmaceuticals 7
and foods 8
BASF Kollicoat® SR 30D Mainly pharmaceuticals with 9
food capabilities
10
Fuiz Technology Cerform and Shearform Taste masking using spun
sugars for food and 11
pharmaceutical applications 12
Particle Dynamics Micromask Pharmaceutical and foods 13
14
15
processes such as heat, light, or moisture, which might degrade the ingredient before it 16
has time to act. It may also involve retarding interactions between substances in a given 17
food formula, thus ensuring product’s acceptable shelf-life. 18
2. Controlled release—Microencapsulation can be used in a delivery capacity whereby an 19
ingredient is released at the required time and place. This could mean the release of a 20
leavening agent at a certain temperature during baking, a flavor upon chewing, or a pro- 21
biotic culture upon digestion in the small intestine. 22
3. Processability—Microencapsulation techniques can simply be used to allow easier han- 23
dling of an ingredient during production of the product. The advantages of providing 24
a dry powdered form of a flavor to a bakery that otherwise only uses powdered ingredi- 25
ents are obvious. 26
4. Taste masking—Vitamins and minerals are increasingly being added to food to increase 27
actual or the consumer’s perceived health benefits of the product by the consumer. Many 28
of these ingredients are unpalatable, and their taste needs to be masked until they have 29
passed the taste buds and the mouth area. This reason for encapsulation is particularly 30
prevalent in the animal feed industry, where larger concentrations of such ingredients 31
are added in order to produce healthier and hence better yielding animals. 32
33
34
Modes of Release
35
If you have encapsulated an ingredient, you will want it to be released from that encapsu- 36
lant in order for it to function within the food product at the desired time and place. The 37
most common modes of release are detailed below: 38
39
1. Thermal release—Whereby the encapsulant melts at a certain temperature, usually dur- 40
ing cooking of the product, releasing the ingredient. By altering the type of coating and 41
its thickness, it is possible to ensure release of an ingredient within a few degrees of the 42
required temperature. 43
2. Physical release—Requiring the physical breaking of the microcapsules; usually this 44
mode of release is designed for ingredients that need to be released during chewing. 45
Factors which can be altered to prolong or otherwise the release profile include the size S46
of the capsules and the strength and flexibility of the coating. N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 218

218 Chapter 10

1 3. Dissolution—Most food products contain at least a small amount of water, which can be
2 used to ensure the release of an ingredient enclosed in a water-soluble coating. The
3 chemistry of the coating can be designed so that this only occurs at a particular pH, tem-
4 perature, or salt concentration.
5
6
Market Drivers
7
8 Examples of key market drivers for microencapsulated food ingredients include:
9
10
Increased Consumption of Processed Foods which Require
11
Ingredient Stabilization
12
13 Sales of processed and pre-prepared foods, commonly known as de-cooking phenomenon,
14 continue to increase. This essentially means that people are devoting less and less time to
15 the preparation of their food, and so are using less fresh ingredients and more that are pre-
16 pared in some way to speed cooking. The increased processing that these products may
17 undergo, including pre-cooking and freeze–thaw cycles, can harm many ingredients in the
18 food products, resulting in lower quality for the consumer. Microencapsulation can protect
19 many of these ingredients during processing and then be designed so as to release them
20 when needed, either during the final cooking process or upon consumption.
21
22
Rapid Growth in the Functional Food Market
23
24 Sales of functional foods have increased dramatically since the mid 1990s. This category is
25 best represented by probiotic milk drinks or vitamin fortified sports bars and cereals. Obvi-
26 ously, such products need to contain some ingredients which will impart this benefit, and
27 many such ingredients are unstable or difficult to handle such as vitamins, minerals, amino
28 acids and herbal extracts.
29
30
Drive for Brand Differentiation Increases Call
31
for Microencapsulated Ingredients
32
33 Food manufacturers are responding to more complex consumer requirements and increas-
34 ing competition, particularly from the powerful supermarket’s own innovative brand prod-
35 ucts and product lines. The use of ingredients or processes which impart some novel
36 characteristic to a food product, such as a new taste or health benefit, is a key tool in selling
37 more expensive branded products to consumers. Many such innovations are always copied
38 in some way by the supermarkets, creating a cycle of innovation, which can only increase
39 the use of such high-tech processes within the food industry.
40
41
Consumer Demand for Natural Food Products
42
43 The continuous string of public health scares surrounding the food industry, as well as
44 greater awareness among consumers of the benefits of a healthy diet, are driving the
45 demand for food products which are natural in origin. Many of these natural ingredients are
46S however unstable to harsh environment of food processing and cooking, so in order to
47N
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 219

Marketing Perspective of Encapsulation Technologies 219

ensure the survival of such ingredients through consumption, microencapsulation is being 1


used to protect them. 2
3
4
Microencapsulation Reduces Amount of Ingredient Required
5
for Same Effect
6
Whilst many microencapsulation techniques are often considered too expensive for appli- 7
cation to food ingredients, on some occasions, the use of such technology can actually 8
result in a cost saving. The protection of food ingredients until they are required can result 9
in the need for much less of that ingredient in the product, possibly producing significant 10
savings on raw materials. 11
12
13
Complexity of Technical Requirements Creates Opportunities
14
for Specialist Companies
15
The complexities of encapsulation processes often mean that an entirely new procedure 16
needs to be developed for a given application or a new ingredient. Different problems may 17
also call for the use of totally different technologies. Food ingredient manufacturers cannot 18
possibly have all the equipment and technical know-how in-house in order to cater for each 19
possible new problem or innovation. This creates an increasing demand for contract 20
microencapsulation services. Small contract companies have the flexibility to respond to 21
customer needs. They will often produce very small volumes, which would not be econom- 22
ically feasible for a larger company to do. 23
24
25
Tightening of Laws on Product Claims may Force Companies
26
to Use Microencapsulation
27
The increasing focus on quality of food products, driven by consumer demand for more 28
information about the food they consume, will undoubtedly lead to tighter regulations gov- 29
erning the product claims that food manufacturers make. At present, in many countries, it is 30
simply necessary for companies to introduce an ingredient at the outset of the production 31
process, without ensuring that the stated level is actually present in the finished product. 32
Often, sensitive ingredients such as some vitamins are destroyed by the cooking process, 33
making claims for the extra health benefits of such added substances essentially false. If 34
producers have to ensure that the ingredients survive until consumption, the protection of 35
these ingredients would become vital, leading to new opportunities for developing and 36
using novel microencapsulation technologies. 37
38
39
New Product Development Creates Future Markets
40
The development of an encapsulation process for a new ingredient or new purpose is often 41
an expensive and time-consuming procedure, which will not be undertaken unless a decent 42
return on that investment is expected. One of the best ways for companies providing 43
microencapsulated food ingredients to identify where to focus their research is by under- 44
taking contract work. Each new request, whether it results in an order or not, brings a new 45
idea or indication of a market need, which can hopefully be exploited. S46
N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 220

