J. Paulo Sousa e Silva, Ana Cristina Freitas - Probiotic Bacteria - Fundamentals, Therapy, and Technological Aspects-Pan Stanford Publishing (2014)
J. Paulo Sousa e Silva, Ana Cristina Freitas - Probiotic Bacteria - Fundamentals, Therapy, and Technological Aspects-Pan Stanford Publishing (2014)
Bacteria
This page intentionally left blank
edited by
J. Paulo Sousa e Silva
Ana C. Freitas
Probiotic
Bacteria
Fundamentals, Therapy, and Technological Aspects
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742
© 2014 by Taylor & Francis Group, LLC
CRC Press is an imprint of Taylor & Francis Group, an Informa business
This book contains information obtained from authentic and highly regarded sources. Reason-
able efforts have been made to publish reliable data and information, but the author and publisher
cannot assume responsibility for the validity of all materials or the consequences of their use. The
authors and publishers have attempted to trace the copyright holders of all material reproduced in
this publication and apologize to copyright holders if permission to publish in this form has not
been obtained. If any copyright material has not been acknowledged please write and let us know so
we may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced,
transmitted, or utilized in any form by any electronic, mechanical, or other means, now known or
hereafter invented, including photocopying, microfilming, and recording, or in any information
storage or retrieval system, without written permission from the publishers.
For permission to photocopy or use material electronically from this work, please access www.
copyright.com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc.
(CCC), 222 Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organiza-
tion that provides licenses and registration for a variety of users. For organizations that have been
granted a photocopy license by the CCC, a separate system of payment has been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and
are used only for identification and explanation without intent to infringe.
Visit the Taylor & Francis Web site at
http://www.taylorandfrancis.com
and the CRC Press Web site at
http://www.crcpress.com
Contents
Foreword ix
Preface xi
Index 295
Contents ix
Foreword
Nagendra P. Shah
Professor of Food Science
School of Biological Sciences
The University of Hong Kong, Hong Kong
Contents xi
Preface
the colon and thus improve host health (Gibson & Roberfroid, 1995).
When we combine probiotic organisms and prebiotics in a product
to obtain a presumably synergistic relationship, the term synbiotic
is used.
Although any microorganism that would produce health
beneits could be considered a probiotic, only some genera have
proven to be probiotic. The genera of bacteria and fungi that have
been employed for their probiotic properties are most commonly
species of Lactobacillus and Biidobacterium and species of the yeast
genus Saccharomyces; other bacterial genera, such as Streptococcus,
Enterococcus, and Bacillus, have also been studied. However, with
regard to the genera Enterococcus and Bacillus, particular concerns
have been raised concerning their safety properties (Hempel et al.,
2011). Some of these genera have been used as single cultures or
in mixed formulations. In a very recent revision, Bengmark (2012)
reported some probiotic starter cultures assumed with no identiiable
adverse effects in clinical studies (e.g., no effect in terms of bacteria
translocation, gastric colonization with enteric organisms, or septic
morbidity, serum C reactive protein levels or mortality). This included
isolated strains such as Lactobacillus plantarum 299v (Pro-Viva) or
L. rhamnosus GG or multiple strains, Ecologic 641 (Winclove Bio
Industries, Amsterdam, the Netherlands), a supplemented synbiotic
composition consisting of six different strains of freeze-dried, viable
bacteria: L. acidophilus, L. casei, L. salivarius, Lactococcus lactis,
Biidobacterium biidum, and B. lactis along with corn-starch and
maltodextrins, Trevis (Chr Hansen Biosystem, Denmark) with L.
acidophilus La5, B. lactis Bb-12, Streptococcus thermophilus, and L.
bulgaricus as well as VSL#3 (VSL Pharmaceuticals, Ft Lauderdale,
Florida, USA) with four strains of Lactobacillus (L. casei, L. plantarum,
L. acidophilus, and L. delbrueckii subsp. bulgaricus) along with three
strains of Biidobacterium (B. longum, B. breve, and B. infantis) and S.
salivarius subsp. thermophilus.
Several health attributes have been ascribed to probiotics, which
has increased commercial interest in exploiting different applications
leading to the rapid growth and expansion of this market sector.
From the selection of a probiotic strain to its incorporation in a inal
product, both in food matrices or pharmaceutical formulations,
several steps must be accomplished if a safe and biological active
product is to be achieved (Fig. 1.1).
Probioc Bacteria 3
Toxicity: ,0
Potenal probioc
Colonizaon:
+
Health benefits:
++
(
Stability
Food Pharmaceucal
applicaons applicaons
References
Bengmark, S. (2013) Gut microbiota, immune development and function.
Pharmacol. Res., 69(1), 87–113.
FAO/WHO (2001) Report on joint FAO/WHO expert consultation on
evaluation of health and nutritional properties of probiotics in food
including powder milk with live acid bacteria, Cordoba, Argentina.
Gibson, G.R. and Roberfroid, M.B. (1995) Dietary modulation of the human
colonic microbiotia: introducing the concept of prebiotics. J. Nutr.,
125, 1401–1412.
Hempel, S., Newberry, S. Ruelaz, A., Wang, Z. Miles, J.N.V., Suttorp, M.J.,
Johnsen, B., Shanman, R. Slusser, W., Fu, N., Smith, A., Roth, B., Polak,
J., Motala, A., Perry, T. and Shekelle, P.G. (2012) Safety of probiotics to
reduce risk and prevent or treat disease. Evidence Report/Technology
Assessment, 200, 1–94.
Saad, N., Delattre, C., Urdaci, M., Schmitter, J.M. and Bressollier, P. (2013) An
overview of the last advances in probiotic and prebiotic ield. LWT -
Food Sci.Technol., 50, 1–16.
This page intentionally left blank
Chapter 2
2.1.1 Introduction
Oral use is the most convenient way for administering probiotic
products not only due to the easiness of the method but also to
promote one of the main effects attributed to probiotics— improving
intestinal microbial balance. Nevertheless, the upper gastrointestinal
tract (GIT) is an adverse medium to probiotics; hence, there are
Figure 2.1 Anatomic structure of the human GIT and accessory digestive
organs.
2.1.2 GIT pH
Among other factors (transit time, bacteria metabolism, and
chemical reactions) that might inluence the performance of
probiotic products, the pH is perhaps the most important due to
general sensibility of probiotics to acidic conditions. According to
Evans et al. (1988), the average pH in stomach is 1.8 and rises to 6.6
in the proximal intestine reaching 7.5 in the distal intestine, and then
it decreases to 6.3 in right colon and it rises again, until it reaches 7.1
in left colon. The pH depends on prandial condition of individuals.
Table 2.1 illustrates physiological pH in humans in the fasting and
fed states (Gray and Dressman, 1996; Dressman et al., 1998).
Apart from prandial condition, the pH in GIT is a function of
many variables, including meal volume and content, and volume
of secretions (Mudie et al., 2010). It should be noted that the
extrapolation of GIT pH data from healthy situations to pathological
conditions must be made with precaution.
10 Gastrointestinal Tract
Table 2.1 Physiological pH in the GIT in the fasted and fed states
2.1.5 Stomach
Stomach has a J-shape with an approximate capacity of 1.5 l, but in a
fasting state, it normally contains 20–30 ml of luid (predominantly
wet mucus). Stomach acts as a reservoir for food while it is mixed
with acid, mucus, and pepsin in order to be released (as chime) into
the duodenum at a controlled rate. The gastric emptying time may
vary from few minutes to several hours, depending on the time of the
last food ingestion, anxiety, position, and level of individual activity,
among other factors.
During fasting, an interdigestive cycle of motility (migrating
myoelectric complex, MMC) composed of four phases governs
stomach activity. This cycle begins in the stomach and moves along
the small intestine into the distal ileum. The Phase III of MMC (a burst
of contractile activity of 5–15 min) opens the pylorus and clears the
stomach. The ingestion of food stops the cycle.
The peristaltic movements appear 5–10 min after the intake of
foods and remain until the gastric emptying, which lasts from 1 to
several hours depending on the meal composition (Phillips, 1993).
The hydrochloric acid secreted by parietal cells kills many bacteria
and provides the pH for pepsin to begin protein digestion (Ganong,
2005).
Gut
Microbiota
Functional
Foods Metabolites
Role in Host
Physiology
and Health
"
!
!!!
#
"#$
$%
!
"#$
!
!
%
!
"#$&
'
!
*
!!
Metabolic endotoxaemia
Figure 2.4 Possible mechanisms for describing how gut microbiota are
involved in the onset of obesity and associated metabolic
disorders. Adapted from Cani and Delzenne, 2011.
2.2.2.3 Immunomodulation
The immune regulatory function of the intestinal microbiota
consists of priming the mucosal immune system and maintenance
of intestinal epithelium homeostasis. The intestinal microbiota may
inluence the content of lamina propria T cells, immunoglobulin A
producing B cells, intraepithelial T cells, and serum immunoglobulin
levels (Tlaskalová-Hogenová et al., 2004). As already stated, studies
in germ-free animals demonstrated impairment of immune function
in the absence of the intestinal microbiota. Defective development of
gut-associated lymphoid tissues, antibody production (i.e., sIgA), and
maturation of isolated lymphoid follicles have been shown in germ-
free animals, together with reduced Peyer’s patches and mesenteric
lymph node number and dimension (Bouskra et al., 2008).
In this symbiotic relationship, it is also important that immune
system adequately recognizes microbial components and establishes
a state of tolerance towards them (Aureli et al., 2011). According
to Aureli et al. (2011), such tolerance of microbiota is mediated
by complementary mechanisms, including (i) microbiota bacteria
do not express virulence factors such as antigenic structures
recognized by toll-like receptors on the surface of immune system
22 Gastrointestinal Tract
(Continued)
24 Gastrointestinal Tract
thiamine, nicotinic acid, folic acid, pyridoxine, biotin, and B12 (Lee
et al., 1999), oligosaccharides, organic acids (Bhat and Bhat, 2011),
and polyunsaturated fatty acids such as conjugated linoleic acid
(Rodrigues et al., 2011; Rodriguez-Alcalá et al., 2011). Furthermore,
iron bioavailability may be increased by L. acidophilus and bile acids
may be deconjugated by numerous lactobacilli.
An overall review of the mechanistic insights that many studies
have provided concerning probiotics and their interactions with
the composition and function of the intestinal mucosa via speciic
molecules (cell wall components such as peptidoglycan, teichoic
acids or capsular polysaccharides, and immunomodulatory proteins)
is given by Bron et al. (2012). In this review, the importance of
human individuality is discussed as previously mentioned and the
possibility of a personalized application of probiotics in the near
future is proposed (Bron et al., 2012). These mechanisms will be
further probed in Chapter 3.
Criteria for rational use of probiotics will be given in Chapter 7,
but in summary may include adequate identiication and functional
characterization of bacteria; administration of an appropriate
quantity of bacteria (which is inluenced by nature of strain
employed, whether in combination with other strains of same or
different species, and by the food/pharmaceutical matrix selected)
and their enumeration; beneits for the host and their deinition; and
safety of use.
model that simulates food transit through the upper part of the GIT to
enable us to evaluate the probiotic potential of food products. More
recently, Mercuri et al. (2010) created the dynamic gastric model,
which was the irst dynamic in vitro model of the human stomach,
and as such, it is able to mimic the digestive processes of the gastric
compartment not only in terms of temporally varying enzymatic and
acid secretions but also in terms of simulation of the mechanical
forces exerted by the muscular tissue of the stomach, an important
model for assessment of digestion and its impact on probiotics.
2.4 Conclusions
A balanced host–intestinal microbiota relationship is essential
for intestinal homeostasis due to important roles in digestive,
immune, and metabolic functions. Gut microbiota provides an array
of functions for the host ranging from epithelial barrier function,
through protection against infectious diseases to energy recovery
from nutrients, generating SCFAs with different physiological
roles. Different studies using advanced analytical strategies have
highlighted the importance of modulation of intestinal microbiota
toward the improvement of human health.
Speciic dietary intervention programs involving food-based
strategies or supplements, such as probiotics and prebiotics, are
constantly being proposed as relevant solutions to be used in
the management of microbiota-associated disorders. The use of
probiotics in the treatment and prevention of gastrointestinal
diseases has yielded important results, and in order to select the
most adequate binomial strain-effect in vitro, gastrointestinal models
are available for a irst phase of development.
References
Anderson, R.C., Cookson, A.L., McNabb, W.C., Park, Z., McCann, M.J., Kelly, W.J.,
Roy, N.C. 2010. Lactobacillus plantarum MB452 enhances the function
of the intestinal barrier by increasing the expression levels of genes
involved in tight junction formation. BMC Microbiology 10, 316.
Atarashi, K., Tanoue, T., Shima, T., Imaoka, A., Kuwahara, T., Momose, Y.,
Cheng, G., Yamasaki, S., Saito, T., Ohba, Y., et al. (2011). Induction of
colonic regulatory T cells by indigenous Clostridium species. Science
331, 337–341.
Aura, A.-M, Karppinen, S., Virtanen, H., Forssell, P., Heinonen, S.-M., Nurmi,
T., Adlercreutz, H. and Poutanen, K. (2005a). Processing of rye bran
inluences both the fermentation of dietary ibre and the bioconversion
of lignans by human faecal lora in vitro. Journal of Science and Food
Agriculture 85(12), 2085–2093.
Aura, A.-M., Martin-Lopez, P., O´Leary, K.A., Williamson, G., Oksman-
Caldentey, K.-M., Poutanen, K. and Santos-Buelga, C. (2005b). In vitro
metabolism of anthocyanins by human gut microlora. European
Journal of Nutrition 44, 133–142.
Aura, A.-M., O´Leary, K.A., Williamson, G., Ojala, M., Bailey, M., Puupponen-
Pimiä, R., Nuutila, A.M., Oksman-Caldentey, K.-M. and Poutanen, K.
(2002). Quercetin derivatives are deconjugated and converted to
hydroxyphenylacetic acids but not methylated by human fecal lora in
vitro. Journal Agriculture Food Chemistry 50, 1725–1730.
Aureli, P., Capurso, L., Castellazzi, A.M., Clerici, M., Giovannini, M., Morelli, L.,
Poli, A., Pregliasco, F., Salvini, F. and Zuccotti, G.V. (2011). Probiotics
and health: an evidence-based review. Pharmacological Research 63,
366-376.
van Baarlen, P., Troost, F., van der Meer, C., Hooiveld, G., Boekschoten, M.,
Brummer, R.J. and Kleerebezem, M. (2011). Human mucosal in vivo
transcriptome responses to three lactobacilli indicate how probiotics
may modulate human cellular pathways. Proceedings National Academy
Science USA 108(suppl 1), 4562-4569.
Bäckhed, F., Ding, H-, Wang, T., Hooper, L.V., Koh, G.Y., Nagy, A., Semenkovich,
C.F., Gordon, J.I. (2004). The gut microbiota as an environmental factor
that regulates fat storage. Proceedings National Academy Sciences USA
101, 15718–15723.
Barnich, N., Carvalho, F.A., Glasser, A-L., Darcha, C., Jantscheff, P., Allez, M.,
Peeters, H., Bommelaer, G., Desreumaux, P., Colombel J-F. and Darfeuille-
Michaud, A. (2007). CEACAM6 acts as a receptor for adherent invasive
E. coli, supporting ileal mucosa colonization in Crohn disease. Journal
Clinical Investigation 117, 1566–1574.
