Biofilm Formation in Food Industries - A Food Safety Concern

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Food Control 31 (2013) 572e585

Contents lists available at SciVerse ScienceDirect

Food Control
journal homepage: www.elsevier.com/locate/foodcont

Review

Biofilm formation in food industries: A food safety concern


Sokunrotanak Srey, Iqbal Kabir Jahid, Sang-Do Ha*
School of Food Science and Technology, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myeon, Anseong-si, Gyeonggi-do 456-756, Republic of Korea

a r t i c l e i n f o a b s t r a c t

Article history: Foodborne diseases have always been a threat to human health. They are considered an emergent public
Received 6 July 2012 health concern throughout the world. Many outbreaks have been found to be associated with biofilm. It
Received in revised form is well documented that biofilm has become a problem in food industries as it renders its inhabitants
26 November 2012
resistant to antimicrobial agents and cleaning. In this review, biofilm formation in dairy, fish processing,
Accepted 1 December 2012
poultry, meat, and Ready-To-Eat foods industries are discussed, as well as the biofilm forming abilities’ of
various microorganisms and the influence of food contact surface materials on biofilm formation. In
Keywords:
addition, the conventional and emergent control strategies used to gain more proximity to efficiently
Biofilms
Food industry
maintain good hygiene throughout food industries is discussed.
Control strategies Ó 2012 Elsevier Ltd. All rights reserved.
Chemical control
Novel control strategies
Hurdle technology

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .573
2. Biofilm and development steps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .573
2.1. Initial attachment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 573
2.2. Irreversible attachment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
2.3. Early development of biofilm architecture (microcolony formation) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
2.4. Maturation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
2.5. Dispersion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
3. Biofilm formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .574
3.1. Biofilm forming strength . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
3.2. Food contact surface materials for biofilm formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 575
4. Biofilm: problems in food industry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .575
4.1. Produce industry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 575
4.2. Dairy industry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 576
4.3. Fish processing industry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 576
4.4. Poultry industry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
4.5. Meat industry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
4.6. Ready-to-eat (RTE) foods industry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
5. Biofilm control strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .578
5.1. Cleaning and disinfection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578
5.2. Clean-in-Place (CIP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578
5.3. Chemical-based control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
5.3.1. Sodium hypochlorite . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
5.3.2. Hydrogen peroxide (H2O2) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
5.3.3. Ozone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
5.3.4. Peracetic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579

* Corresponding author. Tel.: þ82 31 670 4831; fax: þ82 31 765 4853.
E-mail address: sangdoha@cau.ac.kr (S.-D. Ha).

0956-7135/$ e see front matter Ó 2012 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.foodcont.2012.12.001
S. Srey et al. / Food Control 31 (2013) 572e585 573

5.4. Other approaches of biofilm control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 580


5.4.1. Ultrasonication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 580
5.4.2. Enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 580
5.4.3. Phages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 580
5.5. Hurdle technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581