220 Chapter 10

1 Market Restraints
2
3 Examples of the key market restraints for microencapsulated food ingredients include:
4
5 Cost of Many Microencapsulation Techniques Reduces Scope for
6 Use in Food Products
7
8 The cost of most microencapsulation techniques is a serious constraint to the use of this
9 technology in the food industry, where traditionally the end-user markets have been mature
10 and price sensitive. An alternative scenario is its use in those situations where encapsula-
11 tion is the most cost-effective solution to a particular problem.
12
13 Lack of Industry Awareness of Microencapsulation
14
15 It is still felt that many food processors are not aware of the possible advantages which
16 microencapsulated ingredients can bring to their products. Combined with this are miscon-
17 ceptions about such things as the cost of these ingredients and concerns over the increased
18 level of processing which they might represent. The desired widespread knowledge of these
19 techniques within the food industry will undoubtedly take a long time to build up and will
20 limit the wider use of the technology in the process.
21
22 Difficulties in Technical Communication with Customers
23
24 Related to the above constraint is a common complaint from many encapsulation special-
25 ists that companies approaching them do not understand the nature of microencapsulation.
26 Whilst they generally welcome any approaches for business, this is obviously a source of
27 frustration within the industry, and the wasted time and effort can have financial implica-
28 tions. In addition, if clients go away with a poor impression of microencapsulation due to
29 unrealistic expectations, this will have an impact on the future market potential for such
30 technology. It is the encapsulation companies themselves, as providers of the service, who
31 need to communicate effectively with their potential clients, so that realistic project objec-
32 tives are agreed upon, which will result in satisfaction for both sides.
33
34 Limitations on Encapsulant Materials
35
36 There are a number of limitations on the materials which can be used to encapsulate actives
37 which may ultimately lead to technical and supply problems:
38
39
BSE and Foot and Mouth
40
41 The Bovine spongiform encephalopathy (BSE) epizoolic has created a consumer confi-
42 dence crisis in the safety of materials derived from animal sources. Products using encap-
43 sulation are often functional foods bought by very health conscious consumers, who are
44 aware of such issues. Thus, the presence of gelatin may cause concern among consumers.
45 The recent foot and mouth disease epizoolic in the UK and the resulting ban on the export
46S of animal products also caused an increase in gelatin prices by as much as 20 percent.
47N
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 221

Marketing Perspective of Encapsulation Technologies 221

GMO 1
2
Consumer demand has led to most food manufacturers in Europe, in particular, being very
3
careful not to use ingredients that might come from genetically modified organisms in their
4
products. This has affected supplies of certain materials such as maize-derived starches
5
from the US.
6
7
Approval of Ingredients 8
9
The European Union maintains a list of chemicals that have been approved for use in food. 10
This may limit the development of novel processess that may require materials not on the 11
approved list. 12
13
Competitive Factors 14
15
One key competitive factor in this market is technology. The ability to consistently deliver 16
added value to food products through the use of a microencapsulation is vital for companies 17
using this technology. It is not an easy process to sell simple products such as these at much 18
higher prices than normal, so customers must be convinced that the microencapsulation is 19
achieving the desired properties in their products. The ability to deliver something different 20
and superior in terms of performance is the key to attracting more customers in this market. 21
Another important competitive factor, as in the provision of microencapsulated products 22
in many other industries, is customer service. With most ingredients being designed for 23
specific applications, often at the request of customers, close attention to delivering on cus- 24
tomer service and maintaining good relationships is important to maintain market share. 25
Another key competitive factor is technical expertise, as the microencapsulation has to 26
achieve the desired result in the final product in order for customers to continue to pay 27
higher prices for these products. Properties of the encapsulation are often tuned to the cus- 28
tomer’s requirements for a particular application, so close attention to customer service is 29
very important to ensure the product is successful and repeat business is gained. 30
Microencapsulation of flavorings undoubtedly adds some cost to the production, which 31
is generally reflected in higher prices. Respondents were very unwilling to discuss what 32
actual price the microencapsulation added to a flavoring, mainly due to the fact that this can 33
vary greatly according to the technique applied, the volume of product made, and the dose 34
rate of that flavoring. In addition, the added value of the microencapsulation was often felt 35
to result in cost savings for customers due to lower wastage of the volatile flavorings and 36
better impact in the product, resulting in the need to use lower volumes. 37
Microencapsulation techniques can result in price increases of 2 to 10 times the price of 38
a non-encapsulated flavoring. The prices for microencapsulated flavorings are not expected 39
to mirror this decline as the added value they bring often means that companies are not 40
competing on price against similar products. 41
42
43
Pricing
44
The food industry tends to be price sensitive, so the extra cost associated with microencap- 45
sulating an ingredient needs to be fully justified in terms of offering a clear improvement in S46
N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 222

222 Chapter 10

1 performance of that ingredient within the food product. The application of microencapsula-
2 tion is more easily justified with higher value ingredients such as flavorings, vitamins and
3 minerals, and probiotics.
4 Price is an important competitive factor, for both non-encapsulated and microencapsu-
5 lated ingredients. Higher prices of microencapsulated ingredients must be shown to be jus-
6 tified by their better performance. There can also be significant price differences between
7 microencapsulated products from different companies, and here also, improved perfor-
8 mance must be demonstrated.
9 Microencapsulation is performed by both the original ingredient manufacturers and
10 companies specializing in microencapsulation technologies. For the latter, it is imperative
11 that their technical expertise allows them to offer something unique in terms of perfor-
12 mance. Their hardest task lies in convincing clients of this performance benefit in relation
13 to the higher price of the ingredient. If their message is strong enough, they will face fewer
14 market barriers and lower competition. Companies need to work closely with clients to
15 identify the end-use products that microencapsulated ingredients could add benefit to and
16 design suitable products to meet this need.
17
18
Industry Structure
19
20 Microencapsulation is performed by both the original ingredient manufacturers and com-
21 panies specializing in microencapsulation technologies. Examples of companies amongst
22 the latter group include Balchem, Particle Dynamics, Bio Dar, and TasteTech. Most of the
23 major flavor houses are actively involved in research into the use of microencapsulation
24 technologies, as are other major food ingredient manufacturers. At a very simplistic level,
25 companies can be divided into two groups according to their capabilities of adding value to
26 their businesses. In practice, there is a significant amount of cross-over between these two
27 definitions:
28
29 1. The first type includes companies that are usually small- or medium-sized enterprises for
30 which the ability to do microencapsulation lies at the core of their business and provides
31 their main revenue stream. Such companies market their ability to perform microencap-
32 sulation in a number of ways. Close cooperation with their customers is vital and can
33 involve licensing a production process, toll manufacturing, co-development work, or sale
34 of a bespoke product line. Such companies may also sell standard microencapsulated
35 products, or products in which microencapsulation is the core enabling technology.
36 2. The second type of companies are usually larger enterprises for which microencapsula-
37 tion is simply another manufacturing technique they have at their disposal and that they
38 can use in one or more product lines. Such companies have usually developed or pur-
39 chased microencapsulation capabilities for a particular in-house application, but have
40 gone on to apply this knowledge in other areas of their business. They do not generally
41 offer their microencapsulation capabilities to other companies. An exception to this trend
42 is companies such as BASF and 3M. These enterprises have been offering their microen-
43 capsulation capabilities to other companies in certain markets. Such companies and few
44 others can really be considered providers of a wide range of microencapsulation expertise,
45 which can be applied for virtually any company in any industry. Such companies specifi-
46S cally advertise their ability to apply their techniques to long lists of substances. Examples
47N of these companies are South West Research Institute, Thies Technology, 3M and Ciba.
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 223