References 35
Barry, J.L., Hoebler, C., Macfarlane, G.T., Macfarlane, S., Mathers, J.C., Reed, K.A.,
Mortensen, P.B., Norgaard, I., Rowland, I.R. and Rumney, C.J. (1995).
Estimation of the fermentability of dietary ibre in vitro: A European
interlaboratory study. British Journal of Nutrition 74, 303–322.
Berg, R.D. (1996). The indigenous gastrointestinal microlora. Trends in
Microbiology 433, 430-435.
Bernalier-Donadille, A. (2010). Fermentative metabolism by the human gut
microbiota. Gastroentérologie Clinique et Biologique 34, S16–S22.
Bevins, C.L. and Salzman, N. H. (2011). Paneth cells, antimicrobial
peptides and maintenance of intestinal homeostasis. Nature Reviews
Microbiology 9, 356–368.
Bouskra, D., Brezillon, C., Berard, M., Werts, C., Varona, R., Boneca, I.G. and
Eberl, G. (2008). Lymphoid tissue genesis induced by commensals
through NOD1 regulates intestinal homeostasis. Nature 456, 507–
510.
Bowly, E., Adlercreutz, H. and Rowland, I. (2003). Metabolism of isolavones
and lignans by the gut microlora: a study in germ-free and human
lora associated rats. Food Chemistry Toxicology 41, 631–636.
Bron, P.A., Van Baarlen, P. and Kleerebezem, M. (2011). Emerging molecular
insights into the interaction between probiotics and the host intestinal
mucosa. Nature Reviews Microbiology 10, 66–78.
Brown, C.T., Davis-Richardson, A.G., Giongo, A., Gano, K.A., Crabb, D.B.,
Mukherjee, N., Casella, G., Drew, J.C., Ilonen, J., Knip, M., Hyoty, H., Veijola,
R., Simell, T., Simell, O., Neu, J., Wasserfall, C.H., Schatz, D., Atkinson,
M.A. and Triplett, E.W. (2011). Gut microbiome metagenomics analysis
suggests a functional model for the development of autoimmunity for
type 1 diabetes. PLoS One 6(10), e25792.
Cani, P.D., Possemiers, S., van de Wiele, T., Guiot, Y., Everard, A., Rottier,
O., Geurts, L., Naslain, D., Neyrinck, A., Lambert, D.M., Muccioli,
G.G. and Delzenne, N.M. (2009). Changes in gut microbiota control
inlammation in obese mice through a mechanism involving GLP-2-
driven improvement of gut permeability. Gut 58, 1091−1103.
Cani, P.D. and Delzenne, N.M. (2011). The gut microbiome as therapeutic
target. Pharmacology & Therapeutics 130, 202–212.
Collado, M.C., Isolauri, E., Laitinen, K. and Salminen, S. (2008). Distinct
composition of gut microbiota during pregnancy in overweight and
normal-weight women. American Journal Clinical Nutrition 88, 894–
899.
36 Gastrointestinal Tract
Evans, D.F., Pye, G., Bramley, R., Clark, A.G., Dyson, T.J. and Hardcastle, J.D.
(1988). Measurement of gastrointestinal pH proiles in normal
ambulant human subjects. Gut 29, 1035–1041.
Evans, D. (1993). Gastrointestinal tract physiology ant its relevance to drug
delivery. Current status on targeted drug delivery to the gastrointestinal
tract, London, Capsugel Library.
Fallani, M., Young, D., Scott, J., Norin, E., Amarri, S., Adam, R., Aguilera, M.,
Khanna, S., Gil, A., Edwards, C., Doré, J. and other members of the
INFABIO team. (2010). Intestinal microbiota of 6-week-old infants
across Europe: geographic inluence beyond delivery mode, breast-
feeding, and antibiotics. Journal Pediatrics Gastroenterology Nutrition
51, 77–84.
Falony, G., Vlachou, A., Verbrugghe, K. and De Vuyst, L. (2006). Crossfeeding
between Biidobacterium longum BB536 and acetate-converting,
butyrate-producing colon bacteria during growth on oligofructose.
Applied Environmental Microbiology 72, 7835–7841.
Fanaro, S., Chierici, R., Guerrini, P. and Vigi, V. (2003). Intestinal microlora
in early infancy: composition and development. Acta Paediatrica 91,
48–55.
Finegold, S.M., Sutter, V.L. and Mathison, G.E. (1983). Normal indigenous
lora. In: Human intestinal microlora in health and disease, pp. 3–31.
Edited by D.J. Hengtes. New York, Academic Press.
Frank, D.N., St. Amand, A.L., Feldman, R.A., Boedeker, E.C., Harpaz, N., Pace,
N.R. (2007). Molecular-phylogenetic characterization of microbial
community imbalances in human inlammatory bowel diseases.
Proceedings National Academy Science U.S.A. 104, 13780–13785.
Gao, K., Xu, A.L., Krul, C., Venema, K., Liu, Y., Niu, Y.T., Lu, J., Bensoussan, L.,
Seeram, N.P., Heber, D. and Henning, S.M. (2006). Of the major phenolic
acids formed during human microbial fermentation of tea, citrus, and
soy lavonoid supplements, only 3, 4-dihydroxyphenylacetic acid has
antiproliferative activity. Journal of Nutrition 136, 52–57.
Ganong, W. (2005) Regulation of gastrointestinal function, Review of Medical
Physiology Lange Medical Books/McGraw-Hill.Glitso, L.V., Mazur, W.M.,
Adlercreutz, H., Wähälä, K., Mäkelä, T., Sandström, B. and Bach Knudsen,
K.E. (2000). Intestinal metabolism of rye lignans in pigs. British Journal
of Nutrition 84, 429-437.
Gmeiner, M., Kneifel, W., Kulbe, K.D., Wouters, R., De Boever, P., Nollet, L. and
Verstraete, W. (2000). Inluence of a synbiotic mixture consisting of
Lactobacillus acidophilus 74-2 and a fructooligosaccharide preparation
on the microbial ecology sustained in a simulation of the human
38 Gastrointestinal Tract
Mainville, I., Arcand, Y. and Farnworth, E.R. (2005). A dynamic model that
simulates the human upper gastrointestinal tract for the study of
probiotics. International Journal of Food Microbiology 99, 287–296.
Mäkivuokko, H., Nurmi, J., Nurminen, P., Stowell, J. and Rautonen, N. (2005).
In vitro effects of polydextrose on colon fermentation and mucosal
cyclooxygenase gene expression. Nutrition Cancer 52(1), 94–104.
Martin, F.P., Dumas, M.E., Wang, Y., Legido-Quigley, C., Yap, I.K., Tang, H.,
Zirah, S., Murphy, G.M., Cloarec, O., Lindon, J.C., Sprenger, N., Fay, L.B.,
Kochhar, S., van,Bladeren, P., Holmes, E. and Nicholson, J.K. (2007). A
top-down systems biology view of microbiome-mammalian metabolic
interactions in a mouse model. Molecular Systems Biology 3, 112:
1–16.
Martin, F-P.J., Collino, S., Rezzi, S. and Kochhar, S. (2012). Metabolomic
applications to decipher gut microbial metabolic inluence in health
and disease. Frontiers in Physiology 3, 1–11 (article 113).
Martín, R., Langa, S., Reviriego, C., Jiménez, E., Marín, M. L., Olivares, M.,
Boza, J., Jiménez, J., Fernández, L., Xaus, J., Rodríguez, J.M. (2004).
The commensal microlora of human milk: new perspectives for food
bacteriotherapy and probiotics. Trends in Food Science and Technology
15, 121–127.
Martín, R., Jiménez, E., Heilig, H., Fernández, L., Marín, M.L., Zoetendal, E.G.,
Rodríguez, J.M. (2009). Isolation of biidobacteria from breast milk
and assessment of the biidobacterial population by PCR-denaturing
gradient gel electrophoresis and quantitative real-time PCR. Applied
and Environmental Microbiology 75, 965–969.
Marzorati, M., Verhelst, A., Luta, G., Sinnott, R., Verstraete, W., Van de
Wiele, T. and Possemiers, S. (2010). In vitro modulation of the human
gastrointestinal microbial community by plant-derived polysaccharide-
rich dietary supplements. International Journal of Food Microbiology
139, 168–176.
Mercuri, A. (2010). The dynamic gastric model: a new in vitro model for
biorelevant dissolution and delivery assessment of oral dosage forms.
UKICRS Newsletter, 28–30.
Minekus, M., Marteau, P., Havenaar, R. and Huis in ‘t Veld, J.H.J. (1995). A
multi-compartmental dynamic computer-controlled model simulating
the stomach and small intestine. Alternatives to Laboratory Animals
(ATLA) 23, 197–209.
Minekus, M., Smeets-Peeters, M., Bernalier, A., Marol-Bonnin, S., Havenaar,
R., Marteau, P., Alric, M., Fonty, G. and Huis in´t Veld, J.H.J. (1999). A
computer-controlled system to simulate conditions of the large
References 41
Rowland, I.R., Bearne, C.A., Fischer, R. and Pool-Zobel, B.L. (1996). The effect
of lactulose on DNA damage induced by DMH in the colon of human
lora associated rats. Nutrition Cancer 26, 37–47.
Salzman, N.H. (2011). Microbiota-immune system interaction: an uneasy
alliance. Current Opinion Microbiology 14, 99–105.
Sanders, M.E. (2007). Probiotics: strains matter. Functional Food and
Nutraceuticals. June, 34–41.
Sanders, M.E. (2011). Impact of probiotics on colonizing microbiota of the
gut. Journal of Clinical Gastroenterology 45(Suppl), S115–S119.
Sanders, M.E. and Marco, M.L. (2010). Food formats for effective delivery of
probiotics. Annual Reviews on Food Science and Technology 1, 65–85.
Sarbini, S.R., Kolida, S., Naeye, T., Einerhand, A., Brison, Y., Remaud-Simeon, M.,
Monsan, P., Gibson, G.R. and Rastall, R.A. (2011). In vitro fermentation
of linear and α-1,2-branched dextrans by the human fecal microbiota.
Applied and Environmental Microbiology, 77(15), 5307–5315.
Schippa, S., Iebba, V., Barbato, M., Nardo, G.D., Totino, V., Checchi, M.P.,
Longhi, C., Maiella, G., Cucchiara, S. and Conte, M.P. (2010). A distinctive
‘microbial signature’ in celiac pediatric patients. BMC Microbiology 10,
175–184.
Schlee, M., Harder, J., Köten, B., Stange, E.F., Wehkamp, J. and Fellermann, K.
(2008). Probiotic lactobacilli and VSL#3 induce enterocyte β-defensin
2. Clinical Experimental Immunology 151, 528–535.
Sekirov, I., Russell, S.L., Antunes, L.C.M. and Finlay, B.B. (2010). Gut microbiota
in health and disease. Physiology Reviews 90, 859–904.
Sokol, H., Pigneur, B., Watterlot, L., Lakhdari, O., Bermúdez-Humarán, L.G.,
Gratadoux, J.J., et al. (2008). Faecalibacterium prausnitzii is an anti-
inlammatory commensal bacterium identiied by gut microbiota
analysis of Crohn disease patients. Proceedings National Academy
Sciences U.S.A. 105, 16731–16736.
Sokol, H., Seksik, P. Furet, J.P., Firmesse, O., Nion-Larmurier, I., Beaugerie,
L., et al. (2009). Low counts of Faecalibacterium prausnitzii in colitis
microbiota. Inlammatory Bowel Disease 15, 1183–1189.
Sousa, T., Paterson, R., Moore, V., Carlsson, A., Abrahamsson, B. and
Basit, A.W. (2008). The gastrointestinal microbiota as a site for the
biotransformation of drugs. International Journal Pharmacology 363,
1–25.
Spratt, P., Nicolella, C. and Pyle, D.L. (2005). An engineering model of the
human colon. Food and Bioproducts Processing 83(2), 147–157.
44 Gastrointestinal Tract
Sumeri, I., Arike, L., Adamberg, K. and Paalme, T.(2008). Single bioreactor
gastrointestinal tract simulator for study of survival of probiotic
bacteria. Applied Microbiology and Biotechnology 80, 317–324.
Tlaskalová-Hogenová, H., Stepánková, R., Hudcovic, T., Tucková, L.,
Cukrowska, B., Lodinová-Zádníková, R., et al. (2004). Comensal bacteria
(normal microlora), mucosal immunity and chronic inlammatory and
autoimmune diseases. Immunology Letters 93, 97–108.
Tuohy, K.M., Hinton, D.J.S., Davies, S.J., Crabbe, M.J.C., Gibson, G.R. and Ames,
J.M. (2006). Metabolism of maillard reaction products by the human
gut microbiota—implications for health. Molecular Nutrition Food
Research 50, 847–857.
Tuohy, K.M., Rouzaud, G.C., Bruck, W.M., and Gibson, G.R. (2005). Modulation
of the human gut microlora towards improved health using prebiotics-
assessment of eficacy. Current Pharmaceutical Design 11, 75–90.
Turnbaugh, P.J., Bäckhed, F., Fulton, L. and Gordon J.I. (2008). Diet-induced
obesity is linked to marked but reversible alterations in the mouse
distal gut microbiome. Cell Host Microbe 3(4), 213–223.
Tzounis, X., Vulevic, J., Kuhnle, G.G.C., George, T., Leonczak, J., Gibson, G.R.,
KwikUribe, C. and Spencer, J.P.E. (2008). Flavanol monomer induced
changes to the human faecal microlora. British Journal of Nutrition 99,
782–792.
Uronis, J.M., Mühlbauer, M., Herfarth, H.H., Rubinas, T.C., Jones, G.S. and
Jobin, C. (2009). Modulation of the intestinal microbiota alters colitis-
associated colorectal cancer susceptibility. PLoS ONE 4(6), e6026.
Vaarala, O., Atkinson, M.A. and Neu, J. (2008). The “perfect storm” for type
1 diabetes: The complex interplay between intestinal microbiota, gut
permeability, and mucosal immunity. Diabetes 57(10), 2555–2562.
Vanhoutte, T., Huys, G., Brandt, E. and Swings, J. (2004) Temporal stability
analysis of the microbiota in human feces by denaturing gradient gel
electrophoresis using universal and group-speciic 16S rRNA gene
primers. FEMS Microbiology Ecology 48, 437–446.
Walker A. (2010). Breast milk as the gold standard for protective nutrients.
Journal of Pediatrics 156, S3–S7.
Walle, T. (2004). Absorption and metabolism of lavonoids. Free Radical
Biology and Medicine 36, 829–837.
Wen, L., et al. (2008). Innate immunity and intestinal microbiota in the
development of Type 1 diabetes. Nature 455, 1109–1113.
Wikoff, W.R., Anfora, A.T., Liu, J., Schultz, P.G., Lesley, S.A., Peters, E. C. and
Siuzdak, G. (2009). Metabolomics analysis reveals large effects of gut
References 45
!