1. Introduction Svabic-Vlahovi 
c, 2004; Stepanovic, Cirkovi 
c, Mijac, & Svabi 
c-
Vlahovic, 2003). The biofilm formation is a stepwise and dynam-
Foodborne diseases cover a wide range of illnesses and are ical process consisting of (i) initial attachment, (ii) irreversible
triggered by agents that were consumed along with food. As stated attachment, (iii) early development of biofilm architecture, (iv)
by the World Health Organization (WHO), the contamination of maturation, and (v) dispersion (Fig. 1).
food may occur at any stage in the process from food production to
consumption (“farm to fork”) and can result from environmental 2.1. Initial attachment
contamination, including the pollution of water, soil or air (WHO,
2007; WHO, n.d). According to the WHO, they are considered an The bacteria’s initial attachment can be active or passive,
emergent public health problem in both developed and developing depending on their motility or the gravitational transportation of
countries (WHO, 2007). The Centers for Disease Control and their planktonic (free floating), diffusion or shear force of the
Prevention (CDC) has stated that foodborne diseases cause surrounding fluid phase (Kumar & Anand, 1998). The cell’s adhesion
approximately 1000 reported disease outbreaks (CDC, 2011) and during this process strongly depends on the physiochemical prop-
a projected 48 million illnesses, 128,000 hospitalizations, and 3000 erties of the bacterial cell surface (Ferreira, Pereira, & Melo, 2010). At
deaths annually in the United States between 1996 and 2010 first, the adherent cells, those that originate biofilm formation on
(Scallan, 2011; Scallan et al., 2011). In addition, many other a surface, possess only a small quantity of extracellular polymeric
outbreaks of pathogens have been found to be associated with substance (EPS) and many are capable of independent movement
biofilms (Aarnisalo, Lundén, Korkeala, & Wirtanen, 2007; Dykes, (O’Toole & Kolter, 1998) by pilus-mediated twitching or gliding
Sampathkumar, & Korber, 2003; Lapidot, Romling, & Yaron, 2006; motility. In this stage, the adhesion is reversible since the attached
Waak, Tham, & Danielsson-Tham, 2002). microorganisms are not yet committed to the differentiation
It is now recognized that biofilms are a frequent source for processda series of morphological changesdwhich leads to biofilm
infections (Costerton, Stewart, & Greenberg, 1999). Around 80% of formation, and many of the cells may detach from the surface and
persistent bacterial infections in the United States were found to be return to the planktonic lifestyle (Stoodley, Sauer, Davies, &
associated with biofilms (Janssens et al., 2008). Since they can Costerton, 2002).
render their inhabitants more resistant to disinfectant (Bower, The surface properties also play an important role in bacterial
McGuire, & Daeschel, 1996; Sidhu, Langsrud, & Holck, 2001), bio- adhesion. Generally, any surface is vulnerable to biofilm develop-
films have become problematic in a wide range of food industries, ment including plastic, glass, metal, wood, and food products. The
including brewing (Flemming & Ridgway, 2009), seafood process- adhesion to the surface is also dependent upon the physicochemical
ing (Shikongo-Nambabi, 2011), dairy processing (Chmielewski & properties of the surface such as texture (rough or smooth) (Donlan,
Frank, 2003), poultry processing (Harvey, Keenan, & Gilmour, 2002), surface charge (Abdallah, Chaieb, Zmantar, Kallel, & Bakhrouf,
2007), and meat processing (Sofos & Geornaras, 2010). 2009), hydrophobicity (Donlan, 2002), pH, temperature (Nilsson,
Much research has been performed to gain deeper under- Ross, & Bowman, 2011), and nutrient composition of the pre-
standing of biofilms and to identify a solution in order to avoid conditioning solution (Donlan, 2002; Gerstel & Römling, 2001). For
contamination of foodstuffs. The objective of this review is to instance, the research of Sinde and Carballo (2000) found that
summarize the problem of biofilms in food industries and the Salmonella and Listeria can attach in a higher numbers to hydro-
current and innovative control strategies that have been used to phobic surfaces than the hydrophilic ones. Furthermore, the surfaces
combat the challenges caused by biofilms. overlaid by a so-called conditioning film comprising macromole-
cules, such as organic substances, will enhance the attachment of
bacterial cells (Tang, Flint, Bennett, Brooks, & Morton, 2009). It has
2. Biofilm and development steps

Biofilms are an aggregation of microorganism attached to and


growing on a surface (Costerton & Stewart, 2001). The formation and
development of biofilms is affected by many factors, including the
specific bacteria strain (Borucki, Peppin, & White, 2003; Chae & Schraft,
2000), material surface properties, and environmental parameters
such as the pH and nutrient levels and temperature (Donlan, 2002).
Biofilm cells are more resistant to antimicrobial agents than planktonic
bacteria, as they have a barrier which prevents or lessens the contact
with antimicrobial agents (O’Toole & Kaplan, 2000).
Any type of microorganisms, including those that cause spoilage Fig. 1. Stages of biofilms development. This diagram is a cartoon of the 5 stages of
and pathogenic microorganisms, could form a biofilm and play biofilm development: 1 reversible attachment, 2 irreversible attachment, 3 early
development of biofilm architecture, 4 maturation and 5 finally dispersion. Under the
a key role in many infections (Parsek & Singh, 2003). Experiencing cartoon are 5 electron micrographs showing what the biofilm actually looks like at
an inhospitable environment, it is important for bacteria to form each stage. Image by D. Davis from Monroe, D “Looking for chinks in the armor of
a biofilm as a survival strategy (Stepanovi c, Cirkovi
c, Ranin, & bacterial biofilms” PLoS Biol, Vol. 5, issue 11.
574 S. Srey et al. / Food Control 31 (2013) 572e585

also been shown that any pre-existing EPS will facilitate the adhe-
sion (Flemming & Schaule, 1988).

2.2. Irreversible attachment

The change from reversible to irreversible attachment is a shift


from a weak interaction of the bacteria with the surface to
a permanent bonding with the presence of EPS (Stoodley, Sauer,
et al., 2002). After irreversible attachment, strong shear force or
chemical breaking of the attachment forces by enzymes, deter-
gents, surfactants, sanitizers (Sinde & Carballo, 2000), and/or heat
is needed for biofilm removal (Augustin, Ali-Vehmas, & Atroshi,
2004; Maukonen et al., 2003; Sinde & Carballo, 2000). Gerke, Kraft,
Süssmuth, Schweitzer, and Götz (1998) worked on Staphylococcus
epidermis and has shown that adherent bacteria produce a poly-
saccharide intercellular adhesion that bonds the cells together and
facilitates microcolony formation and biofilm maturation.