Marketing Perspective of Encapsulation Technologies 223

Microencapsulators in Specific Industries 1


2
Examples include Coletica and Lipotec in the cosmetic industry. In reality such companies, 3
though specialized in certain applications, are often looking for new business opportunities 4
for applying their techniques and would gladly try to adapt them to other industries. Cross- 5
over capabilities are often seen in the cosmetic and food industries. 6
7
Companies for which Microencapsulation Performs a Vital Role in their 8
Main Product Line 9
10
For carbonless paper manufacturers, microencapsulation is thus an inherent part of their 11
production lines. Although most companies originally licensed the technology from its 12
developers, through using it for a number of years, they were able to have build up a high 13
degree of expertise in the area. Similar examples can be found in the textile industry. 14
15
Companies which Apply Microencapsulation to Certain 16
Product Lines 17
18
Examples of this category are flavor suppliers that offer some of their flavors for certain end 19
uses in microencapsulated form. Such companies usually bring the technology from an out- 20
side source, but through running the processes for a number of years, they develop their 21
own expertise and even research capabilities. Table 10.2 gives examples of companies that 22
are involved in the provision of microencapsulation technologies to the food industry. 23
24
Balchem Encapsulates 25
26
Balchem Encapsulates is a wholly owned subsidiary of Balchem Corporation, which was 27
founded in 1967 by the merger of several entities, including Dr Leslie Balassa who owned 28
several technology inventions in the field of encapsulation. It operates in two business seg- 29
ments, Encapsulated Products and Specialty Products. 30
31
32
Table 10.2. Examples of companies active in the food ingredients 33
microencapsulation market (source: various) 34
Company Website 35
36
Aveka www.aveka.com
Balchem www.balchem.com 37
Brace www.brace.com 38
Coating Place www.encaps.com 39
Micap www.micap.com 40
Particle Coating Technologies www.pctusa.com
41
Particle Dynamics www.particledynamics.com
Ronald T. Dodge Co. www.rtdodge.com 42
Sono-Tek www.sono-tek.com 43
Southwest Research Institute www.swri.com 44
TasteTech, UK www.tastetech.co.uk 45
Thies Technology www.thiestechnology.com
S46
3M www.mmm.com
N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 224

224 Chapter 10

1 Balchem’s Encapsulated Products business utilizes proprietary microencapsulation


2 technologies in an ever-expanding variety of applications, with food and feed ingredients at
3 its core business. Its major product line ranges from ingredients for bakery products
4 (Bakeshure), confectionary (Confecshure) and wellness products (Vitashure), as well as
5 encapsulated flavors (Flavourshure) and meat-processing ingredients (Meatshure). Its ani-
6 mal feed additives have the trade name Reashure. Balchem also offers partnerships and toll
7 manufacturing capabilities to solve particular problems faced by companies, which might
8 be solved using its technologies.
9 Balchems’ acquisition of the encapsulation and agglomeration capabilities of Loders
10 Croklaan in July 2005 underlines the ambition of this company to grow its core business.
11
12
Brace
13
14 The Germany-based company, Brace, specializes in laminar flow break microcapsule
15 forming. The firm holds a wide variety of patents worldwide. Examples of their products
16 include microcapsules of gelatin, alginate, and agar, filled with flavors for use in breath
17 freshening, chewing gum, and ready meal applications.
18
19
Karmat Coating Industries Ltd
20
21 Karmat is an Israeli company formed in 1993 and jointly owned by Kibbutz Ramot
22 Menashe and Coating Place Inc. Karmat’s microencapsulation techniques are mainly based
23 on fluid-bed technology, which employs an open air system allowing the use of many dif-
24 ferent coating materials including modified starches, peptides, cellulose derivatives, fatty
25 acids, and peptides. It applies these to the encapsulation of ingredients for the food, cos-
26 metic, pharmaceutical, chemical, and feed industries.
27 Karmat’s main business is in the microencapsulation of vitamins and minerals, which it
28 incorporates into bespoke premixes for its clients in the food industry. Most products it pro-
29 duces are customized specifically for clients. Its premixes for the dairy sector are called Lac-
30 tomix, those for baking applications, Bakeamix, and those for use in baby food, Babymix.
31 Other product lines include microencapsulated citric acid, ascorbic acid, and ferrous sulfate.
32
33
Bio Dar
34
35 Bio Dar was founded in 1984 as an Israeli–American joint venture. In 1998, the company
36 was acquired by Lycored, a subsidiary of Makhteshim-Agan and part of the large Israeli
37 holding group, Koor Industries. Bio Dar’s main products are microencapsulated vitamins
38 and minerals. It also manufactures microencapsulated nutraceuticals such as carnitine,
39 amino acids, and herbal extracts. Over 50 percent of its products are customized to specific
40 customer requirements.
41
42
Particle Dynamics
43
44 Particle Dynamics, Inc. (PDI) is an American-based company acquired in 1972 by KV Phar-
45 maceutical company. PDI markets specialty raw materials for the pharmaceutical, nutri-
46S tional, food, and personal care industries. It divides its business into three main technology
47N lines; Destab, a direct compression technology used to make tablets for pharmaceutical and
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 225

Marketing Perspective of Encapsulation Technologies 225

nutritional supplement purposes, Descote®, a line of microencapsulated vitamins, minerals, 1