"
#
*#
$
Number of
Study Regimen patients Main results
Hamilton- L. acidophilus 8 × 1010 for 20 Four patients
Miller, 2003 8 weeks cleared
L. acidophilus NSA in 14 Six patients cleared
“acidophilus milk” c. 5 ×
10 9 cfu/day for 8 weeks
L. johnsonii La1 20 Breath test values
supernatant, 200 ml for 2 reduced
weeks
B. longum BB536 2.5 × 68 Breath test values
1010 cfu/day for 4 weeks reduced
in pasteurized yogurt
L. gasseri OLL 2716 c. 2 × 31 Breath test and
109 cfu/day for 8 weeks in pepsinogen values
yogurt base reduced
Yogurt made with six 27 Breath tests
“lactic acid” strains 7 × 109 remained positive
cfu/day for 30 days in
26/27
Myllyluoma L. rhamnosus GG (LGG), 13 Breath test and
et al., 2007 L. rhamnosus LC705, P. gastrin-17 reduced
freudenreichii
ssp. shermanii JS, and B.
lactis Bb12, 2.5 × 109 cfu/
day for 8 weeks
Gotteland L. acidophilus La1 product 295 Breath test values
et al., 2008 (80 ml) daily with >107 reduced and
cfu/ml for 3 weeks eradication of 14.9%
alone or combined with for La1 and 22.9%
cranberry jus when combined with
cranberry juice
Inhibition of Helicobacter pylori and Intestinal Pathogens 59
(Continued)
66 Probiotics and Their Therapeutic Role
et al. (2004).
73
74 Probiotics and Their Therapeutic Role
strain GG before and after birth split the risk of eczema [95%
conidence interval (CI) for relative risk, 0.3–0.8] but not that of
allergen sensitization. In a large and recent study of 925 mother–
infant pairs, prenatal administration of probiotics (containing four
bacterial strains) during the last month of gestation and postnatal
administration of probiotics and prebiotics from birth to age 6
months resulted in short-lived changes in the neonatal gut lora
and diminished the incidence of atopic eczema (but had no effect
on other atopic ailments or allergic sensitization) at age 2 years.
In general, these studies demonstrate an increased production of
cytokines by Treg cells (IL-10 and TGF-β) associated with a reduced
severity of atopic dermatitis in a small number of infants.
Concerning prevention of allergy, no study showed signiicant
evidences to recommend probiotics. Regardless of all of the
immunomodulatory effects described in experimental models, so
far, none of these studies has shown any clear effect on preventive
sensitization or any allergic disease other than eczema (Prescott and
Bjorkste, 2007), which alerts us to the needs of further systematic
studies. However, the study (Van Overtvelt et al., 2010) of 11 lactic
acid bacteria as an adjuvant for sublingual allergy vaccines has been
performed as a promising application, in which they compared
immunomodulatory properties of 11 strains as well as their capacity
to enhance sublingual immunotherapy eficacy in a murine asthma
model, proving that probiotics acting as a Th1/possibly Treg, but
not Th1 adjuvant, potentiate tolerance induction via the sublingual
route.
3.12 Conclusions
The different mechanisms of action explored in this chapter may lead
to relevant health beneits, namely in infection defence, prevention
of cancer, in stabilizing or reconstituting the physiological balance
between the intestinal microbiota and its host, and inluencing
immunological processes at different locations. However, it must be
highlighted that one probiotic cannot exhibit all activities at the same
time, at least not to that extent that it could be used for the prevention
or cure of all mentioned kinds of disease. The overall effect attributed
to a certain probiotic strain depends on the metabolic properties, the
kind of surface molecules expressed, and components to be secreted
with probiotic actions (Oelschlaeger, 2010).
The studies analyzed during this chapter will increase our
knowledge concerning the functional attributes of known probiotics
and facilitate probiotic product improvements.
The capability to control and envisage probiotic-based effects
will permit both industry and the consumer to choose, based on
science, the probiotics with well-known health beneits. These
multidisciplinary efforts to develop more probiotics to be delivered
through medication or food additives will increase available solutions
to ight old and new diseases.
References
Allakera, R.P. and Douglas, I. (2009) Novel anti-microbial therapies for dental
plaque-related diseases. Int. J. Antimic. Agents, 33, 8–13.
Armuzzi, A., Cremonini, F., Ometti V., Bartolozzi, F., Canducci, F., Candelli, M.,
et al. (2001) Effect of Lactobacillus GG supplementation on antibiotic-
associated gastrointestinal side effects during Helicobacter pylori
eradication therapy: a pilot study. Digestion, 63, 1–7.
Baroutkoub, A., Mehdi, R.Z., Beglarian, R., Hassan, J., Zahra, S., Mohammad,
M. S. and Hadi, E.M. (2010) Effects of probiotic yoghurt consumption
on the serum cholesterol levels in hypercholestromic cases in Shiraz
Southern Iran. Sci. Res. Essays, 5, 2206–2209.
Barrons, R. and Tassone, D. (2008) Use of Lactobacillus probiotics for
bacterial genitourinary infections in women: a review. Clin. Ther., 30,
453–468.
86 Probiotics and Their Therapeutic Role
Bhatia, S.J., Kochar, N., Abraham, P., Nair, N.G. and Mehta, A.P. (1989)
Lactobacillus acidophilus inhibits growth of Campylobacter pylori in
vitro. J. Clin. Microbial., 27, 2328–2330.
Bosch, M., Nart, J., Audivert, S, Bonachera, M.A.,Alemany, A.S, Fuentes, M.C
and Jordi C. (2011). Isolation and characterization of probiotic strains
for improving oral health. Arch. Oral Biol., 57, 539–549.
Boutron-Ruault, M.C. (2007) Probiotics and colorectal cancer. Nutrition
Clinique et Métabolisme, 21, 85–88.
Canducci, F, Cremonini, F., Armuzzi, A., Di Caro, S., Gabrielli, M., Santarelli,
L., Nista. E., Lupascu, A., De Martini, D. and Gasbarrini, A. (2002a)
Probiotics and H. pylori eradication. Digest. Liver Dis., 34, S81–S83.
Canducci, F., Armuzzi, A., Cremonini, F., Cammarota, G., Bartolozzi, F., Pola,
P., et al. (2002b) A lyophilized and inactivated culture of Lactobacillus
acidophilus increases Helicobacter pylori eradication rates. Aliment
Pharmacol Ther., 14, 1625–1629.
Cabré, E. and Gassull, M.A. (2007) Probiotics for preventing relapse or
recurrence in Crohn’s disease involving the ileum: are there reasons
for failure? J. Crohn’s Colitis, 1, 47–52.
Cani, P.D. and Delzenne, N.M. (2009) The role of the gut microbiota in energy
metabolism and metabolic disease. Curr. Pharm. Des., 15, 1546–1558.
Cani, P.D., Neyrinck, A.M., Fava, F., Knauf, C., Burcelin, R.G., Tuohy, K.M., Gibson,
G.R. and Delzenne, N.M. (2007) Selective increases of biidobacteria in
gut microlora improve high-fat-dietinduced diabetes in mice through
a mechanism associated with endotoxaemia. Diabetologia, 50, 2374–
2383.
Capurso, G., Marignani, M. and Fave, G.D. (2006) Probiotics and the incidence
of colorectal cancer: when evidence is not evident. Digest. Liver Dis.,
38, Suppl. 2, S277–S282.
Chalk, C.S. and Chalk, A.J. (2003) Probiotics and allergy: current understand-
ing and application for the PCCP (Primary Care Chiropractic Physician).
J. Chiropractic Med., 3, 131–133.
Chen, Z., Ma K.Y., Liang Y., Peng C. and Zuo Y. (2011) Role and classiication of
cholesterol-lowering functional foods. J. Functional Foods, 3, 61–69.
Colbére-Garapin, F., Martin-Latil, S., Blondel, B., Mousson, L., Pelletier, I.,
Autret, A., Francois, A., Niborski, V., Grompone, G., Catonnet, G., and van
de Moer, A. (2007) Prevention and treatment of enteric viral infections:
possible beneits of probiotic bacteria. Microbes Infect., 9, 1623–1631.
Commane, D., Hughe, R., Shortt, C. and Rowland, I. (2005) The potential
mechanisms involved in the anti-carcinogenic action of probiotics.
Mutation Res., 591, 276–289.
References 87
Desbonnet, L., Garrett, L., Clarke, G., Kiely, B., Cryana, J.F. and Dinana, T.G.
(2010) Effects of the probiotic Biidobacterium infantis in the maternal
separation model of depression. Neuroscience, 170, 1179–1188.
Fazeli, H., Moshtaghian, J., Mirlohi, M. and Shirzadi, M. (2010) Reduction
in serum lipid parameters by incorporation of a native strain of
Lactobacillus plantarum A7 in mice. Iranian J. Diabetes Lipid Disord.,
9, 1–7.
Felley, C. and Michetti, P. (2003) Probiotics and Helicobacter pylori. Best
Practice Res.Clin. Gastroent., 17, 785–791.
Figueroa-González, I., Cruz-Guerrero A. and Quijano G. (2011) The beneits
of probiotics on human health. J. Microbial. Biochem. Technol., S1:003.
Fiocchi, C. (2006) Probiotics in inlammatory bowel disease: yet another
mechanism of action? Gastroenterology, 131, 2009–2011.
Fooks L.J. and Gibson, G.R. (2003) Mixed culture fermentation studies on the
effects of synbiotics on the human intestinal pathogens Campylobacter
jejuni and Escherichia coli. Anaerobe, 9, 231–242.
Forestier, C., De Champs, C., Vatoux, C. and Joly, B. (2001) Probiotic activities
of Lactobacillus casei rhamnosus: in vitro adherence to intestinal cells
and antimicrobial properties. Res. Microbiol. 152, 167–173.
Forsytha, C. B., Farhadia, A. Jakate, S.M., Tanga, Y. Shaikha, M. and
Keshavarziana, A. (2010) Lactobacillus GG treatment ameliorates
alcohol-induced intestinal oxidative stress, gut leakiness, and liver
injury in a rat model of alcoholic steatohepatitis. Alcohol, 43, 163–
172.
Geier, M.S., Butler, R.N. and Howarth, G.S. (2007) Inlammatory bowel
disease: current insights into pathogenesis and new therapeutic
options; probiotics, prebiotics and synbiotics. Int. J. Food Microbiol.,
115, 1–11.
Glass, M.D., Courtneyb, P.D., LeJeunea, J.T., and Warda, L.A. (2004) Effects
of Lactobacillus acidophilus and Lactobacillus reuteri cell-free
supernatants on Cryptosporidium viability and infectivity in vitro. Food
Microbiol., 21, 423–429.
Gotteland, M., Andrews, M., Toledo, M., Muñoz, L., Speisky,H. and Salazar, G.
(2008) Modulation of Helicobacter pylori colonization with cranberry
juice and Lactobacillus johnsonii La1 in children. Nutrition, 24, 421–
426.
Gratz, S.W., Mykkanen, H. and El-ezami, H.S. (2010) Probiotics and gut health:
a special focus on liver diseases. World J. Gastroenterol., 16, 403–410.
88 Probiotics and Their Therapeutic Role
Guo, Z., Liu, X.M., Zhang, Q.X., Shen Z., Tian, F.W., Zhang, H., Sun, Z.H., Zhang
H.P. and Chen, W. (2011) Inluence of consumption of probiotics on the
plasma lipid proile: a meta-analysis of randomized controlled trials.
Nutrition Metab. Cardiovasc. Dis., 21, 844–850.
Hamilton-Miller, J.M.T. (2003) The role of probiotics in the treatment and
prevention of Helicobacter pylori infection. Int. J. Antimic. Agents, 22,
360–366.
Hatakka, K., Holma, R., El-Nezami, H., Suomalainen, T., Kuisma, M., Saxelin,
M., Poussa, T., Mykkänen, H. and Korpela, R. (2008a) The inluence
of Lactobacillus rhamnosus LC705 together with Propionibacterium
freudenreichii ssp. shermanii JS on potentially carcinogenic bacterial
activity in human colon. Int. J. Food Microbiol., 128, 406–410.
Hatakka, K., Mutanen, M., Holma, R., Saxelin, M. and Korpela, R. (2008b)
Lactobacillus rhamnosus LC705 together with Propionibacterium
freudenreichii ssp shermanii JS administered in capsules is ineffective
in lowering serum lipids. J. Am. Coll. Nutrition, 27, 441–447.
Hemsworth, J., Hekmat, S. and Reid, G. (2011) The development of
micronutrient supplemented probiotic yogurt for people living with
HIV: laboratory testing and sensory evaluation. Innovative Food Sci.
Emerg. Technol. 12, 79–84.
Hoesl, C.E. and Altwein, J.E. (2005) The probiotic approach: an alternative
treatment option in urology. Eur. Urol., 47, 288–296.
Isolauri, E. (2004) The role of probiotics in paediatrics. Curr. Paediatrics, 14,
104–109.
Jeun, J., Kim, S., Cho, S., Jun, H., Park, H., Seo J., Chung, M. and Lee, S. (2010)
Hypocholesterolemic effects of Lactobacillus plantarum KCT3928 by
increased bile acid excretion in C57BL/6 mice. Nutrition, 26, 321–
330.
Jonkers, D. and Stockbrügger, R. (2007). Review article: probiotics in
gastrointestinal and liver diseases. Alimentary Pharmacol. Therap., 26,
133–148.
Kirpich, I.A., Solovieva, N.V., Leikhter, S.N., Shidakova, N.A., Lebedeva, O.V.,
Sidorov, P.I., Bazhukova, T.A., Soloviev, A.G., Barve, S.S., McClain, C.J.
and Cave, M. (2008) Probiotics restore bowel lora and improve liver
enzymes in human alcohol-induced liver injury: a pilot study. Alcohol,
42, 675–682.
Kumar, M., Verma, V., Nagpal, R., Kumar, A., Gautam, S.K., Behare, P.V., Grover,
C.R. and Aggarwal, P.K. (2011) Effect of probiotic fermented milk and
chlorophyllin on gene expressions and genotoxicity during AFB1-
induced hepatocellular carcinoma. Gene, 490, 54–59.
References 89
Lam, E.K.Y, Yu, L., Wong, H.P.S., Wu, W.K.K, Shin, V.Y., Tai, E.K.K, So, W.H.L.,
Woo, P. C.Y, and Cho, P.C.Y. (2007) Probiotic Lactobacillus rhamnosus
GG enhances gastric ulcer healing in rats. Europ. J. Pharmacol., 565,
171–179.
LeBlanc, A.M., Matar, C., Thériault, C. and Perdigón, G. (2005) Effects of
milk fermented by Lactobacillus helveticus R389 on immune cells
associated to mammary glands in normal and a breast cancer model.
Immunobiology, 210, 349–358.
Lewis, S.J. and Burmeister, S.A. (2005) A double-blind placebo-controlled
study of the effects of Lactobacillus acidophilus on plasma lipids. Eur. J.
Clin. Nutrition, 59, 776–780.
Linsalata, M. and Russo, F. (2008) Nutritional factors and polyamine
metabolism in colorectal cancer. Nutrition, 24, 382–389.
Lorca, G.L., Wadstrom, T., Valdez, G.F. and Ljungh, A. (2001) Lactobacillus
acidophilus autolysins inhibit Helicobacter pylori in vitro. Curr.
Microbiol., 42, 39–44.
Ly, N.P, Litonjua, A., Gold, D.R. and Celed. J.C. (2011) Gut microbiota,
probiotics, and vitamin D: interrelated exposures inluencing allergy,
asthma, and obesity? J. Allergy Clin. Immunol., 127, 1087–1094.
Lye, H., Rusul, G. and Liong M. (2010) Removal of cholesterol by lactobacilli
via incorporation and conversion to coprostanol. J. Dairy Science, 93,
1383–1392.