2.3. Early development of biofilm architecture (microcolony


formation)

Microcolony formation results from concurrent accumulation


and growth of microorganisms and is associated with the produc-
tion of EPS (Chmielewski & Frank, 2003), which helps strengthen
the bond between the bacteria and the substratum and stabilizes
the colony from any environmental stress (Donlan, 2002). Studies
of bacterial species in natural systems have shown that accumu-
Fig. 2. SEM images of biofilm formation on polystyrene coupons in A. hydrophila.
lation could embrace the recruitment of planktonic cells from the
surrounding medium as a result of cell-to-cell communication
(quorum sensing) (McLean, Whiteley, Stickler, & Fuqua, 1997; Pesci possible causes of biofilm detachment (Kaplan, Ragunath,
et al., 1999). O’Toole and Kolter (1998) indicated that in order to Ramasubbu, & Fine, 2003; Kaplan, Ragunath, Velliyagounder,
allow microcolony formation of Pseudomonas aeruginosa PA14 to Fine, & Ramasubbu, 2004). Detachment seems to be an active
take place type IV pili are required. Swimming motility was process which allows for the colonization of new niches (Sauer
considered to enable the microorganisms to overcome repulsive et al., 2002). In addition, starvation is also considered as a reason
forces at the surfaceewater interface, allowing them to reach the of detachment and allows bacteria to search for a nutrient-rich
substratum and form microcolonies by twitching motility powered environment (O’Toole & Kaplan, 2000).
by extension and retraction of type IV pili (Skerker & Berg, 2001). Other researchers have also proposed the involvement of
Microcolonies can be very beneficial as they provide interspecies enzymatic activity in the degradation of EPS. For instance, the
substrate exchange and/or mutual end-product removal to bacteria production of surface protein releasing enzyme dispersin B that
(Costerton, Lewandowski, Debeer, Caldwell, & Korber, 1994). degrades the biofilms of Staphylococcus epidermidis is found in the
Actinobacillus actinomycetemcomitans (Kaplan et al., 2004). Another
2.4. Maturation study indicates that biofilms can be removed from surfaces by
gentle rinsing if there is an overexpression of alginate lyase which
The biofilm maturation is the step where it develops into an would cause the degradation of alginate.
organized structure which can be flat or mushroom-shaped (Fig. 2),
depending on the nutrient source it is reliant upon (Chmielewski & 3. Biofilm formation
Frank, 2003; Klausen et al., 2003). In order to reach structural
maturity, periods of 10 days or more are required (Stoodley, Sauer, 3.1. Biofilm forming strength
et al., 2002). Bacteria grow under sessile form in heterogeneous
complex-enclosed microcolonies scattered with open water chan- The ability to form biofilm varies greatly not just between species.
nels (Davey & O’Toole, 2000). In a study, the mature biofilms was Even in the same species with different strains and serovars, biofilm
evaluated and compared with chemostat cultures of P. aeruginosa formation strength can vary significantly. For instance, in a study on
by DNA microarray technology (Whiteley et al., 2001). The study the ability of five Salmonella enterica serovars to attach to and colo-
showed that over 70 genes were altered, including genes encoding nize intact and cut lettuce and cabbage surfaces (Patel & Sharma,
proteins involved in translation, metabolism, membrane transport 2010) showed that S. enterica serovars Tennessee and Thompson
and/or secretion, and gene regulation. can produce considerably more biofilm than serovars Braenderup,
Negev, and Newport; and could be considered as strong biofilm
2.5. Dispersion formers according to the criteria suggested by Stepanovi c et al.
(2004). In another work, Listeria monocytogenes Scott A and 3990
Dispersion is the last step in the biofilm formation cycle, and it were known to be high biofilm producers, while L. monocytogenes
allows the cells to revert into their planktonic form (Sauer, Camper, strains YM96 and 303 were moderate biofilm producers and
Ehrlich, Costerton, & Davies, 2002). External perturbation, such as L. monocytogenes 17 was the least productive biofilm former
increased fluid shear (Stoodley, Cargo, Rupp, Wilson, & Klapper, (Chmielewski & Frank, 2006). However, there are also other factors
2002), internal biofilm processes, such as endogenous enzymatic that influence biofilm forming capacity, specifically surface proper-
degradation, or the release of EPS or surface-binding protein, are all ties and nutrient availability. Salmonella spp. and L. monocytogenes
S. Srey et al. / Food Control 31 (2013) 572e585 575