and herbal extracts, and MicroMask which encompasses a series of technologies for taste 2
masking mainly in over-the-counter medicines. 3
PDI’s proprietary microencapsulation technology is based on the trapping of ingredients 4
in a matrix, in which particles of the ingredient are effectively embedded. The advantage of 5
this technology lies mainly in its physical stability, being able to better withstand processes 6
such as tabletting. The company follows a lower risk business approach of modifying exist- 7
ing compounds and formulations rather than discovering new molecular entities. Particle 8
Dynamics serves the vitamin, food, and herbal supplement industries and is also involved 9
in pan coating. 10
11
12
Particle Coating Technologies
13
Particle Coating Technologies, Inc. (PCT) is a research and development company dedi- 14
cated to microencapsulation technologies. Formerly part of the University of Washington, 15
Department of Chemical Engineering in St. Louis, MO, the company became independent 16
in 1994. 17
PCT pioneered a spinning-disk coating technique and also specializes in the formation 18
of narrow particle-size distribution products, using a proprietary atomizer. PCT claims 19
more than 20 of the world’s largest 100 public companies as customers and has some toll 20
manufacturing capabilities in addition to its core work in feasibility studies. 21
22
23
TasteTech
24
TasteTech is a privately owned British company founded in 1992 and currently distributes 25
its ingredients worldwide. It utilizes its proprietary microencapsulation technology to 26
encapsulate flavors, spice extracts, and key ingredients used in the food industry. It also 27
applies its technology in the pharmaceutical and cosmetic industries. TasteTech employs 28
three main microencapsulation technologies as well as spray drying capabilities. Its con- 29
trolled release techniques are labeled CR100, CR200, and CR300 and are used to encapsu- 30
late the ingredients mainly using vegetable fats and oils. The advantage of using these 31
encapsulant materials is the ability to control the release of the ingredients by adjusting the 32
melting point of the fat coating. 33
34
35
Key End-User Groups
36
In terms of the microencapsulation technology itself, almost any company in the chemical, 37
food, and related industries is a potential end user. The number of areas the techniques are 38
being applied to continues to grow as more research is carried out and more projects 39
attempting to harness microencapsulation capabilities are undertaken. 40
41
42
Competitive Factors
43
Table 10.3 gives examples of some of the factors taken into consideration in the selection of 44
a microencapsulation technology. 45
For those companies marketing their ability to perform microencapsulation for other S46
companies in any market, an important competitive factor is reputation. If a company is N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 226

226 Chapter 10

1 Table 10.3. Microencapsulated food ingredient market: Important factors to consider


2 (source: Frost and Sullivan)
3 Issue Questions Importance
4
Function Primarily taste-masking is the Vital function
5
raison d’etre of microencapsulation
6 Cost What is my budget? Is the technology Need to weigh effectiveness,
7 cost-effective? new methods are expensive!
8 Size How small do my encapsulated Important for mouthfeel, taste,
9 ingredients need to be? and effectiveness
Production/ Will the encapsulated ingredient Microencapsulation is ineffective
10
conditions survive processing and storage? unless it can protect the ingredient
11 from processing and storage
12 Effects How will the encapsulated ingredient Unintentional interactions
13 interact with other ingredients can ruin a product
14 in the finished product?
15
16
17 recognized as an expert in the field and has previously participated in a large number of
18 successful projects, then approaches from clients are far more likely. Given the unpre-
19 dictable, project-based nature of much of this business, such industry recognition is vital if
20 a company is to generate a steady stream of work in the area. Other activities which can
21 raise a company’s profile in this way might be to hold as many patents as possible in the
22 area concerned or to regularly publish scientific papers on the subject and give talks at
23 conferences.
24 For any company using microencapsulation, innovative ways of applying the technology
25 are likely ultimately to lead to greater profitability. If the technology can be applied to give
26 a product a competitive edge or to introduce an entirely new concept to the market, high
27 rates of growth in that product may well result.
28
29
Examples of Microencapsulation in the Food Ingredient Industry
30
31 This section reviews the use of microencapsulation technology in the flavors, vitamins,
32 salts and acids, and probiotics markets. In these markets, the use of microencapsulation has
33 moved beyond basic coating for handling purposes toward its use for stability purposes and
34 more importantly, controlled release of the encapsulated substance.
35 The largest market using microencapsulation technologies is that for flavorings. Accord-
36 ing to Frost and Sullivan (Oxford, UK), microencapsulation of flavors in the European mar-
37 ket was valued at $340 million in 2001.
38 Other market sizes and growth rates vary widely, depending on the food ingredients
39 concerned, but overall growth in the use of microencapsulation is undoubtedly being expe-
40 rienced as more food and beverage manufacturers begin to recognize the benefits that some
41 of these technologies can bring. The two major growth areas will continue to be in the func-
42 tional food sector, where microencapsulation can be used to stabilize ingredients such as
43 vitamins and minerals, and elsewhere in techniques which offer controlled release of ingre-
44 dients. Both these markets are expected to see double-digit growth in the next few years.
45 The increased processing of foods, coupled with the desire for increased quality and
46S innovation, is driving growth in the use of techniques which can effectively stabilize sensi-
47N tive ingredients.
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 227

Marketing Perspective of Encapsulation Technologies 227

Microencapsulated Flavors Market 1


2
Flavor is an essential part of our experience when eating food. Four basic flavor types have 3
been identified: sweet, sour, bitter, and salty, with a fifth defined by the Japanese as Umami, 4
a savory flavor. Obviously, many natural foods have their own inherent flavors, but today’s 5
complex food products often require the addition of one or more flavors in order to satisfy 6
the consumer. The major areas of flavoring use are in dairy foods, confectionary, beverages, 7
bakery foods, and other savory foods. Flavors can be derived by physical extraction from 8
natural sources or synthesized. 9
10
Why Microencapsulate Flavors? 11
12
Microencapsulation can help protect flavors from the rigors of harsh production processes 13
and in providing a much high sensory impact in the final product. In addition, a more natu- 14
ral flavor can be provided by protecting sensitive extracts and further releasing them at the 15
time of consumption. 16
Finally, flavor manufacturers are simply looking to offer innovative new products 17
to their customers. The protection provided by microencapsulation can be harnessed to 18
allow the use of certain flavors or flavor combinations which were not previously possible 19
in certain products. As discussed earlier, microencapsulating flavors can help in various 20
ways: 21
22
1. Protection from degradation. 23
2. Controlled release at a desired time and rate. 24
3. Stabilization and shelf-life extension. 25
26
End-Use Applications 27
28
The segmentation of market revenues for microencapsulated flavors does not necessarily 29
follow that of the total flavor market due to certain end-use requirements. The major end- 30
use markets for microencapsulated flavorings are listed below: 31
32
1. Confectionery products—sustained release of flavorings is often required to add value 33
to the products. The most common type of confectionary that use microencapsulated 34
flavorings is chewing gums. Confectionery is estimated to account for 35 percent of 35
European microencapsulated flavorings market revenues. 36
2. Bakery products—dry powdered flavorings are required for easier mixing with other 37
powdered ingredients. Controlled release of flavors in baking is also a growing feature 38
of baked goods. 39
3. Powdered beverages—encapsulated dry powders are necessary for better mixing with 40
other dry ingredients of the beverage components and for enhanced shelf life of the dry 41
mix. Release is achieved through dissolution of the coating. Spray dried flavors are 42
widely used in this market for ease of handling and instant solubility. 43
4. Processed foods—all the reasons for microencapsulating flavors mentioned above are 44
applicable to its application in processed foods. More advanced microencapsulation 45
techniques are being used in this market sector to address a range of shelf-life and con- S46
trolled release issues. N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 228