Marco, M.L., Pavan, S. and Kleerebezem, M. (2006) Towards understanding
molecular modes of probiotic action. Curr. Opin. Biotechnol., 17, 204–
210.
Marteau, E. (2001) Prebiotics and probiotics for gastrointestinal health. Clin.
Nutrit., 20, 41–45.
Marteau, P., Seksik, P. and Jian, R. (2002) Probiotics and health: new facts
and ideas. Curr. Opin. Biotechnol., 13, 486–489.
Marteau, P. and Shanahan, F. (2003) Basic aspects and pharmacology of
probiotics: an overview of pharmacokinetics, mechanisms of action
and side-effects Best Pract. Res. Clin. Gastroenterol., 17, 725–740.
Marteau, P., Sokol, H., Dray, X. and Seksik, P. (2009) Bacteriotherapy for
inlammatory bowel disease: therapeutic tool and/or pharmacological
vectors? Gastroentérologie Clinique Biologique, 33, Suppl. 3, S228–
S234.
McFarland, L.V. and Elmer, G.W. (1997) Pharmaceutical probiotics for the
treatment of anaerobic and other infections. Anaerobe, 3, 73–78.
90 Probiotics and Their Therapeutic Role
Storm, D.W., Koff, S.A., X Horvath, D.J., Li, B. and Justice, S.S. (2011) In vitro
analysis of the bactericidal activity of Escherichia coli Nissle 1917
against pediatric uropathogens. J. Urol., 186, 1678–1683.
Sullivan, A. and Nord, C.E. (2002) The place of probiotics in human intestinal
infections. Int. J. Antimicrob. Agents, 20, 313–319.
Taylor, A. L., Dunstan, J.A. and Prescott, S.L. (2007) Probiotic supplementation
for the irst 6 months of life fails to reduce the risk of atopic dermatitis
and increases the risk of allergen sensitization in high-risk children: a
randomized controlled trial. J. Allergy Clin. Immunol., 119, 184–191.
Tuohy, K.M. Probert, H.M. Smejkal, C.W. and Gibson, G R. (2003) Using
probiotics and prebiotics to improve gut health. Drug Discovery Today,
8, 692–700.
Tuomola, E.M., Ouwehand, A.C. and Salminen, S.J. (1999) The effect of
probiotic bacteria on adhesion of pathogens to human intestinal
mucus. FEMS Immunol Med Microbiol, 26, 137–142.
Uehara, S., Monden, K., Nomoto, K., Seno, Y., Kariyama, R., and Kumona,
H. (2006) A pilot study evaluating the safety and effectiveness of
Lactobacillus vaginal suppositories in patients with recurrent urinary
tract infection. Int. J. Antimicrob. Agents, 28, S30–S34.
Vasiljevic, T. and Shah, N.P. (2008) Probiotics—from Metchnikoff to
bioactives. Int. Dairy J., 18, 714–728.
Viljanen, M., Pohjavuori, E., Haahtela, T., Korpela, R., Kuitunen, M., Sarnesto,
A. Vaarala, O. and Savilahti, E. (2005) Induction of inlammation as a
possible mechanism of probiotic effect in atopic eczema–dermatitis
syndrome. J. Allergy Clin. Immunol., 115, 1254–1259.
Van Overtvelta, L. Moussua, H., Horiota, S., Samsonb, S., Lombardia,V.,
Mascarella, L., van de Moerb, A., Bourdet-Sicard, R. and Moingeona, P.
(2010) Lactic acid bacteria as adjuvants for sublingual allergy vaccines.
Vaccine, 28, 2986–2992.
Zhang, M., Cheng J., Lu, Y., Yi, Z., Yang, P. and Wu, X. (2010) Use of pre-, pro-
and synbiotics in patients with acute pancreatitis: A meta-analysis.
World J. Gastroenterol., 16, 3970–3978.
Zhao, H.Y., Wang, H.J., Lu, Z. and Xu, S.Z. (2004) Intestinal microlora in
patients with liver cirrhosis. Chin J Dig Dis., 5, 64–67.
Wade, W.G. (2010) New aspects and new concepts of maintaining
“microbiological” health. J. Dentistry, 38, S21–S25.
Wilson, N.L., Moneyham, L.D. and Alexandrov, A.N. (2008) A systematic
review of probiotics as a potential intervention to restore gut health in
HIV infection. Mol. Immunol., 46, 172–180.
References 93
4.1 Introduction
The demand for functional foods has, over recent years, increased
markedly (Siro et al., 2008). These foods, when include probiotics
and prebiotics as biologically active components, produce metabolic
and physiological health beneits in addition to their nutritional
properties or at least enhance the normal homeostatic mechanisms
in the intestine. Considering that scientiic evidences indicate that
ingestion of some microbial strains provokes health beneits, which
are extensively discussed in previous chapters, no clear indications
exist about the effective dose for these strains; however, it has been
consensual that high numbers of viable bacteria are recommended
to assure probiotic foods eficiency. The viability and metabolic
activity of probiotic microorganisms are important factors that
should be controlled throughout processing operations, maturation
Yoghurt
Traditional
Fermented Milks
Therapeutic and
Nature Thermophilic Intestinal Flora
strains
Gelled Miscellaneous
Viscous Dietetic
Pasty, Foamy
Frozen, Dried Fermented Milks +
Mesophilic
Concentrated Plant Material
strains Slimy Fermented
Flavoured Milks
Non-slimy
Buttermilks
Lactic and
Alcoholic Kef ir and Koumiss
Figure 4.1 The family tree of fermented milks. Adapted from Kurmann
(1984) and Robinson and Tamime (1990). All fermented
milks are lactic fermentations (largest group worldwide)
with exception of shaded boxes that may include lactic-yeast
fermentations.
Strain
Survival in upper
Safety Capacity for large
intestinal tract and
considerations scale production
resistance to bile salts
Possible addition of
Growth with 1-10% Coagulation of milk
growth-promoting Oxygen tolerance for
inoculum within 24 h within 24 h
factors for lactic growth in milk
(108-109 cfu/ml) (pH < 4.6)
acid bacteria (LAB)
Maintenance of viability
(Continued)
Dairy Products
109
110
- Delivery vehicle is also essential A meta-analysis has shown that delivering Sachdeva and
for potential probiotic activity— probiotic strains via fermented milk products Nagpal, 2009
metabolic activity and probiotic may be more eficient in eradication of intestinal
properties are, in some cases, H. pylori than delivering them via capsules or
only stimulated if fermented sachets.
milk is the delivery vector.
(Continued)
Dairy Products
111
112
and lentil lour (Zare et al., 2012), lactulose (Oliveira et al., 2011),
tryptone, yeast extracts, certain amino acids, nucleotide precursors
(Gomes et al., 1998a), and a mineral source such as iron or zinc
(Seleet et al., 2011).
Other options currently pursued for improving viability include
enhancement of bacterial stress response (Corcoran et al., 2008)
and quorum-sensing interference. The release of quorum-sensing
signal molecules by probiotics may interact with human intestinal
epithelial cells and contribute to the modulation of the different
physiological functions mentioned previously in Chapter 2.
Although survival in fermented milks has been discussed herein
as a challenge for probiotic strain stability, the relevance of the
protective effect of fermented milk on passage through the GIT
(Ranadheera et al., 2010) should not be overlooked. Indeed, buffering
capacity and pH have been indicated as the major reasons for such
protection, yet how these properties regulate the metabolism and
cellular processes of the probiotics at the molecular level is still
unknown. Recent indings by Wang et al. (2012) unravel some of
these phenomena for L. casei Zhang, a strain isolated from koumiss
in Inner Mongolia, China. The authors used microarray technology
to detect the gene expression proile of the L. casei strain with
and without fermented milk (strain suspension) used as a carrier
during transit in simulated gastrointestinal juice. Interestingly, a
large proportion of genes involved in translation and cell division
were downregulated in the bacteria that were in strain suspension
during transit in simulated intestinal juice. This may hamper protein
biosynthesis and cell division and may partially explain the lower
viability of L. casei Zhang during transit in the GIT without the
fermented milk (Wang et al., 2012).
4.2.2 Cheeses
In the last decade, research efforts on the development of probiotic
cheeses containing viable cells of Lactobacillus and Biidobacterium
sp. with potential health beneits have been made to lead this type of
product into the functional foods category (Ross et al., 2002). Cheese
has been claimed as a good carrier of probiotic bacteria because it
enables their passage as viable cells throughout the GIT (Gomes
et al., 1995; Mäkeläinen et al., 2009). In addition, the organoleptic
characteristics, nutritional values, and the suitability of cheeses to
116 Food as Vehicles of Probiotics
Lactococci
Lact. lactis ssp. lactis Fresh cheese/57 days—4 or 12°C Roy et al., 1997
117
118 Food as Vehicles of Probiotics
Cheese type Probiotic strain Process remarks Ripening Viable cell counts References
parameters/ (log cfu/g)
Storage Fresh Ripened
conditions cheese cheese
Pategrás L. paracasei Probiotic strain used as adjunct; 60 days/12°C 6.0–7.8 9.0 Bergamini
cheese (semi- L. acidophillus Starter culture: Streptococcus /80 %HR 5.5–7.2 7.5–8.5 et al., 2009
hard cheese) B. lactis thermophilus 7.0–7.5 7.2–7.8
Cream cheese L. plantarum RL34 Cheese aseptically illed in plastic 90 days/4°C 8.5 7.0 Georgieva
boxes et al., 2009
Turkish Beyaz L. fermentum Mixed probiotic culture 120 days/4°C 9.4 7.9 Kiliç et al.,
cheese L. plantarum Cheeses packed in plastic bags 2009
with brine
Cremoso L. casei I90 Each probiotic strain used as 60 days/5°C – 7.4 Milesi et al.,
cheese (soft L. plantarum I91 adjunct; Starter culture: Str. 8.2 2009
cheese) L. rhamnosus I73, I77 thermophilus. 7.3–7.7
Vacuum-packed cheeses.
Pategrás L. casei I90 Each probiotic strain used as 60 days/12°C/80 – 8.7 Milesi et al.,
cheese (semi- L. plantarum I91 adjunct; Starter culture: Str. %HR 8.9 2009
hard cheese) L. rhamnosus I73, I77 thermophilus. 8.6–8.7
(Continued)
Dairy Products
123
124
Cheese type Probiotic strain Process remarks Ripening Viable cell counts References
parameters/ (log cfu/g)
Storage Fresh Ripened
conditions cheese cheese
Cheddar B. longum 1941 Mixed probiotic culture used 24 weeks/4–8°C 8.3 8.3 Ong and
cheese B. animalis subsp. as adjunct; Starter culture: 8.4 8.5 Shah, 2009
Food as Vehicles of Probiotics
(Continued)
Dairy Products
125
126
Cheese type Probiotic strain Process remarks Ripening Viable cell counts References
parameters/ (log cfu/g)
Storage Fresh Ripened
conditions cheese cheese
Synbiotic L. casei 01 Each probiotic bacteria used as 60 days/12°C 7.8–8.2 8.5–8.7 Rodrigues
curdled milk L. acidophillus L10 starter. 8.2–8.5 8.0–8.2 et al., 2011a
Food as Vehicles of Probiotics
(Continued)
129
130
4.3.1 Beverages
Alternative functional foods with probiotics and dairy products aside
such as juices are of potential interest (Rodrigues et al., 2012b).
The growing interest in the development of nondairy beverages is
associated with the potential alternative ways to deliver probiotic
strains to lactose-intolerant people with reduced cholesterol
content, which is of interest to many regular consumers. According
to the review by Espírito Santo et al. (2011), the probiotic food with
fruits as ingredients has been indicated as consumers’ predilection.
However, fermented milks are still the preferred product to develop
probiotic beverages. Calcium and vitamin-fortiied juices as
functional drinks are consumed regularly (Bhadoria and Mahapatra,
2011), and therefore, probiotic beverages based on fruit juices are
of utmost interest, as full beneits of probiotics are increasingly
recognized by the regular consumer. Apart from allergy to dairy
products, other factors such as tradition and economic reasons
limiting the consumption of dairy products in countries such as
China or Japan reinforce the need of alternative nondairy products
to deliver probiotic agents (Granato et al., 2010).
Nondairy Products 135
Other
Juice Probiotic relevant Storage Viable cell counts (log cfu/
composition strains information conditions mL)/pH Remarks References
Start End
Pomegranate L. plantarum Fermented 4°C 8.6 NA Increments of all Mousavi
L. delbrueckii through 72 h 4 weeks 8.6 NA probiotic strains et al., 2011
L. paracasei at 30 °C 8.5 NA between 24 and
Food as Vehicles of Probiotics
(Continued)
137
138
Other
Juice Probiotic relevant Storage Viable cell counts (log cfu/
composition strains information conditions mL)/pH Remarks References
Start End
Noni L. casei Fermented 4°C NA NA L. casei did not Wang et al.,
Food as Vehicles of Probiotics
(Continued)
Nondairy Products
139
140
Other
Juice Probiotic relevant Storage Viable cell counts (log cfu/
composition strains information conditions mL)/pH Remarks References
Start End
Orange L. salivarius Non- 4°C 7.9/±3.5 NA/±3.5 Strains of L. Sheenan et
Food as Vehicles of Probiotics
D7 at 30°C.
Note: NA - Not available
141
142 Food as Vehicles of Probiotics
(Continued)
146 Food as Vehicles of Probiotics
4.4 Conclusions
The demand for functional foods has been increased over the last
years. These foods, including probiotics and prebiotics as biologically
active components, produce metabolic and physiological health
beneits apart from their nutritional properties. The most common
probiotic strains found in functional foods belong to Lactobacillus
and Biidobacterium genera. Nowadays, a phletora of probiotic
food products are available in the market, some of them including
prebiotic compounds that demonstrate the interest and potential
of these products. The dairy-based products dominate the most
consumed probiotic products (mostly fermented milks and cheeses)
over the world, but beverages (with or without dairy components)
or other products, such as meat products, dry bars, and so on, have
been researched and some of them are already found in the market.
The most challenging aspects to overcome by using food as
vehicles of probiotics are indeed to assure their viability through
152 Food as Vehicles of Probiotics
References
Abraham S., Cachon R., Colas B., Feron G. and De Coninck J. (2007) Eh and
pH gradients in Camembert cheese during ripening: measurements
using microelectrodes and correlations with texture. Int. Dairy J., 17,
954–960.
Akin M. B., Akin, M. S. and Kirmaci, Z. (2007) Effects of inulin and sugar
levels on the viability of yogurt and probiotic bacteria and the physical
and sensory characteristics in probiotic ice-cream. Food Chem., 104,
93–99.
Aragon-Alegro, L. C, Alarcon Alegro, J. H., Cardarelli, H. R., Chiu, M. C. and
Isay Saad, S. M. I. (2007) Potentially probiotic and synbiotic chocolate
mousse. LWT-Food Sci. Technol., 40, 669–675.
Araújo E. A., Carvalho A. F., Leandro E. S., Furtado M. M. and Moares C. A.
(2010) Development of a symbiotic cottage cheese added with
Lactobacillus delbrueckii UFV H2b2O and inulin. J. Functional Foods, 2,
85–89.
Azarnia, S., Robert, N. and Lee, B. (2006) Biotechnological methods to
accelerate Cheddar cheese ripening. Crit. Rev. Biotechnol., 26, 121–
143.