strains preferably adhere to hydrophobic surface materials (Donlan, documented that the safety of prolonging the shelf life of fresh-cut
2002). Furthermore, biofilm production of Bacillus cereus on stainless produce depends on the washing step, while the efficacy of sani-
steel coupons at the aireliquid interface was found to be thicker than tizers on produce can only reduce at most 2 log of bacteria; the
that in submerged systems (Wijman, de Leeuw, Moezelaar, microorganisms presumably are protected by their location in the
Zwietering, & Abee, 2007). Similarly, Staphylococcus isolated from plant tissue (internalized, in stomata, cracks, crevices, cut surfaces),
food and food processing environments was studied, and the biofilm and/or by biofilms produced by the microbes themselves (Whipps,
was thicker with the presence of sodium chloride or glucose Hand, Pink, & Bending, 2008). Trimming, cutting, washing, rinsing,
(Møretrø et al., 2003). It has also been speculated that carbohydrate dewatering and packaging are all used in produce industry and are
metabolism may have an effect on biofilm production among various considered to be the primary source of cross-contamination
gram-positive bacteria (Pillai et al., 2004). As can be seen, biofilm (Suslow, 2001). López-Gálvez, Gil, Truchado, Selma, and Allende
formation capacity varies between genera, species, and strains, and it (2010) investigated the effect of subsequent washing with chlo-
is influenced by other factors such that one type of bacteria can be rine or sodium hypochlorite on short-term cross-contaminated
a strong biofilm producer under a certain environment and become fresh-cut lettuce. The results indicated that subsequent sanitation
weak in another environment. steps cannot efficiently inactivate Escherichia coli cells on the
vegetable tissue. Through scanning electron microscopy, bacteria
3.2. Food contact surface materials for biofilm formation cells were detected in clusters or tissue stomata (Fig. 3) where they
are considered as protected from disinfectants, which could explain
Besides bacteria genus and species, extrinsic factors also play the inefficiency of disinfecting solutions. However, chlorine dioxide
a major role influencing the degree of attachment and biofilm and sodium hypochlorite solutions were able to inactivate a large
formation. Materials that make up food contact surfaces were portion of E. coli cells that were transferred from an inoculated
postulated to have a large effect on the level of attachment and bio- sample to wash water. It has been suggested that the use of sani-
film formation. The materials used for food contact surfaces are tation solutions may be able to reduce cross-contamination
known to be stainless steel, glass, rubber, polyurethane (Chia, Goulter, between clean and contaminated produce during the washing
McMeekin, Dykes, & Fegan, 2009), Teflon, nitrile butyl rubber (NBR, process. It is recommended that disinfectants should be used in
Buna-n) (Storgards, Simola, Sjöberg, & Wirtanen, 1999), and order to avoid cross-contamination from contaminated produce to
wooddfor developing countries (Mariani et al., 2011). Stepanovic clean produce (Keskinen, Burke, & Annous, 2009; López-Gálvez
et al. (2004) reported that Salmonella spp. and L. monocytogenes can et al., 2010).
produce high biofilms on plastic surfaces. In another study, the Produces are generally consumed raw, hence packingdthe last
surface roughness of a polyesterurethane conveyor belt was reported step of processingdis considered as one of the critical control
to have a significant influence on the biofilm forming ability of points. This last step should be controlled attentively and regularly
L. monocytogenes. Even the weak strains demonstrated the ability to to avoid re-contamination. It has been observed that 12.3% of all
form biofilm and could not totally be eliminated (Chaturongkasumrit, foodborne outbreaks from 1990 to 2007 were related to fresh
Takahashi, Keeratipibul, Kuda, & Kimura, 2011). Adetunji and Isola produce whereas 10% caused by improper handling post harvesting
(2011) studied the biofilm formation on wood, stainless steel, and from farm and the rest were associated with growing, packing,
glass surfaces, with the results demonstrating that wood encourages shipping or processing of fresh produce (Alliance For Food and
biofilm formation due to its porosity and absorbency, which can
entrap organic material and bacteria. The authors stated that glass is
the preferred food contact surface owing to its smooth surface and
corrosion resistant properties, while stainless steel can resist the
impact damage better but it is vulnerable to corrosion. Chia et al.
(2009) found that Salmonella attached the best to Teflon, followed
by stainless steel and glass, then Buna-n and polyurethane. It is
suggested that the adhering cells depend on the interfacial free
energies of the surface. Their results suggested that surface rough-
ness has no correlation with adhesion, while many other authors
have stated the opposite (Howell & Behrends, 2006; Scardino,
Harvey, & De Nys, 2006). It has been speculated that the difference
between the degree of surface roughness studied depends on
a subjective assessment (e.g. polished or unpolished), which could be
the reason for the different observations (Chia et al., 2009). Corre-
spondingly, Arnold, Boothe, Suzuki, and Bailey (2004) reported that
there were dramatically less bacterial cells attached to electro-
polished stainless steel compared to an untreated stainless steel
surface. It was also suggested that besides the material itself other
hygienic design such as welding, joints, corners, and equipment
design could also be an important factor affecting biofilm formation
(Guðbjörnsdóttir, Einarsson, & Thorkelsson, 2005).

4. Biofilm: problems in food industry

4.1. Produce industry

Currently, microbial control strategies are not efficient enough


to provide a complete eradication of hazardous microorganisms
without affecting product qualities (FAO/WHO, 2008, p. 151). It is Fig. 3. Attachment of A. hydrophila on iceberg lettuce stomata grown at 25  C for 24 h.
576 S. Srey et al. / Food Control 31 (2013) 572e585