228 Chapter 10

1 Microencapsulation is a very active area of research at most of the major flavor houses.
2 These companies have large R&D budgets needed to invest in the development of microen-
3 capsulation expertise, which can take a lot of practice to perfect. Such a quality-driven mar-
4 ket sector also presents opportunities for smaller technology providers. Although profit
5 margins from partnering may be lower than selling your own products, access to the large
6 marketing and sales forces of the industry leaders will maximize the sales potential of the
7 product, which is something that smaller companies can struggle to achieve on limited
8 budgets.
9
10
Examples of Market Drivers and Restraints in the Flavors Industry
11
12 Product innovation gives market advantage. Innovative new food products which capture
13 the imagination of the public give food manufacturers a competitive edge in the competi-
14 tive food market. Such innovation within the food industry is a major driver of the use of
15 microencapsulated flavors, which can add an extra dimension to a food product. Possibili-
16 ties such as sequential release of flavors, long-lasting flavor effect, or simply the ability to
17 introduce certain flavors into a new food area can all help to increase the appeal of a food
18 product and help it to capture market share.
19 Increasing use of processed foods. Sales of processed and pre-prepared foods con-
20 tinue to increase as the phenomenon known as de-cooking increases. Such systems often
21 require flavors to be protected from the processing they undergo in order that they can be
22 delivered to the consumer when the product is eaten.
23 Consumers demand better food quality. In conjunction with the greater consumption
24 of processed foods, consumers are demanding greater quality from these food products.
25 One particular quality demand is that for more authentic or natural tastes. Microencapsula-
26 tion can be used to deliver individual, authentic tasting flavors at exactly the right time,
27 resulting in greater customer satisfaction with the product.
28
29
Examples of market constraints in the flavors industry
30
31 Drive for lower process costs. Although microencapsulated flavorings cost more than
32 those which are not encapsulated, they can offer cost savings in other ways. Microencapsu-
33 lation of flavors into dry powders can help in this regard. Less expensive equipment might
34 be needed and production steps removed due to the easy mixing of different powders rather
35 than oil inclusion. In addition, less flavor is likely to be released to the atmosphere during
36 processing, resulting in the need to use less of the product to achieve the same effect.
37 Environmental problems with spray drying lead producers to search for alternative
38 technologies. The use of spray drying, where a fine mist of flavor is heated will inevitably
39 lead to the escape of some of these volatile compounds into the atmosphere, which can
40 require the use of expensive scrubbers to control. Even then, certain flavors such as onion
41 or cheese can be particularly troublesome. Companies using spray drying for flavors are
42 monitored by environmental officials at a national level, who have the ability to impose
43 injunctions stopping the use of the equipment for particular flavorings. This has led produc-
44 ers to look to other microencapsulation techniques for flavor applications. It has been sug-
45 gested to Frost and Sullivan that if another suitable technique were available to encapsulate
46S flavorings in water dispersible coatings, it would be take a large share of the market from
47N spray dryers.
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 229

Marketing Perspective of Encapsulation Technologies 229

Consumer and legislative restraints on encapsulant materials. The European approved 1


ingredient list contains details of those substances which are allowed for use as food ingredi- 2
ents and broadly in which applications they can be used in. Such regulatory constraints pres- 3
ent major challenges mainly in limiting the number of materials available, their usage in 4
specific systems, and sometimes at low concentrations that may render them ineffective. 5
Consumer preference can also limit the types of encapsulating material used. A particular 6
example is the movement against genetically modified materials. 7
8
9
Microencapsulated Vitamins Market
10
Vitamins are essential parts of the human diet, without which we are prone to a large num- 11
ber of vitamin deficiency-related diseases and general ill health. Traditionally, we have 12
derived these essential nutrients by consuming a varied diet of natural foods abundant in 13
particular vitamins, such as many fruits and vegetables. The techniques most widely prac- 14
ticed are physical processes such as spray drying, spray cooling, and fluid-bed techniques. 15
Revenues indicated in this document refer to all coated vitamins. 16
17
18
Why Microencapsulate Vitamins?
19
The major reasons for microencapsulating vitamins for use in food products are detailed 20
below. In addition to processability and stability issues discussed above for encapsulating 21
flavors, most vitamins have objectionable taste that needs to be masked so that the active 22
can only be released in the gut. The largest volume of vitamins sold in microencapsulated 23
form include the fat- and water-soluble types such as A, D and E, K1, and the B-group 24
(notorious for medicinal taste). 25
26
27
Vitamins in Animal Feed
28
Vitamins are also microencapsulated for use in animal feed products for both taste-masking 29
and bioavailability reasons. The latter is particularly an issue for ruminants, where rumen 30
by-pass products can utilize microencapsulation to ensure that vitamins reach the second gut 31
and intestines in their intact form. 32
33
34
Microencapsulated Salts and Acids Market
35
A variety of other food ingredients have been microencapsulated using mainly fluid-bed 36
techniques; the most common are salts (sodium chloride, sodium bicarbonate, and sodium 37
diacetate) and acids (citric, malic, sorbic, tartaric, ascorbic, acetic, fumaric, etc.). 38
These ingredients are encapsulated for a number of reasons such as controlled release, 39
extended shelf-life, prevention of color degradation, and protection against moisture. Their 40
main end-use applications include confectionery, bakery, and processed meat products. 41
42
43
Microencapsulated Probiotics Market
44
Probiotics are bacteria, yeasts, or other microorganisms which provide health benefits by 45
contributing to intestinal microbial balance. These live cultures of microorganisms are S46
included in a number of food products and supplements and are generally intended to N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 230

230 Chapter 10

1 colonize the large intestine, where they impart such benefits as impeding harmful bacterial
2 growth, supporting the immune response, increasing the nutrient absorption, and reducing
3 the risk of cancer.
4 Probiotics have been included in a number of food products, which can be broadly clas-
5 sified into two groups—dairy products and other food supplements. Dairy products include
6 yoghurt, sour cream, cottage cheese, ice cream, and fermented milk drinks. Other dietary
7 supplements can be capsules, powders, or tablets.
8
9
Why Use Microencapsulated Probiotics?
10
11 One important constraint in the probiotic market is the technical challenges associated with
12 working with live cultures of delicate microorganisms. The bacteria are often sensitive to
13 the conditions found in food products or encountered during processing such as moisture,
14 temperature, oxygen, or pressure. Microencapsulation can protect the bacteria against these
15 conditions as well as control their delivery.
16 It is not expected that microencapsulated probiotics will cannibalize existing probiotic
17 markets, but they will instead open up new markets, either through the inclusion of probiotics
18 in different food products and supplement forms or through the introduction of previously
19 unavailable bacterial strains. The food supplement market will be the first to benefit, with
20 improved probiotic survival rates during tabletting a particularly attractive proposition. Two
21 main challenges can be faced in this application:
22
23 1. Higher price, which may be two or three times that for non-encapsulated bacteria. This
24 is undoubtedly the main reason why the microencapsulated products will not affect sales
25 of non-encapsulated ones, where they simply could not compete. However, for those
26 bacteria which cannot be included in food products in any other way, price is not
27 expected to be a major constraint. In fact, increasing survival rates of the bacteria would
28 reduce the need for overdosing, potentially offering savings.
29 2. Product development time: It is likely that each new bacterial strain to be microencapsu-
30 lated will need different conditions of encapsulation and will thus pose its own technical
31 challenges. This will undoubtedly slow up the time to market for a wider range of poten-
32 tial microencapsulated probiotic products. Table 10.4 gives examples of companies that
33 have developed microencapsulated probiotic products.
34
35
36
37 Table 10.4. Microencapsulated food ingredients market: Examples of microencapsulation
38 of probiotic bacteria (source: Frost and Sullivan)
39 Company/group Product/brand Specific application
40
Institut Roselle Probiocap Guarantees probiotic stability and
41
survival in cereal bars and yogurt
42 Advanced Bionutrition MicroMatrix® Allows continuous encapsulation
43 of functional foods
44 Rhodia FloraFit® Designed for supplements and
45 nutritional additives
University of Maryland Xanthan-chitosan Protects prebiotic coacervation
46S
products at 0–60°C
47N
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 231