Bergamini, C.V., Hynes, E. R., Palma, S. B., Sabbag, N. G. and Zalazar, C. A. (2009)
Proteolytic activity of three probiotic strains in semi-hard cheese as
single and mixed cultures: Lactobacillus acidophilus, Lactobacillus
paracasei and Biidobacterium lactis. Int. Dairy J., 19, 467–475.
References 153
Borges, S., Barbosa, J., Camilo, R., Carvalheira, A., Silva, J., Sousa, S., Gomes,
A.M., Pintado, M. M., Silva, J. P., Costa, P., Amaral, M. H., Teixeira, P.
and Freitas, A. C. (2012) Effects of encapsulation on the viability of
probiotic strains exposed to lethal conditions. Int. J. Food Sci. Technol.,
47(2), 416–421.
Borriello, S. P., Hammes, W. P., Holzapfel, W., Marteau, P., Schrezenmeir, J.,
Vaara, M. And Valtonen, V. (2003) Safety of probiotics that contain
Lactobacilli or Biidobacteria. Clin. Infect. Dis., 36, 775–780.
Boylston, T. D., Vinderola, C. G., Ghoddusi, H. B. and Reinheimer, J. A. (2004)
Incorporation of biidobacteria into cheese: challenges and rewards.
Int. Dairy J., 14, 375–387.
Capela, P., Hay, T. K. C. and Shah, N.P. (2006) Effect of cryoprotectants,
prebiotics and microencapsulation on survival of probiotic organisms
in yoghurt and freeze-dried yoghurt. Food Res. Int., 39, 203–211.
Champagne, C. P., Tompkins, T. A., Buckley, N. D. and Green-Johnson, J.
M. (2010) Effect of fermentation by pure and mixed cultures of
Streptococcus thermophilus and Lactobacillus helveticus on isolavone
and B-vitamin content of a fermented soy beverage. Food Microbiol.,
27, 968–972.
154 Food as Vehicles of Probiotics
Muller, J. A., Stanton, C., Sybesma, W., Fitzerald, G. F. and Ross, R. P. (2010)
Reconstitution conditions for dried probiotic powders represent a
critical step in determining cell viability. J. Appl. Microb., 108, 1369–
1379.
Muthukumarasamy, P. and Holley, R. A. (2007) Survival of Escherichia coli
O157:H7 in dry fermented sausages containing micro-encapsulated
probiotic lactic acid bacteria. Food Microb., 24, 82–88.
Muthukumarasamy, P. and Holley, R. A. (2006) Microbiological and
sensory quality of dry fermented sausages containing alginate-
microencapsulated Lactobacillus reuteri. Int. J. Food Microb., 111,
164–169.
Nazzaro, F., Fratianni, F., Sada, A. and Orlando, P. (2008) Synbiotic potential
of carrot juice supplemented with Lactobacillus spp. and inulin or
fructooligosaccharides. J. Sci. Food Agric., 88, 2271–276.
Oliveira, R. P. S., Florence, A. C. R., Perego, P., Oliveira, M. N. and Converti, A.
(2011) Use of lactulose as prebiotic and its inluence on the growth,
acidiication proile and viable counts of different probiotics in
fermented skim milk. Int. J. Food Microb., 145, 22–27.
Oliveira, R. P. S., Florence, A. C. R., Silva, R. C., Perego, P., Converti, A., Gioielli,
L .A. an Oliveira, M.N. (2009a) Effect of different prebiotics on the
fermentation kinetics, probiotic survival and fatty acids proiles in
nonfat symbiotic fermented milk. Int. J. Food Microb., 128, 467–472.
Oliveira, R. P. S., Perego, P., Converti, A. and Oliveira, M. N. (2009b) Effect
of inulin on growth and acidiication performance of different
probiotic bacteria in co-cultures and mixed culture with Streptococcus
thermophilus. J. Food Eng., 91, 133–139.
Oliveira, R. P. S., Perego, P., Converti, A. and Oliveira, M. N. (2009c) Growth
and acidiication performance of probiotics in pure culture and co-
culture with Streptococcus thermophilus: the effect of inulin. LWT-Food
Sci. Technol., 42, 1015–1021.
Ong, L. and Shah, N. P. (2009) Probiotic Cheddar cheese: Inluence of
ripening temperatures on survival of probiotic microorganisms,
cheese composition and organic acid proiles. LWT-Food Sci. Technol.,
42, 1260–1268.
Ong, L. and Shah, N. P. (2008) Inluence of Probiotic Lactobacillus acidophilus
and L. helveticus on proteolysis, organic acid proiles, and ACE-
iInhibitory activity of Cheddar cheeses ripened at 4, 8, and 12°C. J.
Food Sci., 73, M111–M120.
Ong, L., Henriksson, A. and Shah, N. P. (2007) Proteolytic pattern and organic
acid proiles of probiotic cheese as inluenced by probiotic strains of
References 163
Rößle, C., Brunton, N. Ronan, Gormley, T., Ross, P. R. and Butler, F. (2010)
Development of potentially synbiotic fresh-cut apple slices. J. Functional
Foods, 2, 245–254.
Rodrigues, D., Rocha-Santos, T. A. P., Freitas, A. C., Gomes, A. M. P., Duarte, A.
C. (2012a). Analytical strategies for characterization and validation of
functional dairy foods. Trends in Anal. Chem., 41, 27–45.
Rodrigues, D., Sousa, S., Gomes, A. M., Pintado, M. M., Silva, J. P., Costa, P.,
Amaral, M. H., Rocha-Santos, T. A. P. and Freitas, A. C. (2012b) Storage
stability of L. paracasei as free cells or encapsulated in alginate based
microcapsules in low pH fruit juices. Food Bioprocess Technol., 5,
2478–2757.
Rodrigues, D., Rocha-Santos, T. A. P., Pereira, C. I., Gomes, A. M., Malcata, F. X.
and Freitas, A.C. (2011a) The potential effect of FOS and inulin upon
probiotic bacterium performance in curdled milk matrices. LWT–Food
Sci. Technol., 44, 100–108.
Rodrigues, D., Santos, C. H., Rocha-Santos, T. A. P., Gomes, A. M., Goodfellow, B.
J. and Freitas A. C. (2011b) Metabolic proiling of potential probiotic or
synbiotic cheeses by nuclear magnetic resonance (NMR) spectroscopy.
J. Agric. Food Chem., 59, 4955–4961.
Rodrigues, D., Rocha-Santos, T. A. P., Gomes, A. M., Goodfellow, B. J. and Freitas,
A. C. (2011c) Lipolysis in probiotic and synbiotic cheese: The inluence
of probiotic bacteria, prebiotic compounds and ripening time on free
fatty acid proiles. Food Chem., 131, 1414–1421.
Rodrigues, D., Sousa, S., Rocha-Santos, T., Silva, J., Sousa Lobo, J., Costa, P.,
Amaral, M. H., Gomes, A., Pintado, M. M., Malcata, F. X. and Freitas, A.
C. (2011d) Inluence of L-cysteine, oxygen and relative humidity upon
survival throughout storage of probiotic bacteria in whey protein-
based microcapsules. Int. Dairy J., 21, 869–876.
Rodríguez-Alcalá, L. M., Braga, T., Malcata, F. X., Gomes, A. and Fontecha,
J. (2011) Quantitative and qualitative determination of CLA
produced by Biidobacterium and lactic acid bacteria by combining
spectrophotometric and Ag+-HPLC techniques. Food Chem., 125(4),
1373–1378.
Roland, N., Bougle, D., Lebeurrier, F., Arhan, P., Maubois, J. L. (1998)
Propionibacterium freudenreichii stimulates the growth of
Biidobacterium biidum in vitro and increases fecal biidobacteria in
healthy human volunteers. Int. Dairy J., 8, 587–588.
Ross, R. P., Fitzgerald, G. F., Collins, K. and Stanton, C. (2002) Cheese delivering
biocultures: probiotic cheese. Austr. J. Dairy Technol., 57, 71–78.
166 Food as Vehicles of Probiotics
Suomalainen, T., Lagström, H., Mättö, J., Saarela, M., Laitinen, L, Ouwehands,
A.C. and Salminen, A. (2006) Inluence of whey-based fruit juice
containing Lactobacillus rhamnosus on intestinal well-being and
humoral immune response in healthy adults. LWT-Food Sci. Technol.,
39(7), 788–795.
Tamime, A.Y. (2002) Fermented milks: a historical food with modern
applications: a review. Eur. J. Clinical Nutr., 56(4), S2–S15.
Tamime, A. Y., Wszolek, M., Bo¡zani, C, R. and Özer, B. (2011) Popular ovine
and caprine fermented milks. Small Rumin. Res., 101, 2–16.
Tang, A. L., Shah, N. P., Wilcox, G., Walker, K.Z. and Stojanovska, L. (2007)
Fermentation of calcium-fortiied soymilk with Lactobacillus: Effects
on calcium solubility, isolavone conversion, and production of organic
acids. J. Food Sci., 72, M431–M436.
Tharmaraj, N. and Shah, P. N. (2004) Survival of Lactobacillus acidophilus,
Lactobacillus paracasei ssp. paracasei, Lactobacillus rhamnosus,
Biidobacterium animalis and Propionibacterium in cheese-based dips
and the suitability of dips as effective carriers of probiotic bactéria. Int.
Dairy J., 14, 1055–1066.
Tuorila, H. and Cardello, A. V. (2002) Consumer responses to an off-lavor
in juice in the presence of speciic healh claims. Food Qual. Preference,
13, 561–569.
Van Baarlen, P., Troost, F., Van Hemert, S., Van der Meer, C., De Vos, W. M.,
Hooiveld, G. J. E. J., Brummer, R. J. M and Kleerebezem, M. (2009)
Differential NF-κB pathways induction by Lactobacillus plantarum in
the duodenum of healthy humans correlating with immune tolerance.
Proc. Natl. Acad. Sci. USA, 106, 2371–2376.
Varga, L., Szigeti, J. and Gyenis, B. (2006) Inluence of chicory inulin on the
survival of microbiota of a probiotic fermented milk during refrigerated
storage. Ann. Microbiol., 56(2), 139–141.
Vinderola, G., Prosello, W., Molinari, F., Ghiberto, D. and Reinheimer, J. (2009)
Growth of Lactobacillus paracasei A13 in Argentinian probiotic cheese
and its impact on the characteristics of the product. Int. J. Food Microb.,
135, 171–174.
Vinderola, C. G., Prosello, W., Ghilberto, D. and Reinheimer, J. A. (2000)
Viability of probiotic Biidobacterium, Lactobacillus acidophilus and
Lactobacillus casei and nonprobiotic microlora in Argentinean Fresco
cheese. J. Dairy Sci., 83, 1905–1911.
Vorobjeva, L. I., Khodjaev, E. Y. and Vorobjeva, N. V. (2008) Propionic acid
bacteria as probiotics. Microbial Ecol. Health Dis., 20, 190–112.
References 169
Wang, J., Zhong, Z., Zhang, W., Bao, Q., Wei, A., Meng, H. and Zhang, H. (2012)
Comparative analysis of the gene expression proile of probiotic
Lactobacillus casei Zhang with and without fermented milk as a vehicle
during transit in a simulated gastrointestinal tract. Res. Microbiol., 163,
357-365.
Wang, C., Ng, C., Su, H., Tzeng, W. and Shyu, Y. (2009) Probiotic potential of
noni juice fermented with lactic acid bacteria and biidobacteria. Int. J.
Food Sci. Nut., 60, 98–109.
Warmiska-Radyko I., Laniewska-Moroz L. and Babuchowski A. (2002)
Possibilities for stimulation of Biidobacterium growth by
propionibacteria. Lait, 82, 113–121.
Weinbreck, F., Bodnár, I. and Marco, M. L. (2010) Can encapsulation lengthen
the shelf-life of probiotic bacteria in dry products? Int. J. Food Microb.,
136, 364–367.
Xanthopoulos, V., Ipsilandis, C. G. and Tzanetakis, N. (2012) Use of a selected
multi-strain potential probiotic culture for the manufacture of set-type
yogurt from caprine milk. Small Ruminant Res., 106(2–3), 145–153.
Yilmaztekin, M., Özer, B.H. and Atasoy, F. (2004) Survival of Lactobacillus
acidophilus LA-5 and Biidobacterium biidum BB-02 in white-brined
cheese. Int. J. Food Sci. Nutr., 55, 53–60.
Yoon, K. Y., Woodams, E. E. and Hang, Y. D. (2006) Production of probiotic
cabbage juice by lactic acid bacteria. Biores. Technol., 97, 1427–1430.
Zare, F., Champagne, C. P., Simpson, B. K., Orsat, V. and Boye, J. I. (2012)
Effect of the addition of pulse ingredients to milk on acid production
by probiotic and yoghurt starter cultures. LWT-Food Sci.Technol., 45,
155–160.
This page intentionally left blank
Chapter 5
Viseu, Portugal
cCESAM & Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
dCBQF, Biotechnology School of Portuguese Catholic University,
5.1 Introduction
The preparation of microparticles is in great development in several
industrial areas, especially in the pharmaceutical and food industries.
In recent years, the food industry has been incorporating probiotics
in microparticles in order to develop new and more diversiied
functional foods that could cause an enhancement in health state of
consumer. Probiotics have also been encapsulated for incorporation
on pharmaceutical dosage forms.
a copy (Green 1955). In the food industry, the irst researches took
place from the late 1950s (Gouin 2004). Lactic acid bacteria (LAB)
was irst immobilized on Berl saddles in 1975 and later Lactobacillus
lactis was encapsulated in calcium alginate beads (Gibbs et al. 1999,
Desai and Park 2005). In the pharmaceutical industry, the early
research also occurred in the same period. In this ield, there was an
important contribution because microencapsulation permitted the
development of controlled-release formulas, that is, those with the
capacity to release active agents in the organs where they shall act
only or where they will be absorbed (Suave et al. 2006).
The microparticles continue to be employed in different sectors
of industry in the present day, performing a variety of functions. The
protection afforded by the wall of polymeric microparticles increases
the lifespan of a volatile compound, extending the shelf time of many
food products and cosmetics (Leimann 2008). In the pharmaceutical
industry, the applications are very varied: concealment of lavors
or odors, converting liquids into solids, protection in relation to
atmospheric agents (light, heat and humidity, and/or oxidation),
reduction or elimination of gastric irritation or secondary effects
caused by certain drugs, reducing volatility, production of modiied-
release dosage forms, and so on (Kas and Oner 2000, Yoshizawa
2004, Burgess and Hickey 2006, Jyothi et al. 2010).
The use of microparticles in the food industry is also of great
importance, because it increases the stability of oils, lavorings,
vitamins, and so on (Leimann 2008). In this area, they have the ability
to reduce the reactivity of the core material with the environment;
decrease the rate of evaporation or the transfer of the core material
toward the outer space; facilitate the encapsulated material handling;
promote the controlled release of some substances; mask unpleasant
odor and lavor; and promote homogeneous dilution of the material
encapsulated in a food matrix (Favaro-Trindade et al. 2008).