Farming, 2010). In 2011, there was an outbreak linked to whole In another study, attachment and biofilm formation by
cantaloupe contaminated with L. monocytogenes (Caron 2011). It is L. monocytogenes on stainless steel (SS, type 304, no. 4 finish) and
speculated that the root cause of the outbreaks is the unsanitary Buna-n rubber (BN, acrylonitrile butadiene, 70 durometer), which
condition of the packing shed. Moreover, the microorganisms were are commonly used in food processing equipment, was evaluated
also found other places include the conveyor belt, drying area and (Helke, Somers, & Wong, 1993). According to the results, lactose had
floor drain (Neuman, 2011). It is supposed that pathogens become no effect on the attachment on either surface compared to the
firmly attached in inaccessible place (Neuman, 2011) and form control PBS, while other compounds showed a significance effect
biofilm then internalize within the produce (Sapers, 2001). on the attachment. The authors suggested that the repulsion
Microorganisms are found to preferentially attach to intact between negatively charged proteins and bacterial cell surfaces
surfaces of produce (Liao & Cooke, 2001). Gandhi, Golding, Yaron, could to some extent provide an explanation for the decrease in
and Matthews (2001) found evidence of bacteria at a depth of attachment. A similar study demonstrated that in the presence of
12 mm within intact alfalfa sprout tissue. In another study by Han, pasteurized whole milk with a relative humidity (RH) of 75%,
Sherman, Linton, Nielsen, and Nelson (2000), it was noticed that L. monocytogenes endured on the BN surface between 25  C and
there was no remarkable growth of E. coli O157:H7 found on 6  C, and 6  C on the SS surface and growing at 25  C on the SS
uninjured green pepper surfaces after inoculation and incubation surface, both at the same RH. At the same time, the attached cells
for 24 h at 37  C; while significant growth and multiplication was decreased over time and the slowest rate of decrease was at 6  C
found on intact surfaces. Sanitizers such as ozone, chlorine, and and 75.5% (Helke & Wong, 1994). It was also documented that the
organic acid were reported to be ineffective against microbial BN surface had an inhibitory effect on the growth of
biofilm (Ölmez & Temur, 2010). Furthermore, E. coli was reported to L. monocytogenes and several other foodborne pathogens (Helke &
have survived a 20,000 ppm chlorine soak and grew to a level of Wong, 1994; Ronner & Wong, 1993). According to Ronner and Wong
6 log CFU/g radish sprouts, which is remarkably higher than the (1993), this inhibitory effect varies depending on the nutrient
initial count on the seeds. It was demonstrated that bacteria were contents in the environment and at one-fifth the amount of
generally detected in the roots of radish sprouts and all across the tryptose-phosphate broth nutrients, there is an increase in lag time.
surface, and the cells are found to be located in biofilms attached to However, the effect can endure about 20 cycles of Clean-In-Place
radish sprout tissues during sprouting (Fransisca, Zhou, Park, & (CIP) (Helke & Wong, 1994). Furthermore, it was suggested that
Feng, 2011). Hydrogen peroxide is also relatively ineffective in a slow growth rate could improve resistance to antimicrobial
E. coli 766 elimination on cantaloupes; it was observed to be agents (Anwar, Dasgupta, & Costerton, 1990), which can to some
reduced by less than 1 log. Furthermore, immersion of cantaloupes extent explain why biofilms on BN are more resistant to sanitizers
in 1% hydrogen peroxide for 2 h did not show any increase in than normally formed biofilms (Wong, 1998).
efficacy. It is speculated that this inefficacy is a result of the Biofilm formation depends on many factors in dairy industry.
oxidation of the agents on the melon surface or from access to Dairy products are very susceptible to contamination by biofilms
endogenous catalase (Sapers & Sites, 2003). and it is challenging to eliminate those microorganisms.