Marketing Perspective of Encapsulation Technologies 231

Examples of Strategic Recommendations for Companies 1


Specializing in Microencapsulation Technologies 2
3
Indeed, changing consumer trends and tastes are primarily responsible for driving innova- 4
tion in the microencapsulation market. Since food manufacturers constantly monitor such 5
trends, food ingredient companies are always looking for ways to meet these ever-changing 6
demands, thereby promoting the need for novel microencapsulation technologies. 7
With consumers showing a growing preference for functional food, which now accounts 8
for a substantial chunk of the global nutrition market, food companies are looking for dif- 9
ferent ways to incorporate health-promoting ingredients that deliver some kind of health 10
benefit to the consumer. 11
12
Business Promotion Strategies 13
14
For companies wishing to promote their microencapsulation capabilities, the promotion 15
of these probably represents the major challenge to the success of the business. Potential 16
clients need be made aware of situations where microencapsulation could add value to 17
a product or solve certain commonly recurring problems. The main method currently 18
employed to get this message across is to personally talk to clients, either through a site visit 19
or at trade shows. Once projects have been undertaken, personal relationships with research 20
departments at clients can hopefully be built, which might result in further business 21
opportunities. 22
To initially catch the attention of potential clients, examples of successful projects pro- 23
ducing interesting and groundbreaking results or simply commercially successful products 24
should be emphasized. Technical expertise should be stressed through highlighting aca- 25
demic publications and patents held. Another method of business promotion is to take part 26
in interesting projects, which receive press coverage and indirectly promote both microen- 27
capsulation technology and the company involved in the project. 28
One way to spread the cost of this would be setting up an industry association represent- 29
ing all companies involved in the market and who aim at promoting the potential benefits of 30
the technology. There is presently an International Microencapsulation Society, but this is 31
aimed more at academic research than industrial issues. The European Commission has 32
also set up a thematic network entitled “Microencapsulation for Low Cost, High Volume, 33
Pharmaceutical Applications,” intended to provide a guidance manual for European Indus- 34
try indicating which technique will be most suitable for a particular application. Such a 35
project is exactly what is needed by the industry and could well prove to be the catalyst for 36
significant market growth. 37
38
Product Proposal Strategies 39
40
Maximize Focus on a Single Market 41
Most companies offering microencapsulation technologies start out by applying them to a 42
particular market area, this usually being the one for which the particular technique turns 43
out to be most suitable. Such a focus is important for these smaller entrepreneurial compa- 44
nies initially. Trying to sell the technology to a range of industries would simply dilute the 45
marketing message and prove very difficult for a small company to achieve effectively. S46
Focusing on a single market allows limited funds to be concentrated on a particular N47
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 232

232 Chapter 10

1 customer group, allowing a company to build a reputation in that industry as a reliable


2 technology provider more quickly.
3
4
5 Application of Techniques Across Different Markets
6 Whilst the above approach is important initially, the ability to apply microencapsulation
7 techniques to areas other than those where they might first have been used can be very
8 important to the long-term growth. High growth markets using the technology may turn out
9 to be part of a trend, which fade away as quickly as they arrived, so companies need to have
10 other areas to fall back on if this occurs. Particularly susceptible to this are consumer mar-
11 kets, such as the cosmetic area, where trends toward certain ingredients or product types
12 move rapidly.
13
14
15 Offer of a Diverse Range of Encapsulation Techniques
16 For companies looking to do business through their ability to do microencapsulation, it is
17 important to be able to offer a broad range of possible solutions to a particular encapsula-
18 tion problem. This will give such companies the ability to serve as many clients as possible.
19 Companies such as South West Research Institute and Thies Technology in the US have
20 expertise in many different encapsulation techniques, making the chances of a company
21 finding a satisfactory solution more likely. Even for companies focusing mainly on one
22 market, a greater number of product lines again increase the chances of a successful match
23 with a client’s product and should result in more repeat business.
24
25
26 Examples of Strategic Recommendations for Larger Companies
27 Using Microencapsulation Technologies In-House
28
Cross-Fertilization of Technology within the Organization
29
30 The number of industries, products, and substances to which microencapsulation can be
31 applied is virtually limitless. The broad applicability of encapsulation technologies should
32 be pursued by larger companies, making sure that research scientists in all business seg-
33 ments are aware of the techniques and what they might be able to achieve. Companies
34 which have clearly benefited from such an approach are 3M and BASF. 3M has long been
35 recognized as the leading large industrial user of microencapsulation technologies.
36 Although originally translated to such applications as scratch and sniff fragrance sampling,
37 these technologies are now an inherent expertise within 3M and have been applied to
38 deliver greater product performance and to develop innovative new product lines in a vari-
39 ety of industries including pheremones, oil extraction chemistry, and adhesives.
40 BASF first developed microencapsulation expertise for use in the carbonless paper mar-
41 ket, in which it still licenses the technology to a number of major paper manufacturers.
42 It has applied various microencapsulation techniques in most of the industries covered by
43 this chapter including its pesticide formulations, cosmetic ingredients, and food ingredi-
44 ents. Its integrated structure has allowed this cross-fertilization of the technology, which is
45 now being used in unexpected applications such as building insulation via phase change
46S encapsulation technology.
47N
Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 233

Marketing Perspective of Encapsulation Technologies 233

Offer of Technical Expertise to Other Organizations 1


2
As has been said time and again about microencapsulation, it is considered as much of an 3
art as a science, so there is no substitute for experience in using the techniques reliably to be 4
able to consistently provide high-quality products. Such experience, especially in the appli- 5
cation of techniques to industrial scale production, is a valuable asset and one which com- 6
panies may be able to directly derive revenues from. A microencapsulation service business 7
could be set up by combining the experience a company has in applying a number of tech- 8
niques in a range of different sectors and offering them to industry at large. 9
Despite the successful approach of 3M in lending its technologies to other industries 10
such as agrochemicals, cosmetics, food and pharmaceuticals, it did however spin-off some 11
of the business resulting partly in the formation of the company Aveka in 1994, which pro- 12
vides microencapsulation and particle processing to industrial clients. Another large com- 13
pany which has formed a microencapsulation business is Ciba. Ciba offers its expertise in a 14
range of different microencapsulation technologies to other companies and is looking to 15
apply its expertise in particular to high-volume industrial uses. 16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
S46
N47
Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 235