Probiotic encapsulation was developed from the technology
involved in cell culture (immobilized cell culture technology, ICT)
(Heidebach et al. 2012). Probiotics have two limitations regarding
encapsulation, namely the size of the microorganisms (with
diameters typically between 1 and 5 μm), which prevents the use
of nanotechnology, and the fact that the cells must be kept viable
(Burgain et al. 2011). This aspect has been crucial to the choice
of encapsulation technology (Champagne and Fustier 2007). The
main purpose of the encapsulation of probiotics is to increase
Microencapsulation 175
viability of these products during its shelf life, protect cells against
an unfavorable environment that occurs in food matrices and in the
gastrointestinal tract, and to enable their release in a viable and
metabolically active state in the gut (Sun and Grifiths 2000, Picot
and Lacroix 2004, Burgain et al. 2011, Nazzaro et al. 2012).
Different approaches have been proposed to increase the
resistance of these sensitive microorganisms against adverse
conditions (Anal and Singh 2007) such as an appropriate selection
of acid and bile-resistant strains, the use of oxygen-impermeable
containers, a two-step fermentation, a stress adaptation, an
incorporation of micronutrients such as peptides and amino acids,
protectors such as sugars and oligosaccharides, and, as previously
referred, the usage of microencapsulation.
After a successful encapsulation, the inner bioactive material
should be released to the outside, where it might implement its
beneicial action. There are four typical mechanisms by which the
core material can be released from a microparticle: mechanical
rupture of the capsule wall, dissolution of the wall, melting of the wall,
and diffusion through the wall. Less common release mechanisms
include ablation (slow erosion of the shell) and biodegradation
(Franjione and Vasishtha 1995, Barbosa-Cánovas et al. 2005).
5.2 Microencapsulation
Several procedures from many ields of application with different
goals led to a great number of encapsulation methods (Dziezak 1988,
Arshady 1989, Dulieu et al. 1999, Gibbs et al. 1999, Heinzen et al.
2002, Gouin 2004, Anal and Singh 2007). The terminology employed
varies between the different scientiic domains. Therefore, the same
technology may have different names in different ields.
The selection of the microencapsulation method depends on
the properties of the core and the coating materials, the size and
morphology of the particle, and the release mechanism desired. Many
of these processes have been adapted from the pharmaceutical and
chemical industries. The use of low-cost materials and manufacturing
processes is very important because food products generally have
lower proit margins than pharmaceutical products (Augustin and
Sanguansri 2008). Generally, microencapsulation can be divided
into three main stages (Poncelet 2006, Burgain et al. 2011) as shown
in Fig. 5.2.
176 Immobilization and Microencapsulation of Probiotics
Figure 5.2 General plan describing the three main phases of microparticles
production. Adapted from Poncelet (2006) and Burgain et al.
(2011).
Burgain et al. 2011, Rodrigues et al. 2011, Huq et al. 2012, Sousa et
al. 2012).
The choice of the encapsulate agent depends on a number of
factors, among them, the nonreactivity with the material to be
encapsulated, the process used for the microparticle formation, and
release mechanism are the most relevant. The ideal encapsulant must
present low viscosity at high concentrations and be easy to handle
during the process; have low hygroscopicity, for ease of handling and
preventing agglomeration; have the ability to seal and hold the active
material within the structure of the capsule; provide maximum
protection to the active material against adverse conditions,
such as light, pH, oxygen, and reactive ingredients; be soluble in
solvents commonly used; have the desired release properties of
the active material; not have an unpleasant lavor in the case of oral
consumption; and be costeffective (Shahidi and Han 1993, Barbosa-
Cánovas et al. 2005, Desai and Park 2005). Biodegradable polymers
and blends of biodegradable polymers natural or synthetic are often
used in controlled release of drugs (Cerini 2001).
5.3.1.1.1 Extrusion
The extrusion can be deined as a forced passage through an opening
(or openings) giving the substance a deined format. One of the most
important features in this procedure is the size of the hole that will
require a higher or lower division of the material to extrude. The
smaller is the inner diameter of the opening (nozzle), the smaller
are the particles.
This technique has been used for many years in the food industry.
The work that originally led to the extrusion/encapsulation process
was done by Schultz et al. of the United States Department of
Agriculture Albany Laboratory (Schultz et al. 1956). Encapsulation
of lavors via extrusion was irst patented in 1957 (Swisher 1957).
The main referred advantage of the extrusion method referred is the
increased stability of lavors or other heat-labile food components,
because in this method, it is not necessary to use high temperatures
(Dziezak 1988, Gibbs et al. 1999, Madene et al. 2006). The
extrusion method is one of the most used techniques for probiotic
encapsulation for food applications (Heidebach et al. 2012). This
category of extrusion technologies presents generally no risks but
sometimes is a very complex technique for microencapsulation (de
Vos et al. 2010).
The probiotic encapsulation by extrusion thus consists in forcing
the liquid dispersion containing the cells through a nozzle using
high pressure at a low temperature. If the extrusion occurs slowly,
droplets that will be formed will fall when the action of gravity
exceeds the surface tension. If the extrusion occurs faster, a jet of
liquid will be formed. If the extrusion (droplet formation) occurs
in a controlled mode (as opposed to atomization—spraying), the
technique is known as prilling (Heinzen 2002, Kailasapathy 2002,
Burgain et al. 2011). Prilling can be a very useful technology if the
solidiication of the exterior material could occur in an instantaneous
way (Meiners 2009). If, as already mentioned, the extrusion is done
only by the action of gravity, the particles formed are large and it is
therefore necessary to reduce the size of the droplets using auxiliary
techniques.
The preparation of microparticles using the extrusion process is
well documented (Gibbs et al. 1999, Krasaekoopt et al. 2003, Gouin
2004, Madene et al. 2006, de Vos et al. 2010, Kuang et al. 2010, Rokka
and Rantamäki 2010, Burgain et al. 2011, Matalanis et al. 2011,
180 Immobilization and Microencapsulation of Probiotics
with the axis of the needle, has been counter-sunk externally. The
counter sunk leads to the aerodynamical effect so that the jet has a
smaller diameter when passing the oriice than before at the needle
(Fig. 5.5). The size of the drops is determined by the product low
rate and the pressure inside the chamber. The product low rate is
controlled by a syringe pump, which is connected to the product
nozzle. With this type of equipment, it is possible to obtain very
small and homogeneous particles (up to about 10 μm) with a slight
risk of clogging (Nisco J30 2009–2011). This unique technology has
the advantage of reaching smaller particles than the needle diameter,
depending on the physical properties of the product (Fig. 5.6).
The parameters that affect the size of particles are the speed
of rotation, the geometry of the disk, and the liquid jet low rate
(Meiners 2009). This method has the advantage that the liquid low
rate can be high (Poncelet 2006). In addition to the method of jet
cutting, this process is the most promising for the food industry
because the preparation conditions are mild, there is no problem of
plugging, the process is continuous, and power costs are generally
low (Teunou and Poncelet 2005).
The main objectives in the disk design are to ensure that the
liquid reaches the speed of disk and to obtain a uniform droplet size
distribution in atomized liquid. Disk diameters vary from 5 cm in
Methods of Microencapsulation 187
5.3.1.1.2 Spraying
Spraying (also called atomization) is one of the most used methods
in the food industry, because in addition to being economical and
lexible produces good quality products (Dziezak 1988, Kailasapathy
2002, Gharsallaoui et al. 2007, Augustin and Sanguansri 2008, de
Methods of Microencapsulation 189
Vos et al. 2010, Zuidam and Shimoni 2010, Burgain et al. 2011,
Peighambardoust et al. 2011). Atomization followed by hot air
drying (spray drying encapsulation) has been used in the food
industry since the late 1950s (Gouin 2004, Desai and Park 2005).
The spray drying of microorganisms dates back to 1914 to the study
of Rogers on dried lactic acid cultures (Peighambardoust et al. 2011).
In 1932, the irst spray-dried lavor powders, in which the lavors
were encapsulated in a thin arabic gum ilm, were produced by an
English company A. Boake, Roberts & Co., Ltd (Barbosa-Cánovas et
al. 2005).
In the spraying process, bacteria are dispersed in a liquid phase
and this system is atomized in a stream of air (Sousa e Silva and M.
Ferreira 1998, Gharsallaoui et al. 2007, de Vos et al. 2010). In the case
of probiotics, dispersion must be made mandatory in an aqueous
phase.
A dispersion or emulsion containing the living cells of the probiotic
in a suitable liquid carrier is initially prepared, usually polymeric. This
carrier should have good emulsifying properties, have low viscosity
at high solids content, and exhibit low hygroscopicity (Barbosa-
Cánovas et al. 2005). This process is characterized by spraying the
liquid dispersion inside a chamber subjected to a controlled stream
of warm air (spray drying) or cold air (spray freeze-drying). This
dispersion is atomized into millions of small individual droplets by
a fast rotating device or a nozzle (Fig. 5.11). Through this process,
the contact surface area of the product sprayed is greatly increased,
giving rise to microparticles. A rapid vaporization of solvent takes
place from the microparticles lying inside the chamber in contact
with the stream of air. After evaporation of the solvent, the dried
microparticles are retrieved.
Atomization can also occur at low temperatures (spray-
congealing or sprayfreezing, (Dziezak 1988, Gouin 2004, Rokka and
Rantamäki 2010, Nazzaro et al. 2012). In this case, the bacterial cells
are in a solution that is atomized in a cold gas phase, which leads to
dispersion of frozen droplets. These droplets can then be dried by
freeze-drying (Wang et al. 2006, Augustin and Hemar 2009, de Vos et
al. 2010). This technique involves the consumption of large amounts
of energy, high processing times, and higher price than that of the
spray-drying technique, especially on a large scale (Gölker 1993,
Johnson and Etzel 1995, Knorr 1998, Mattila-Sandholm et al. 2002,
Burgain et al. 2011). Despite of this, many LAB cannot tolerate the
190 Immobilization and Microencapsulation of Probiotics
5.3.1.1.3 Emulsification
The microparticles can also be produced by emulsiication. An
emulsion is a mixture of two immiscible liquids in which one of
them is in small droplets (the dispersed phase, which may contain
probiotics) within another liquid (the dispersing phase) to form a
stable mixture. Emulsions are thermodynamically unstable and
therefore do not form spontaneously, and it is necessary to provide
energy to obtain them through agitation. Surfactant agents are
mandatory substances added to emulsions to increase its stability.
The simplest method to produce emulsions consists in a reactor
with a turbine for agitation (Fig. 5.12). Emulsions can also be obtained
using continuous systems with static mixers. These systems consist
of tubes wherein elements are inserted in such a way as to cause the
ine division of the liquids. These systems allow the production of
emulsions in a fraction of seconds at a high liquid low rate (Poncelet
2006).
them, but not expel them from such air current (Dziezak 1988,
Dewettinck and Huyghebaert 1999). In conventional air suspension
coating, the basic concept of luidization depends on balancing the
gravity force experienced by the particle by an upward movement of
air low, which ensures the full luidization of those particles (Gouin
2004). In this type of encapsulation, although the core particles
are suspended in the air, the coating material is atomized into the
camera, shocking and depositing on the particle’s nucleus. As the
spraying nozzle is immersed in the airlow and sprays the coating
material concurrently into the solid particles, these droplets travel
a short distance before contacting it, resulting in a more uniform
ilm (Kinam and Yoon 2006). When the particles strike the top of
the camera, their speed becomes slow and by the action of gravity
are thrown by a descent column of air that throws them again in the
luidized bed in which they are coated once more and the cycle begins
again (Fig. 5.13). The application of a coating to a solid particle is a
very complex phenomenon. The uniform coating layer is not formed
on a single passage of the randomly oriented microparticles through
the atomization zone, but on their successive passages through the
zone, ensuring the integrity and uniformity of the coating layer.
(Dziezak 1988, Vidhyalakshmi et al. 2009).
Tangential Spray
Figure 5.14 Air suspension particle coating. Adapted from Werner et al.
(2007).
5.3.2.1 Solidification
This method consists in the dispersion of the active substance, in
this case the living bacteria, in the molten excipient, followed by
emulsiication of this phase with a heated immiscible solvent at the
same temperature. As the formation of this phase requires heating,
198 Immobilization and Microencapsulation of Probiotics
5.3.2.2 Coacervation
The earliest and one of the most widely used microencapsulation
technique involves the separation of phases by coacervation (Kinam
and Yoon 2006). This original microencapsulation technique,
described by Green (Green 1953), for the production of carbonless
copying paper was based on coacervation. It was Tiebackx (Tiebackx
1911) who irst reported the phenomenon of coacervation
without ever mentioning the word (de Kruif et al. 2004). The term
coacervation was only introduced in chemistry by Bungenberg
de Jong e Kruyt in 1929 (Bungenberg de Jong and Kruyt 1929) to
describe the phenomenon of macromolecular aggregation from a
liquid system, forming a colloidal system in which two liquid phases
separate: one rich in polymers (coacervate) and another low in
polymers (supernatant) (Dervichian 1954, Phares and Sperandio
1964, Burgess and Carless 1984, Nixon 1985, Arshady 1990, Sousa e
Silva and M. Ferreira 1998). The word derives from Latin “acervus,”
which means aggregation, and the preix “co,” which means together.
So, this word “coacervation” signiies the union of the colloidal
particles. The physical and chemical basis behind the coacervation
process is now well developed and understood (Bungenberg de Jong
and Kruyt 1929, Dervichian 1954, Bakan Joseph 1971, Burgess 1990,
Schmitt et al. 1998).
The coacervation can be deined to be simple or complex
depending on whether it has one or more polymers involved (Nixon
1985, Schmitt et al. 1998, Gibbs et al. 1999, Burgess and Hickey
2006, Kinam and Yoon 2006, Madene et al. 2006, Jyothi et al. 2010,
Zuidam and Shimoni 2010). The simple coacervation occurs when
Methods of Microencapsulation 199
2006, Kinam and Yoon 2006, Madene et al. 2006). Heating and add-
ing counter polyions or other cross-linking reagents are alterna-
tive cross-linking methods (Kinam and Yoon 2006, Augustin and
Sanguansri 2008). Enzymatic crosslinkers are more acceptable in
the food industry (Gouin 2004, Augustin and Hemar 2009). Both
chemical cross-linking agents and the application of heat may be
harmful to the encapsulated materials, such as live cells like probi-
otic bacteria.
Complex coacervation is mainly used for microencapsulation
of oils, but it can also be used for nutrients, vitamins, enzymes, and
probiotics (Gouin 2004, Oliveira et al. 2007, Rokka and Rantamäki
2010).
5.3.2.3 Gelation
Gelation is one of the most used entrapment techniques to obtain
microparticles. The contact of droplets of a gel-forming solution in a
gelation bath leads to the formation of these particles. The gelation
may be due to ionic bonding between polymer chains or by cooling
(Poncelet 2006).
The use of natural polymers in formulating dosage forms
containing food and drugs has received much attention because
of its excellent biocompatibility and biodegrability. Among them,
the alginic acid is a very promising natural polymer and has been
widely explored (Gouin 2004, Anal and Singh 2007, Rokka and
Rantamäki 2010, Ain Riaz and Masud 2011, Huq et al. 2012). It is
a linear heteropolysaccharide consisting of β-D-mannuronic acid
(M) and α-L-guluronic acid (G) (Anal and Singh 2007, de Vos et al.
2010, Draget and Taylor 2011), composed of blocks of homopolymer
MM or GG extracted from several species of Brown algae (Aslani and
Kennedy 1996, Tonnesen and Karlsen 2002). Figure 5.16 shows the
structure of sodium alginate that is the sodium salt of the alginic
acid. Depending on the origin, composition and sequence in M and G
vary considerably, affecting their functional properties as supporting
material (Tonnesen and Karlsen 2002, de Vos et al. 2010, Draget and
Taylor 2011, Teoh et al. 2011).