4.3. Fish processing industry


4.2. Dairy industry
In the fish processing industry, both equipment and water
Milk is a very perishable product and is truly vulnerable to quality are considered to be major concerns. Also, many types of
contamination by various microorganisms. The major sources of fish-contaminated-bacteria are found to be biofilm-forming,
contaminated milk and milk products are usually considered to including Vibrio cholerae (Faruque et al., 2006), Vibrio para-
come from improperly cleaned and sanitized equipment (Jessen & haemolyticus (Enos-Berlage, Guvener, Keenan, & McCarter, 2005),
Lammert, 2003; Koutzayiotis, 1992). It is well documented that Vibrio vulnificus (Joseph and Wright 2004), and Vibrio alginolyticus
the bacteria frequently encountered in the dairy environment (Kogure, Ikemoto, & Morisaki, 1998). Many genera other than Vibrio,
belong to the genus Enterobacter (Salo, Ehavald, Raaska, Vokk, & such as L. monocytogenes, Salmonella spp., Bacillus spp., Aeromonas,
Wirtanen, 2006), Listeria (Waak et al., 2002), Lactobacillus, Micro- and Pseudomonas spp., are also known to be biofilm forming in fish
coccus, Streptococcus, Bacillus (Sharma & Anand, 2002) and Pseu- and seafood processing (Rajkowski, 2009).
domonas (Wiedmann, Weilmeier, Dineen, Ralyea, & Boor, 2000). In As important as any of the other components used in seafood
2011, there was a recall of approximately 20 pounds of raw milk industry, seawater is used instead of freshwater for economical
distributed in Washington state due to L. monocytogenes contami- reason. Even though the water is treated in a combined chlorina-
nation (Anonymous, 2011). It is speculated that the type of bacteria tion and Ultraviolet (UV) radiation system, treated seawater was
in the milk samples may have shown biofilm formation. For found to be contaminated with Vibrio spp., caused by biofilm
instance, the larger the amount of thermoduric Streptococci and formation in the seawater distribution system after the treatment
Bacillus species in pasteurized milk compared to raw milk could be process (Shikongo-Nambabi, Kachigunda, & Venter, 2010). Various
caused by the contamination of the dispersion of biofilm (Flint, authors have stated that the failure of chlorine to inhibit biofilm
Bremer, & Brooks, 1997). formation and mature biofilm was not due to the effect of pH
To date, there are many studies focused on the various bacteria (Momba & Binda, 2002; Shikongo-Nambabi et al., 2010). It has been
in dairy factories. Several studies (Mafu, Roy, Goulet, & Magny, established that there is a loss in water quality after undergoing
1990; Vanhaecke et al., 1990) have indicated that there is no treatment process, which is usually caused by cells detaching from
correlation between surface irregularities or roughness and the the biofilm in the distribution system due to the ineffectiveness of
bacteria attachment ability. However, Latorre et al. (2010) con- residual chlorine, and that this is common in water distribution
ducted a study on the presence of L. monocytogenes-containing systems (Momba & Makala, 2004; September, Els, Venter, & Brözel,
biofilm in milking equipment as a potential source of contamina- 2007; Shikongo-Nambabi et al., 2010). Furthermore, biofilm
tion on a dairy farm. After having obtained positive results, electron formation by seafood contaminated Vibrio harveyi on surfacesdbe
microscopy scanning of the equipment showed the presence of it plastic, cement slab or steel coupondwas also evaluated. Results
individual and clusters of bacteria, and it was postulated that it was showed that there was only a slight reduction of biofilm on the
mainly associated with scratched surfaces. concrete slabs and plastic (chlorine 20 ppm; 10 min), while the cells
S. Srey et al. / Food Control 31 (2013) 572e585 577