Encapsulation and Controlled Release: Technologies in Food Systems


Edited by Jamileh M. Lakkis
Copyright © 2007 by Blackwell Publishing

1
Index 2
3
4
5
6
7
8
9
Active transport, 179 for delivering acetylsalicylic 10
Actives in food matrices, entrapment of acid, 189 11
amorphous matrix, 3–5 for delivering antimicrobial agents, 12
complexation into cyclodextrins, 5–7 187–188 13
cross-linked or coacervated polymers, 8 for delivering caffeine, 185–187 14
emulsions and micellar systems, 7 for delivering flavors and nonmedicated 15
fat- or wax-based matrices, 7 actives, 183–185 16
hydrogel matrices, 8–9 for delivering vitamins, 187 17
microporous matrices, 7 manufacture, 183 18
Alginates, 89 vs. lozenge delivery profile, 189–190 19
polycation-coated, 90 Chitosan, 92 20
prebiotics-coated, 90–91 Coacervate phase, 150–151 21
Allyl isothiocyanate, 210–211 Coacervation, 8 22
Antimicrobial agents, 129–130 Co-crystallization, 5 23
Aspartame, 184 Coenzyme Q10, 31–34 24
Complex coacervation, 150–151 25
cross-linking of gelatin-based coacervate 26
Balchem Encapsulates, 231–232
capsule shells, 160–162 27
Beeswax, 119
encapsulation process, 157–160 28
Bifidobacteria, 92
encapsulation technology issues, 29
Bio Dar, 232
162–165 30
Bioadhesive devices, 192–194
properties, 151–157 31
Bioerodible devices, 192–194
solvent exchange, 165–167 32
Brace, 232
Confectionery products as delivery 33
systems, 181 34
Candelilla wax, 120 Controlled release systems, complex 35
Carbohydrates coacervate-based, 136–139 36
encapsulation in amorphous matrices, 3 Controlled release, 1 37
as wall-forming material, 2 Core materials, 2 38
Carnauba wax, 119 Cosolubilization effect, 18 39
Cellulose acetate phthalate, 91–92 Covalent bridging, 45 40
Chemical leaveners, 127–129 Cross-linked or coacervated polymers, 41
Chewable tablets, 196 encapsulation in, 8 42
Chewing gums, 181 Cyclodextrins, 5 43
as delivery systems for oral health, guest molecules and, 6 44
188–189 molecules, 6–7 45
composition, 182–183 type and degree of complexation, 6 S46
N47

235
Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 236

236 Index

1 Dairy products (probiotic survival) Emulsions and micellar systems,


2 cheese, 93–94 encapsulation in, 7–8
3 frozen dairy desserts, 94 Emulsions systems, 41–42
4 yogurt, 94 Encapsulants. See Core materials
5 Dietary fats delivery as o/w emulsion, 65–69 Encapsulated particles (bakery applications),
6 Dilution lines, 17 properties for
7 Dissolution, 225 adhesion and cohesiveness, 125
8 Double emulsions, 55 film thickness, 125
9 general applications of w/o/w emulsions, 58 flexibility, 124
10 taste control using w/o/w emulsions, 58–59 good barrier properties, 124
11 transport and release mechanism of mechanical strength, 125
12 water-soluble components, 57–58 melting properties, 125
13 w/o/w emulsion instability, 56–57 particle size distribution, 125
14 w/o/w emulsion production, 56 surface morphology, 125
15 Dough conditioners, 129 Encapsulated probiotics in dairy products
16 Droplet size distribution, 17, 18 (practical applications)
17 Drug delivery via oral route cheese, 106
18 advantages, 180 frozen desserts, 106–107
19 buccal delivery, 178 yogurt, 106
20 disadvantages, 180–181 Encapsulating agents. See Wall-forming
21 local oropharyngeal delivery, 178 materials
22 periodontal delivery, 178 Encapsulation, 83–84, 135
23 sublingual delivery, 178 Encapsulation, reasons for
24 Drugs, transport mechanisms of controlled release, 225
25 active transport, 179 processability, 225
26 endocytosis, 180 protection, 224–225
27 facilitated diffusion, 179 taste masking, 225
28 passive diffusion, 179 Encapsulation and controlled release in bakery
29 Duplex emulsions. See Double emulsions applications
30 antimicrobial agents, 129–130
31 Effervescent tablets, 196 chemical leaveners, 127–129
32 Electrostatic bridging, 45 dough conditioners, 129
33 Emulsion stabilization, 42–43 flavors, 130
34 coalescence, 46 sweeteners, 130
35 creaming /sedimentation, 43–44 yeasts, 125–127
36 flocculation, 44–46 Encapsulation process
37 Ostwald ripening, 47 extrusion, 3–5, 84–85
38 Emulsions spray drying, 3–4, 5, 84
39 delivery of hydrophobic food actives, Encapsulation technologies for bakery
40 59–65 applications
41 delivery of water-soluble food actives, high-pressure congealing, 117–118
42 53–59 hot melt particle-coating technology,
43 dietary fats delivery, 65–69 113–115
44 formulation design for food, 47–52 spray congealing, 116–117
45 future trends, 69–73 Encapsulation technologies in food
46S release triggers, 52–53 applications (marketing perspective)
47N stabilization, 42–47 business promotion strategies, 239
Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 237