The alginic acid (or sodium alginate) can react with bivalent
cations or with polyvalent cation, as for example calcium, forming
cross-links leading to the formation of a gel (Morris et al. 1978). Gel
formation using calcium cation occurs mainly with GG and therefore
blocks (Fig. 5.17) forming a structure called “egg box” (Grant et al.
202 Immobilization and Microencapsulation of Probiotics
Figure 5.17 Formation of calcium alginate gel: (a) alginic acid molecules
dispersed; (b) link between the strings through the groups
of guluronic acid and calcium ions; (c) formation of the gel
network.
have been developed (Ma et al. 2012). Some other polymer systems
that can be used for ionic gelation are chitosan/triphosphate, pectin/
calcium, gelan gum/calcium, and carboxymethylcellulose/aluminum
(Kinam and Yoon 2006).
Gelation may also be obtained by spraying a thermogel (spray
chilling) or, alternatively by the extrusion method, through
emulsiication followed by cooling or pH change (Poncelet 2006,
Nag et al. 2011).
The emulsiication technique can also be used for the preparation
of calcium alginate microparticles. In this technique, an aqueous
solution of sodium alginate is dispersed in an immiscible liquid
phase and the gel droplets formed react with calcium ions, which are
added to the external phase, forming microspheres. It is possible to
get a high yield of encapsulation of hydrophilic drugs and particles
with size less than 150 μm (Wan et al. 1992). Other gelifying agents
can be used. Heidebach et al. (2009b) obtained microparticles
by transglutaminase-catalyzed gelation of casein suspensions
containing probiotic cells, namely commercial strains of L. paracasei
ssp. paracasei F19 and B. lactis Bb12. L. casei cells were successfully
entrapped into a gel matrix by a water-in-oil emulsion in which the
matrix was formed by sodium caseinate and gellan gum mixture
that was gelled by gradually decreasing pH with glucono-δ-lactone
(Nag et al. 2011). Probiotic cells L. paracasei ssp. paracasei F19 and
B. lactis Bb12 were microencapsulated in milk protein matrices by
means of an enzymatic induced gelation with rennet (Heidebach et
al. 2009a).
It is also possible to obtain microparticles of calcium alginate
by emulsiication-internal gelation (Fig. 5.18). Calcium is dispersed
in the form of an insoluble salt (e.g., calcium carbonate or calcium
citrate) in sodium alginate solution. This mixture is emulsiied in
an oily phase in order to obtain a W/O emulsion and the calcium
present in the internal phase is released by acidiication of the
external oily phase causing the gelling of alginic acid (Poncelet et al.
1992, Poncelet et al. 1995, Poncelet et al. 1999).
5.3.2.5 Polymerization
The polymerization can be in situ or interfacial, that is, it occurs in
the continuous or in the interphase of dispersed system.
In the in situ polymerization, no reactive agents are added to
the core material, and the polymerization occurs exclusively in
the continuous phase and on the side of continuous phase, on the
interface with the core dispersed (Jyothi et al. 2010). Initially, a
prepolymer with low molecular weight will be formed, which with
the passage of time will grow in size, and will deposit on the surface
of the dispersed core material generating a solid coating (Jyothi et al.
2010). The particles obtained are very small, but, typically, have high
concentrations of associated monomer. Emulsion polymerization
in external organic phase is not so used because it requires large
amounts of organic solvents and surfactants and also due to the toxic
nature of the monomers used. It is a very fast and easily scale-up
technique (Silva et al. 2003). The particles obtained have a uniform
coating with a size between 0.2 μm and 75 μm (Vidhyalakshmi et al.
2009).
In the interfacial polymerization technique, the coating can
be formed on the surface of the particle by the polymerization of
reactive monomers (Kinam and Yoon 2006, Vidhyalakshmi et al.
2009, Jyothi et al. 2010). It is characterized by the polymerization
of a monomer in two immiscible phases (Nixon 1985, Burgain et
al. 2011). If the internal phase is a liquid, it is possible to disperse
or solubilize the monomer in this phase and emulsify the mixture
206 Immobilization and Microencapsulation of Probiotics
References
Aaftabrouchad, C. and E. Doelker (1992). “Preparation methods for
biodegradable microparticles loaded with water-soluble drugs.” STP
Pharma Sciences 2: 365–380.
Acosta, E. (2009). “Bioavailability of nanoparticles in nutrient and
nutraceutical delivery.” Current Opinion in Colloid & Interface Science
14(1): 3–15.
Alli, S. M. (2011). “Preparation and characterization of a coacervate
extended-release microparticulate delivery system for Lactobacillus
rhamnosus.” International Journal of Nanomedicine 6: 1699–1707.
Amsden, B. G. and M. F. A. Goosen (1997). “An examination of factors
affecting the size, distribution and release characteristics of polymer
microbeads made using electrostatics.” Journal of Controlled Release
43(2–3): 183–196.
Anal, A. K. and H. Singh (2007). “Recent advances in microencapsulation of
probiotics for industrial applications and targeted delivery.” Trends in
Food Science & Technology 18(5): 240–251.
Ananta, E., M. Volkert and D. Knorr (2005). “Cellular injuries and storage
stability of spray-dried Lactobacillus rhamnosus GG.” International
Dairy Journal 15(4): 399–409.
Anantachoke, N., M. Makha, C. L. Raston, V. Reutrakul, N. C. Smith and M.
Saunders (2006). “Fine tuning the production of nanosized beta-
carotene particles using spinning disk processing.” Journal of the
American Chemical Society 128(42): 13847–13853.
Annan, N. T., A. Borza, D. L. Moreau, P. M. Allan-Wojtas and L. T. Hansen
(2007). “Effect of process variables on particle size and viability of
Biidobacterium lactis Bb-12 in genipin-gelatin microspheres.” Journal
of Microencapsulation 24(2): 152–162.
Arshady, R. (1989). “Microspheres and microcapsules: a survey of
manufacturing techniques. Part 1: Suspension cross-linking.” Polymer
Engineering and Science 29(24): 1746–1758.
Arshady, R. (1990). “Microspheres and microcapsules, a survey of
manufacturing techniques Part II: Coacervation.” Polymer Engineering
and Science 30(15): 905–914.
212 Immobilization and Microencapsulation of Probiotics
6.1 Introduction
The use of health-beneicial microorganisms was irst introduced
in the beginning of the 20th century. As reported in the previous
chapters, probiotics represent alternative therapeutic agents. In
order to improve the clinical eficacy of probiotics, optimization will
still be achieved by the application of new approaches (Sleator and
Hill 2008). Several European Union projects have shown that with
coordinated efforts toward a scientiic approach to the selection
and application of probiotics, effective functional products can be
developed with health beneits for consumers (Saarela et al. 2000).
This chapter is concerned with formulated preparations to
administer probiotics (medicines or dietary/food supplement).
6.2.1 Powders
Oral powders are dosage forms consisting of solid, loose, dry,
inely divided particles that are intended for internal use (Eur.
Pharmacopeia 2008; USP31 2008). They may contain bioactive
compounds and/or other excipients such as coloring, lavoring, and
sweetening agents. In general, oral powders are limited to relatively
nonpotent drugs or dietary/food supplements. The administration
of oral powders could overcome the dificulty in swallowing tablets
experienced by children or even some adults. In the production of
oral powders, measures are taken to ensure an adequate particle
size, which is generally done by milling and/or sieving.
Powders containing probiotics are frequently used to disperse
in water (extemporaneous oral suspensions). Some examples of
marketed powders for oral suspension containing probiotics are
presented in Table 6.2.
234 Development of Probiotic Dosage Forms
6.2.2 Capsules
Capsules are solid dosage forms with hard or soft container or shell
made from gelatin or, less often, from other suitable material (Eur.
Pharmacopeia 2008; USP31 2008). There are two types of capsules,
hard and soft (one-piece). Hard capsules are generally preferred in
order to administer probiotics.
The hard capsules, which consist of two cylindrical pieces (body
and cap) closed at one end, are typically used to contain powders.
These capsules are produced by a process based in a patent dated
from 1846 (Jones 2004). Empty standard size hard capsules are
supplied in bulk containers.
The illing of hard capsules may employ machines that form
powder plugs and eject them into the capsule bodies after having
separated the cap of the shell.
Hard capsule sizes are standard, generally ranging from n° 5
(0.13 ml), the smallest, to n° 000 (1.37 ml), although for human
administration the largest size used is n° 00 (0.95 or 0.93 ml
depending on the manufacturing). Gelatin capsules are readily
soluble in water at 37°C.
Probiotics in the form of powders are also used to ill capsules.
The powder formulations often require some type of excipients such
as illers, glidants, or even disintegrants. It is also possible to prepare
Manufacturing of Dosage Forms Containing Probiotics 235
6.2.3 Tablets
Tablets are solid dosage forms usually obtained by compressing
uniform volumes of particles (powders or particles aggregates)
(Eur. Pharmacopeia 2008). Tablets are the most widely used
pharmaceutical dosage form. The irst tablets comparable to those
used today have their roots in the invention of William Brockedon, in
the 19th century. In 1843, Brockedon obtained a patent for “Shaping
Pills, Lozenges and Black Lead by Pressure in Dies.” It is believed
that John Wyeth and his brother Frank were the irst to use the term
“compressed tablet” and register it in 1877 to protect and restrict its
use (Troy 2005).
Tablets are prepared by the application of a high pressure to
uniform volumes of powders (direct compression) or granules
(obtained by wet or dry granulation) using tablet presses (Fig. 6.2).
Tablets may be coated in order to facilitate its administration, to
protect its bioactive content, or to modify its release.
systems. HPMC has the ability to form a gelling barrier limiting the
release of viable probiotics.
In the studies carried out by Klayraung et al. (2009) to evaluate
the effects of formulation and processing parameters on bacterial
viability, tablets were also prepared by direct compression. In this
study, an exactly weighed amount of powder mixture containing LAB
powder and HPMC phthalate was illed into a die and tablets were
formed under a determined force ranging from 2 to 20 kN.
Tablets can be designed to modify the release and enhance the
adhesion and colonization of the probiotic microorganisms to the
epithelial mucosa of human host by using suitable kinds of tablet
excipients (Maggi et al. 2000).
Different polymers have been studied to form the protective
matrix. For example, Rodrigues et al. (2011) studied the viability of
ive probiotic strains (L. casei 01, L. acidophilus La-5®, L. acidophilus
Ki, B. animalis BB-12®, and B. lactis Bo) immobilized on various
polymers [sodium alginate, xanthan gum, L-carrageenan, cellulose
acetate phthalate (CAP), chitosan/sodium alginate, and whey protein
concentrate] at various concentrations and concluded that alginate
and whey proteins were the most suitable polymers, except for L.
acidophilus Ki and L. casei 01 at 2% (m/v) alginate.
Some examples of polymers used in the production of GR tablets
containing probiotics are summarized in Table 6.4.
Polymers Reference
Sodium alginate in combination with Chan and Zhang, 2002, 2005
hydroxypropylcellulose
HPMC acetate succinate Stadler and Vernstein, 2003
Carboxymethyl high amylase starch Calinescu et al., 2005
Carboxymethyl high amylase starch in Calinescu and Mateescu,
combination with chitosan 2008
HPMC phthalate Klayraung et al., 2009
6.2.5.2 Gels
Gels are semisolid systems consisting of either suspensions made
up of small inorganic particles, forming a network of small discrete
particles (two phase gel), or large organic molecules interpenetrated
by a liquid in such a manner that no apparent boundaries exist
between them (single phase gel) (USP31 2008). In Europe, gels are
deined in a slightly different manner as “liquids gelled by means of
suitable gelling agents” (Eur. Pharmacopeia 2008).
Gels can be used to administer drugs topically, into body cavities
or orally. Few commercialized vaginal gels containing probiotics
246 Development of Probiotic Dosage Forms
6.2.5.4 Pellets
Pellets can be deined as small, free-lowing, spherical, or semi-
spherical solid units, typically from about 0.5 mm to 1.5 mm, usually
intended for oral administration and generally manufactured by
the process of extrusion-spheronization (process usually used in
the pharmaceutical industry to produce spheroids with uniform
size) (Troy 2005). The death of intestinal microlora by antibiotic
or radiation treatment can lead to overgrowth of pathogenic
microorganisms. Reintroduction of a normal lora into the
“sterilized” gut may help to resolve this problem. Pellets containing
bacteria have been prepared by extrusion-spheronization to assess
the effects of processing on bacterial survival. Gram-negative
aerobic (Escherichia coli), gram-positive aerobic (Staphylococcus
saprophyticus and B. subtilis), and gram-positive anaerobic (B.
Dosage Forms Characterization 247
dissolved in the medium and the mean values are calculated from
six parallel measurements. The dissolution test is used to determine
the dissolution rate of the active ingredients of dosage forms placed
in an immersion luid at 37 ± 0.5°C. The basket (apparatus 1), the
paddle (apparatus 2), the reciprocating cylinder (apparatus 3),
or the low-through cell (apparatus 4) may be used (Fig. 6.4). At
speciied time, samples of the dissolution liquid are withdrawn,
iltered, and the amount of bioactive ingredient released is evaluated
by suitable analytical methods (USP31 2008). In the case of dosage
forms containing probiotics, the enumeration of viable cells released
should be done.
investigated. In this study, two strategies were used: (i) the direct
incorporation of bacterial suspensions into pellets, sodium alginate
beads, and ilms, and (ii) the mixture of freeze-dried bacteria with
tabletting excipients to produce tablets. The vegetative forms of L.
plantarum, L. rhamnosus GG, L. lactis, and L. bulgaricus and spore
form of B. subtilis were considered as model bacterial strains. With
this study, the successful production of various dosage forms for the
delivery of viable lactobacilli with an antibacterial activity against
multi-resistant bacteria was demonstrated.
As stated above, probiotics viability should also be maintained
after the manufacture of dosage forms. For example, one of the
methods described to evaluate the relationship between the applied
pressure during tablets manufacturing and bacterial viability
consisted in producing tablets using different compression forces.
After compression, the tablets are dissolved/disintegrated in an
accurate volume of SIF, and the resulting suspension is diluted and
plated in MRS (Man, Rogosa and Sharpe) agar. After incubation for
48 h at 37°C, the number of viable cells was determined. The initial
number of viable cells in the individual tablet doses of probiotics
should be also evaluated following the same procedure without
compressing the powder mixture.
In the studies of Stadler and Viernstein (2003), the degradation
of lyophilized viable LAB cells during the tablets formulation and
preparation as well as the survival in artiicial gastric juice were
evaluated. The loss of bacteria due to the compression of tablets was
evaluated by calculation from the number of viable cells in the mixed
powders before compression and in the tablets resulting from the
compression. The results of these studies showed that for achieving
the resistance of the gastric juice, high content of HPMCAS as well as
a compaction force of high or medium intensity are required.