were completely killed at a higher concentration (chlorine Besides Salmonella spp., Campylobacter spp. are also commonly
100 ppm; 10 min) on the steel coupons. This is particularly found pathogens in poultry and poultry processing (Deming et al.,
important for shrimp hatcheries given that water storage tanks, 1987; Harris, Weiss, & Nolan, 1986; Hopkins & Scott, 1983;
polythene water pipes, and surfaces of larval tanks are susceptible Sanders, Boothe, Frank, & Arnold, 2007). Many researchers have
to biofilm formation (Karunasagar, Otta, & Karunasagar, 1996). been trying to understand the behavior of Campylobacter jejuni in
In a study by Guðbjörnsdóttir et al. (2005), it was shown that poultry processing. As a result, it has been determined that
adhered bacteria are found in many locations on the seafood pro- temperature is a significant factor influencing their survival (Chan,
cessing lines, despite the fact that thorough cleaning and disin- Tran, Kanenaka, & Kathariou, 2001; Dykes et al., 2003; Trachoo,
fection are carried out regularly. Gram-negative rods, namely Frank, & Stern, 2002). Other authors have stated that wild-type
Pseudomonas spp., Aeromonas spp., Enterobacteriaceae, and yeast, cultures of C. jejuni planktonic survive longer at lower tempera-
were isolated from a shrimp processing plant, while Pseudomonas tures than higher temperatures when faced with multiple stressors
spp. and Enterobacteriaceae were found mainly in the fish pro- (Chan et al., 2001; Dykes et al., 2003). In contrast, Hanning, Jarquin,
cessing plant. Pseudomonas putida and Pseudomonas fluorescens and Slavik (2008) showed that culturable C. jejuni can endure
were the main species of Pseudomonas spp. isolated from the longer in biofilms at 32  C, compared with survival of culturable
shrimp factories. These organisms are considered as the causes of planktonic cells and biofilms at 10  C. The results of the same study
spoilage in fresh or chilled fish (Gram and Huss 1996). More demonstrated that the attachment of C. jejuni to surfaces is facili-
importantly, it was noted that the presence of Pseudomonas spp. tated by a pre-existing biofilm so it is important to strengthen the
would significantly enhance the colonization of L. monocytogenes control of biofilms in poultry processing.
on stainless steel (Guðbjörnsdóttir et al., 2005). It was seen that
Vibrio spp. are not the only genus found in seafood processing 4.5. Meat industry
plants. Other genera such as Pseudomonas spp., Aeromonas spp., and
so on are also found to be significant biofilm producers and their Organic residues in food processing could be a niche for micro-
presence would enhance the biofilm formation of other genus. In organism accumulation and biofilm formation as it is a source of
another study on the correlation of the biofilm forming abilities of cross-contamination, and has become quite a concern for numerous
Salmonella and its persistence in fish meal- and feed factories, researchers (Brooks & Flint, 2008; McLandsborough, Rodriguez,
Salmonella agona and Salmonella montevideo were found to be good Pérez-Conesa, & Weiss, 2006; Simões, Simões, & Vieira, 2010).
biofilm producers, while Salmonella typhimurium was found to be Many studies have been conducted in order to gain a deeper
a poor biofilm producer. The results of the study also indicated that understanding of biofilms. For example, E. coli has been studied by
the persistence of Salmonella in the factory environment depends many researchers to investigate the “attachment and biofilm
significantly on the strain’s biofilm forming ability (Vestby, formation by E. coli O157:H7 at different temperatures, on various
Møretrø, Langsrud, Heir, & Nesse, 2009). However, it also indi- food-contact surfaces encountered in beef processing” (Dourou
cated that the level of biofilm formation can be affected positively et al., 2011). In the results from the study, the cells attached to the
or negatively by environmental factors, such as natural microflora surface increased over time even under low temperatures, which
(Carpentier & Chassaing, 2004; Guðbjörnsdóttir et al., 2005). could result from the migration of cells toward the surface by
flagellar-mediated motility and Brownian motion (Van Houdt &
4.4. Poultry industry Michiels, 2005). It has been seen that at low temperatures, micro-
organisms can adhere and survive on food contact surfaces, and
Many studies have been carried out on the biofilm formation in even increase in population as time progressed (Dourou et al., 2011;
the poultry processing industry. Under many investigations, it has Kim, Ryu, & Beuchat, 2006). Another important factor influencing
been identified that dust, surfaces, feces, poultry feed, and trans- the attachment of E. coli O157:H7 is the presence of other micro-
portation of live poultry between production and processing units organisms on the surfaces (Castonguay et al., 2006; Klayman,
are known to be the important risk factors in Salmonella contami- Volden, Stewart, & Camper, 2009; Marouani-Gadri, Augier, &
nation (Marin, Hernandiz, & Lainez, 2009; Park, Jarquin, Hanning, Carpentier, 2009). As an example, E. coli O157:H7 was found to be
Almeida, & Ricke, 2011; Ramesh, Joseph, Carr, Douglass, & incapable of forming a biofilm under dynamic-flow conditions due
Wheaton, 2002). Furthermore, approximately 50% of the strains to the shear forces, while Acinetobacter calcoaceticusdisolated from
isolated on poultry farms were able to produce biofilms (Marin meat-processing plantsdmonospecies biofilms are heterogeneous,
et al., 2009). The attachment of 25 Salmonella strains to four well organized, and can develop under both static and dynamic
different materials (polytetrafluoroethylene-also known as Teflon, conditions. The research of Habimana, Heir, Langsrud, Asli, and
stainless steel, rubber, and polyurethane), which are commonly Møretrø (2010) indicated that E. coli O157:H7 cells were
used in poultry industry, was studied. Among those, Salmonella embedded and cover by a Acinetobacter calcoaceticus biofilm under
sofia isolates (except S1635 and S1636) can generally adhere in both static and dynamic growth conditions. It is now well docu-
higher numbers to the different surfaces than the other isolates. It mented that multispecies biofilms may increase the opportunities
was suggested that S. Sofia serovar may have more pili or fimbriae for pathogens to thrive in the food industry (Burmølle et al., 2006;
than others (Chia et al., 2009). The results of the study also agree Habimana et al., 2010; Stewart & Franklin, 2008).
with the data from Helke et al. (1993) that S. typhimurium can
attach more readily to stainless steel than rubber. In a different 4.6. Ready-to-eat (RTE) foods industry
study, the characterization of biofilm formation and the attachment
of S. typhimurium DT104 (Kim & Wei, 2009) was evaluated, with the Due to lifestyle changes, RTE foods have become very popular.
results indicating that the main contributor to lipopolysaccharide However, RTE foods can be considered as a relatively high risk food
(LPS) synthesis and biofilm formation of S. typhimurium DT104 is since the products will be consumed directly without undergoing
rfbA gene. According to the study, many factors, such as production any bactericidal process. Even though RTE foods have been well
of EPS and their efficient transportation through outer membranes, processed, the chances of being contaminated are relatively high.
expression of flagella and regulation of exoribonucleases and RNA- Furthermore, the storage times and conditions are known to
binding protein, were suggested to be involved in biofilm formation be important factors affecting RTE foods quality (Sofos & Geornaras,
and the attachment of S. typhimurium DT104 on contact surfaces. 2010). RTE foods could potentially be susceptible to cross-
578 S. Srey et al. / Food Control 31 (2013) 572e585