Index 237

client communication, 223 Food packaging, microencapsulation in 1


costs associated with use of future trends, 218 2
microencapsulation, 222 microencapsulated actives for packaging 3
cross-fertilization of technology within the applications, 210–215 4
organization, 240 microencapsulated pigments, 5
identification of high-volume end uses, 215–217 6
223–224 Freeze drying, 5 7
identification of technology potential, 224 8
manufacturing high-volume products, 223 Gellan gum, 91 9
new applications identification, 222 Genetic Algorithms, 104 10
product proposal strategies, 239–240 11
technical expertise to other organizations, 241 High-pressure congealing, 117–118 12
technical innovation, 222–223 Hot melt coating (film-forming materials), 13
technique differentiation, 223 118–119 14
technique potential awareness, 222 fats and glycerides, 120–121 15
Endocytosis, 180 glycol polymers, 120 16
Enzymatic covalent cross-linking, 45–46 resins and rosins, 120 17
Enzymes, encapsulation and controlled waxes, 119–120 18
release of food, 139–140. See also Hot melt extrusion, 4 19
Phytochemicals Hydrogel matrices, encapsulation in, 8–9 20
Extrusion, 3–5 Hydrophobic food actives – delivery via 21
o/w emulsions 22
Facilitated diffusion, 179 aroma release, 60–63 23
Fat- or wax-based matrices, encapsulation in, 7 lipophilic health ingredients, 59–60 24
Fats and glycerides structured emulsions in hydrogels for aroma 25
lauric acid group, 120–121 release, 63–65 26
oleic/linoleic acid group, 121 Hydrophobicity, 122 27
palmitic acid group, 121 28
Flavor encapsulation, 149–150. See also Incomplete surface coverage bridging, 45 29
Complex coacervation 30
Flavor-loaded microcapsules, 149–150 Karmat Coating Industries Ltd., 232 31
Flavors, 130 κ-Carrageenan and locust bean gum, 91 32
Flocculation, 44 33
bridging, 45–46 Lozenges, 190 34
depletion, 44 as delivery systems for dry mouth relief, 35
Fluid bed coating, 113–116 191–192 36
Food actives – delivery via emulsions as delivery systems for teeth 37
double emulsions for controlling remineralization actives, 192 38
water-soluble actives, 55–59 for delivering flavors and sensates, 190 39
effect of o/w emulsions on taste release and for delivering throat relief actives, 191 40
perception, 53–55 vs. chewing gum delivery profile, 41
water-in-oil emulsions for controlling 189–190 42
water-soluble actives, 53 Lutein and leutin ester, 27 43
Food emulsions, formulation design for, 47–48 bioavailability, 27 44
aqueous phase design, 48–49 role in age-related-macular degeneration, 45
choice of lipid phase, 49–50 26–27 S46
interfacial formulation and design, 50–52 solubilization, 29 N47
Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 238

238 Index

1 Lycopene, 19 Nonsoluble nutraceuticals, solubilization of,


2 bioavalaibility, 20 17–19
3 formulation as additive in food systems, 20 lutein and lutein ester, 26–28
4 hydrophilic–lipophilic balance of lycopene, 19–24
5 surfactant, 23–24 phytosterols, 24–26
6 solubilization capacity, 20–23 vitamin E, 28–30
7 Nutraceuticals in nanosized self-assembled
8 Micellar systems, encapsulation in, 7–8 liquid (NSSL) vehicles, 13–17
9 Microcapsules, manufacturing of, 4–5 bioavailability, 31–34
10 Microemulsion, 13, 30 oxidative stability, 30–31
11 formulation and characterization, 14–15 solubilization of, 15
12 industrial applications, 13–14 solubilization of nonsoluble.
13 phase diagram, 13–14 See Nonsoluble nutraceuticals,
14 Microencapsulated actives for packaging solubilization of
15 applications U-type microemulsions, swollen micelles,
16 antimicrobial food packaging materials, and progressive and full dilution, 17
17 210–212 water binding, 34–35
18 insect and/or rodent repellent food
19 packages, 212–213 Oil-in-water microemulsions, 18
20 scented fragrance-inserts and flavor- Oral cavity. See also Drug delivery via oral
21 releasing systems, 213–215 route; Oral transport routes,
22 Microencapsulated flavors market, 235–237 physiological and structural basis of
23 Microencapsulated food ingredients, 224 division, 172
24 competitive factors, 233–234 esophagus, 173–174
25 industry structure, 230–233 permeability and barrier functions, 174
26 key end-user groups, 233 tongue, 173
27 market drivers, 226–227 trigeminal nerve, 174
28 market restraints, 228–229 Oral delivery routes
29 pricing, 229–230 local, 172, 181
30 Microencapsulated pigments, 215–216 systemic, 172, 181
31 inks and time–temperature indicators, Oral transport routes, physiological and
32 216–217 structural basis of
33 Microencapsulated probiotics market, epithelial membranes, 175
34 237–238 keratinization, 177
35 Microencapsulated salts and acids market, 237 membrane coating granules, 177
36 Microencapsulated vitamins market, 237 oral mucosa, 175
37 Microporous matrices, encapsulation in, 7 pH, 178
38 Molecular sieve, 7 plasma membranes, 174–175
39 Monodispersed emulsions, 73 polarity, 177–178
40 Mucoadhesion, 192 saliva, 176–177
41
42 Nanoemulsions, 143–144 Paraffin wax, 119
43 Nanoparticles, 144 Particle Coating Technologies, 233
44 Nature-made emulsions Particle Dynamics, Inc., 232–233
45 oil or lipid bodies, 69–71 Passive diffusion, 179
46S plant cells, 72 Payload, 2
47N yeast cells, 71–72 Phase diagram, 13–14, 16
Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 239

Index 239

Physical release, 225 Release rates, 10–11 1


Phytochemicals Release triggers, 2 2
bioconjugation, 144 for emulsions, 52–53 3
encapsulation and controlled release, Release 4
140–143 delayed, 1 5
encapsulation by nanoemulsion, 143–144 sustained, 1 6
Phytosterols 7
chemical structure, 24–25 SDP method, 101–104 8
solubilization capacity, 25–26 Seamless capsules, 194–196 9
Plasticizers, 123 Shellac resin, 119 10
Pressed tablets, 196 Solid fat index, 122 11
Probiotic capsules, manufacturing Solubilization capacity, 24 12
conditions for Solubilization efficacy, 24 13
encapsulating according to experimental Spray congealing, 116–117 14
design, 98–99 Spray dryers, 117 15
optimal manufacturing conditions Spray drying, 3–4, 5, 84 16
verification, 105 Steroid alcohols. See Phytosterols 17
optimization using genetic algorithms, Surfactants, 13 18
104–105 polysorbates and sugar esters, 15 19
optimization using SDP technique, 101–104 Sweeteners, 130 20
performing optimization, 101 21
response surface models and optimization TasteTech, 233 22
model formulation, 99–101 Thermal release, 225 23
screening experiments and experimental 24
design, 96–98 U-type microemulsions, 17 25
Probiotic encapsulation techniques in dairy U-type phase diagram, 16, 17 26
products 27
advantages and disadvantages, 87–89 Vitamin E, 28 28
emulsion, 85–87 solubilization capacity, 28–30 29
extrusion, 84–85 30
spray-drying, 84 Wall-forming materials, 1–2 31
Probiotic survival (effect of encapsulation) in Water binding, 34–35 32
dairy products Wax- and fat-coating material, 33
effect of carrier matrix, 89–92 characteristics of 34
effect of spray drying, 92–93 chain length, 121 35
in dairy products, 93–94 degree of unsaturation, 121 36
in gastrointestinal conditions, 94–96 hydrophobicity, 122 37
Probiotics, 83 melting point, 123–124 38
Protein/polysaccharide coacervation, 136–139 polarity, 121 39
polymorphism, 122–123 40
Release mechansisms solid fat index, 122 41
burst, 10–11 Wax macro-microemulsions, 120 42
combination, 10 43
matrix systems, 9–10 Xanthum gum, 91 44
reservoir-type systems, 9 45
Release modes, 225 Yeasts, 125–127 S46
N47

You might also like