Klayraung et al. (2009) also studied the inluence of matrix-
forming excipients and compression forces on the viability of
probiotics. The results of these studies showed that the percentage of
matrix-forming agents and the compression forces can signiicantly
inluence the tensile strength and disintegration of tablets containing
probiotics and also the survival of bacteria. These studies also
demonstrated that the tablets produced with high compression forces
exhibited a slow disintegration time and a bacterial viability greater
than 80%. Furthermore, the addition of sodium alginate resulted in
Packaging and Storage of Dosage Forms Containing Probiotics 253
References
Ahmad, F., M. Alam, et al. (2008). “Development and in vitro evaluation of
an acid buffering bioadhesive vaginal gel for mixed vaginal infections.”
Acta Pharmaceutica 58(4): 407–419.
Baerheim, A., E. Larsen, et al. (1994). “Vaginal application of lactobacilli in
the prophylaxis of recurrent lower urinary tract infection in women.”
Scandinavian Journal of Primary Health Care 14(1): 11–16.
Bora, P., V. Puri, et al. (2009). “Physicochemical properties and excipient
compatibility studies of probiotic Bacillus coagulans spores.” Scientia
Pharmaceutica 77: 625–637.
Borges, S., J. Barbosa, et al. (2012). “Effects of encapsulation on the viability
of probiotic strains exposed to lethal conditions.” International Journal
of Food Science & Technology 47(2): 416–421.
Brachkova, M. I. (2010). Evaluation of the Viability of Lactobacillus spp. in
Different Dosage Forms. PhD, University of Lisbon.
Bruno, F. A. and N. P. Shah (2003). “Viability of two freeze-dried strains
of Biidobacterium and of commercial preparations at various
temperatures during prolonged storage.” Journal of Food Science
68(7): 2336–2339.
Çaglar, E., S. Kavaloglu Cildir, et al. (2006). “Salivary mutans streptococci
and lactobacilli levels after ingestion of the probiotic bacterium
Lactobacillus reuteri ATCC 55730 by straws or tablets.” Acta
Odontologica Scandinavica 64(5): 314–318.
Çaglar, E., S. Kavaloglu, et al. (2007). “Effect of chewing gums containing
xylitol or probiotic bacteria on salivary mutans streptococci and
lactobacilli.” Clinical Oral Investigations 11(4): 425–429.
Calinescu, C., J. Mulhbacher, et al. (2005). “Carboxymethyl high amylose
starch (CM-HAS) as excipient for Escherichia coli oral formulations.”
European Journal of Pharmaceutics and Biopharmaceutics 60(1): 53–
60.
Champagne, C. P., F. Mondou, et al. (1996). “Effect of polymers and storage
temperature on the stability of freeze-dried lactic acid bacteria.” Food
Research International 29(5–6): 555–562.
References 257
7.1 Introduction
To better understand the regional or country-speciic regulations,
this chapter will irst provide a broad overview of the global legal
framework concerning probiotics.
There is no universal deinition of probiotics that can be
considered generically either as a food (including food additives and
dietary supplements) or as a drug (medicines). Nevertheless, the
deinition of probiotics by a joint Food and Agriculture Organization
of the United Nations (FAO)/World Health Organization (WHO)
expert consultation in October of 2001 (already mentioned in
Chapter 1) has been widely used. These experts produced a report on
Evaluation of Health and Nutritional Properties of Probiotics in Food
Including Powder Milk with Live Lactic Acid Bacteria. This report,
agreement deines how nations can regulate food safety and apply
animal and plant health measures. The goal of these agreements is
that technical measures and health concerns do not create artiicial
obstacles to trade.
Codex Alimentarius also contributes directly to probiotics and all
related issues from a legal point of view via the Nutrition and Health
Claims standard developed and revised in the beginning of the 21st
century (Codex Alimentarius: www.codexalimentarius.net)
2001 and 2002 and of the concept of QPS for the safety assessment
of bacteria, recently introduced by EFSA, as previously mentioned).
These documents together with other information published in the
meantime, identify essential steps needed to select and validate
a probiotic strain (Table 7.2). Then according to each country’s
of isolation (human, animal, environmental, food), acid resistance, bile resistance, and
other such traits may not be relevant for speciic applications, and therefore cannot
be considered essential.
bRandomized, double blind, placebo controlled.
7.3.1 Asia–Pacific
7.3.1.1.1 Regulations
http://www.comlaw.gov.au/ Australian Government Law site
containing all standards included in Australian New Zealand Food
Standards Code
7.3.1.2 China
In China, probiotics tend to be considered a foodstuff either as
a novel food or as a health food, if health claims are invoked. The
central government authority with jurisdiction over food and health
food (as well as medicines and cosmetics) is the State Food and
Drug Administration (SFDA) but the Ministry of Health and other
entities such as the General Administration of Quality Supervision,
Inspection and Quarantine, the State Administration for Industry
and Commerce, and so on are also in charge of the regulation of
food, drug and cosmetic products. (http://www.chinafdc-law.com/
glossary.html Accessed January 2012)
The irst Food and Hygiene Law of the People’s Republic of China
was promulgated in 1995 (Kaushik and Kaushik, 2010) and the
current basic food law is Food Safety Law, which entered into force
in 1 June 2009; they both deine food as any inished product or raw
materials provided for people to eat or drink, and articles that are
traditionally food and medicine, excluding articles that are used for
the purpose of medical treatment.
Novel food has been regulated by the regulation by “Administrative
Measures on Novel Food 2007” and included four types (article 2).
Type 1 refers speciically microorganisms that are not traditionally
consumed in China. Types 2 and 3 also refer microorganisms, which
are not traditionally consumed in China or new varieties, as raw
food materials derived (type 2) or as food processing. According to
the regulation, production and import of novel food are subject to
pre-market approval by China’s Ministry of Health. Probiotic strains
have already been approved by the current regulation (Lactobacillus
paracasei GM-080 and L. paracasei GMNL-33 by GenMont in 2008)
http://www.genmont.com.tw/englishwebsite/company.html
Accessed December 2011.
276 Guidelines and Regulations
7.3.1.2.1 Regulations
Food Safety Law of the People’s Republic of China. June 1 2009.
(Food Safety Law)
Ministry of Health. Administrative Regulations for Health Food.
Order No. 46; 1996-06-01. (Health Food Regulation)
Ministry of Health: Technical Standards for Testing and Assessment
of Health Food. Order No. 42; 2003
Probiotics Legal Status 277
7.3.1.3 Japan
Japan is a key player in probiotic-based functional foods accounting
for more than half of global probiotic foods market. These probiotic
products have different applications, including immunity, allergy,
cold-like symptoms, gastrointestinal health, blood pressure levels,
cholesterol levels, and so on (Amagase, 2008; He and Benno, 2011).
In terms of food regulation, Japan began a systematic research
on health beneits of foods in 1984. As a consequence, a deinition
for functional food was provided by the Japanese scientiic academic
community involved. In this context, functional foods were deined as
those foods with three functions: apart from nutrient provision and a
sensory function, functional foods also have additional physiological
functions contributing to health maintenance (Shimizu, 2003).
Dynamic changes began in 1991 when the Japanese Ministry of
Health, Labour and Welfare established the regulatory system “Foods
for Speciied Health Use” (FOSHU) in which foods wishing to bear
health claims on the label have to be examined and approved, prior
to use, by the Council of Pharmaceutical Affairs and Food Hygiene
(practical minimum period required for approval is approximately
one year) in terms of speciic safety and eficacy requirements
including:
(i) Proven scientiic effectiveness on the human body functions;
278 Guidelines and Regulations
&'(
Figure 7.1 Categories of food for speciied uses for regulation purposes.
7.3.1.3.1 Regulations
Food for Speciied Health Uses: http://www.mhlw.go.jp/english/
topics/foodsafety/hc/02.html
Food with nutrient function claim: http://www.mhlw.go.jp/english/
topics/foodsafety/hc/01.html
Food Sanitation Law: http://www.tokio.polemb.net/iles/
Gospodarka/Handel/food-e.pdf
7.3.2 Europe
Probiotics are consumed by Europeans in foodstuffs (food and food
supplements) (Saxelin 2008). The legislation on foodstuffs in the 27
country states of the EU is not harmonized.
The principles concerning food safety and consumer protection
are established in the legislation of the member states. However,
EU has established general principles and requirements of food law
with the aim of protecting human life and health and to integrate a
food safety policy based on “farm to fork” approach. EU food law also
aims to harmonize member states requirements in order to allow
the free movement of food in EU. Food safety and general consumer
protection is under competence of Directorate General for Health
and Consumer (DG SANCO). For registration purposes, as already
mention in Section 7.2.1, EU has developed the QPS list for microbial
cultures, which does not need further safety determination.
Food and food supplements containing probiotics are covered by
Regulation 178/2002/EC (which is the legal general framework) and
Directive 2000/13/EC. EU Regulation 178/2002/EC deines food (or
foodstuff) as “any substance or product, whether processed, partially
processed or unprocessed, intended to be, or reasonably expected to
be ingested by humans.” The Directive 2002/46/EC relating to food
supplements [which deine as “foodstuffs the purpose of which is
to supplement the normal diet and which are concentrated sources
of nutrients (vitamins, and minerals) or other substances with a
nutritional or physiological effect, alone or in combination, marketed
in dose form, …”] establishes harmonized rules for their labeling.
Dose forms may be capsules, tablets, pastilles, pills, powders (in
sachets) or liquids in measured doses, and so on. This legislation
also aim to ensure that these products are safe and appropriately
labeled so that consumers can make informed choices.
In EU, food labels may invoke nutrition claims, if they are listed
in the annex of EC Regulation 1924/2006, and health claims, after
approval by European Commission, according to the requirements
set out in this Regulation. Any statement relating food and health
may be interpreted as a claim and should be based on generally
accepted scientiic evidence and easily understood by consumers.
Health claims are deined as any “claim that states, suggests or
implies that a relationship exists between a food category, a food
282 Guidelines and Regulations
of Food Additive Safety (OFAS) in the Center for Food Safety and
Applied Nutrition (CFSAN) at FDA. This petition has four essential
elements for the safety assessment: identity of the additive, probable
exposure, evaluation of safety, and limitations of conditions of use.
FDA recommends that manufacturers consult the FDA about food
ingredient status (pre-petitions consultations).
If the probiotic product is considered a biologic product (a
subset of drugs), a Biologics License Application is necessary (which
includes applicant information, product/manufacturing information,
pre-clinical studies, clinical studies, labeling) and the pertinent
drug regulations, including applying for an Investigation New Drug
Application, should be fulilled.
A probiotic product categorized as a dietary supplement does not
need a premarket approval, but the act requires that to market dietary
supplements that contain “new dietary ingredients,” manufacturers
and distributors must previously notify FDA (it is necessary to wait
at least 75 days after the iling date of the notiication in order to
market the product). Dietary Supplement Health and Education Act
(DSHEA Act) of 1994, which signiicantly amends the Act, deines
“new dietary ingredients” as a dietary ingredient that was not
marketed in the United States before 15 October 1994. In a position
paper, the International Probiotics Association, the European Food
and Feed Cultures Association, and the International Food Additives
Council state that “individual strains belonging to microbial species
with a history of safe use as starter culture used for food fermentation
or as a probiotic in food are not New Dietary Ingredients under
the DSHEA Act;” on the contrary, each strain of microbial species
without a history of safe use as food (including dietary supplements
before 15 October 1994) must be evaluated individually and a
premarket notiication for a new dietary ingredient submitted to
the FDA (http://internationalprobiotics.org/news/upload/pdf/11.
pdf). There are three categories of claims that could be made for
dietary supplements and foods: health claims, structure/function
claims, and nutrient content claims. However, for food or dietary
supplements containing probiotics, only the irst and the second
claim are relevant.
Health claims describe the relationship of a food, or ingredient to
a disease or health-related condition. Legislation the 1990 Nutrition
Labeling and Education Act (NLEA), the 1997 FDA Modernization Act,
and the 2003 FDA Consumer Health Information for Better Nutrition
286 Guidelines and Regulations
7.3.3.1 Legislation
Federal Food Drug and Cosmetic Act of 1938
Public Health Service Act of 1944
Nutrition Labeling and Education Act of 1990
Dietary Supplement Health and Education Act of 1994
FDA Modernization Act of 1997
Probiotics Legal Status 287
Parameter Details
7.3.4.1 Legislaon
• Resolution no. 16: Procedures for registration of food and/or
new ingredients (Brazil, 1999d).
• Resolution no. 17: Risk assessment and food safety (Brazil,
1999e).
• Ministerial Order 398/1999 and Resolution no. 18: Basic
guidelines for analysis and approval of claims for functional
and/or health property mentioned on the labeling of the foods
(Brazil 1999a, 1999b).
• Resolution no. 19: Procedures for registration of foods
claiming functional and/or health properties (Brazil, 1999c).
• Resolution RDC no. 2: Technical Regulation of bioactive
substances and isolated probiotics with claim for functional
and/or health properties, annexed to that resolution (Brazil,
2002).
• Resolution RDC no. 27: Demonstrates about the categories
of food and packaging, which are excepted or compulsory to
sanitary registration, which apply to all types of food (Brazil,
2010).
290 Guidelines and Regulations
7.4 Conclusions
The global regulatory situation related with probiotics is still
undergoing development and change despite the tentative
harmonization by all countries. It is clear from the different
standpoints assumed by the different countries that probiotics are
of importance in terms of health promotion and disease prevention
among consumers. Nevertheless, there are still many challenges
to overcome until a steady, harmonized, and overall regulatory
framework is implemented worldwide to ensure the quality and
safety for proper utilization of probiotics in different countries; in
this context, regulatory bodies, food scientists, manufacturers, and
even consumers need to brace forces in order to achieve such goal.
References
Amagase, H. (2008) Current marketplace for probiotics: a Japanese
perspective. Clin. Infect. Dis., 46, S73–S75.
Brazil (1999a) Ministério da Saúde. Agência Nacional de Vigilância Sanitária.
Portaria no 398, de 30 de abril de 1999. Regulamento Técnico que
Estabelece as Diretrizes Básicas para Análise e Comprovação de
Propriedades Funcionais e ou de Saúde Alegadas em Rotulagem de
Alimentos. Diário Oicial da República Federativa do Brasil. Brasília, 03
maio, 1999. Available at: http://www.crn6.org.br/LEGISLACAO_%20
A L I M E N TAC AO _ E _ % 2 0 N U T R I C AO / p o r t a r i a / Po r t a r i a % 2 0
ANVISA%20398-1999%20Rotulagem%20de%20alimentos.pdf.
Accessed 24 september 2012.
Brazil (1999b) Ministério da Saúde. Agência Nacional de Vigilância Sanitária.
Resolução no 18 de 30 de abril de 1999. Aprova o Regulamento Técnico
que Estabelece as Diretrizes Básicas para Análise e Comprovação de
Propriedades Funcionais e ou de Saúde Alegadas em Rotulagem de
Alimentos. Diário Oicial da República Federativa do Brasil. Brasília, 03
nov. 1999. Available at: http://www.anvisa.gov.br/legis/resol/18_99.
htm. Accessed 24 september 2012.
Brazil (1999c) Ministério da Saúde. Agência Nacional de Vigilância Sanitária.
Resolução no 19, de 30 de abril de 1999. Aprova o Regulamento
Técnico de Procedimentos para Registro de Alimento com Alegação de
Propriedades Funcionais e ou de Saúde em sua Rotulagem. Diário Oicial
da República Federativa do Brasil. Brasília, 10 dez. 1999. Available at:
http://e-legis.bvs.br/leisref/public/showAct.php?id=110. Accessed
24 September 2012.
References 291