contamination during processing and handling. In particular, after prevent the contamination of food products. A good cleaning
cooking, the products may be recontaminated during loading, process that removes any food residues and other compounds that
conveying, weighing, and packaging (Osaili, Alaboudi, & Nesiar, may promote bacteria proliferation and biofilm formation is
2011). Garrido et al. (2009) evaluated the occurrence and levels of particularly effective (Simões et al. 2010). Many different chemical
L. monocytogenes in refrigerated RTE products for prolonged products may be used in cleaning, including surfactants or alkali
consumption in northern Spain. It was found that RTE smoked fish products, or used to suspend and dissolve food debris by decreasing
was the most common pathogen-contaminated food among the surface tension, emulsifying fats, and denaturing proteins (Forsythe
samples analyzed. The slicing materials used in processing were & Hayes, 1998; Maukonen et al., 2003). Cleaning should be carried
speculated to be the source of contamination due to biofilm out in a way that can break-up or dissolve the EPS matrix associated
formation. Comparably, smoked salmon was found to be the most with the biofilms so that disinfectants can gain access to the
L. monocytogenes contaminated product in the study by Di Pinto, bacteria cells (Simões et al. 2006). It is evident that the use of high
Novello, Montemurro, Bonerba, and Tantillo (2010). According to temperatures in cleaning can reduce the physical force required,
a survey by Wagner, Auer, Trittremmel, Hein, and Schoder (2007), such as water turbulence and scrubbing (Maukonen et al., 2003); as
fish and seafood were also found to be highly contaminated with well as inactivate biofilm cells at a certain level (Chmielewski &
L. monocytogenes among the samples analyzed. The survey also Frank, 2006). Besides, cleaning only allows the removal of
shows that there is more chance of contamination for unpackaged approximately 90% of the bacteria from the surfaces and does not
or repackaged RTE foods; furthermore, raw meat sausages were kill them. They might later re-attach to other surfaces and form
postulated to be potentially contaminated. In addition, the a biofilm, thus disinfection is indispensible with the intention of
production of biofilms and quorum sensing by E. coli O157:H7 and eliminating them (Gram, Bagge-Ravn, Ng, Gymoese, & Vogel, 2007).
its transfer from contact surfaces to meat, poultry, RTE deli prod- Antimicrobial agents are used in the disinfection process so as to
ucts, and produce products was studied. As a result, the E. coli kill microorganisms so that the surface population is reduced along
O157:H7 strain could be a concern in a wide variety of food prod- with microbial growth on the surfaces. However, the effectiveness
ucts since it can form biofilms on food contact surfaces during food of disinfectants is limited by the presence of organic material
processing (Silagyi, Kim, Lo, & Wei, 2009). including fat, carbohydrates, and protein-based materials. Other
than that, pH, temperature, water hardness, chemical inhibitors,
5. Biofilm control strategies concentration, and contact time are also important factors influ-
encing disinfectants’ effectiveness (Bremer, Monk, & Butler, 2002;
Since biofilms are a great concern in the food sectors, many Cloete, Jacobs, & Brözel, 1998; Kuda, Yano, & Kuda, 2008). There are
studies have been done in order to gain a better understanding of many types of disinfectants including chlorine, hydrogen peroxide,
their development and spread. Consequently, many studies have iodine, ozone, peracetic acid (Chmielewski & Frank, 2007).
also come up with countermeasures. The first and most important
thing to do is to prevent biofilm formation by regularly cleaning 5.2. Clean-in-Place (CIP)
and disinfecting so as to not allow the cells to firmly attach
(reversible attachment) to contact surfaces (Midelet & Carpentier, Clean-in-Place (CIP) is a process allowing a complete system to
2004; Simões, Simões, Machado, Pereira, & Vieira, 2006). Meyer be cleaned without dismantling it or the manual involvement of
(2003) suggested three different strategies: (i) disinfection “in the operator. It includes jetting and spraying the surfaces or the
time”, before biofilm develops, (ii) disinfection of biofilms using circulation of cleaning solutions throughout the plant with an
harsh disinfectants, and (iii) inhibition of attachment of microbes increased turbulence and flow velocity (Romney, 1990). There are
by selecting surface materials that do not promote attachment or many factors that can influence CIP efficacy, including the nature
by supplementing with nutrients. Many other researchers have of the biofilm layer, cleaning chemical composition and concen-
accounted for the incorporation of antimicrobial products in the tration, time (Boulange-Petermann, Jullien, Dubois, Benezech, &
surface materials themselves (Knetsch & Koole, 2011; Park, Faille, 2004; Changani, Belmar-Beiny, & Fryer, 1997), cleaning
Daeschel, & Zhao, 2004) by coating surfaces with antimicrobials temperature (Lelièvre, Faile, & Bénézech, 2001), cleaning flow rate
(Knetsch & Koole, 2011; Thouvenin et al., 2003) or by modifying the and hydrodynamic (Lelièvre, Antonini, Faille, & Bénézech, 2002),
surfaces’ physiochemical properties (Chandra et al., 2005; and the cleaning surface characteristics (Lelièvre, Legentilhomme,
Rosmaninho et al., 2007). In a study on biofilm control, micropar- et al., 2002). Relatively, Walton (2008) also summarized the basic
ticles (CaCO3) coated with benzyldimethyldodecylammonium principles of cleaning to (i) consider the physical nature and
chloride were found to effectively inactivate biofilm formation construction of the equipment to be cleaned, (ii) assess the nature
(Ferreira, Pereira, & Pereira, 2011). Many others reported biofilm of the soil to be removed; (iii) select a detergent appropriate to
formation was inhibited by silver coating surfaces (Hashimoto, the removal of that soil, (iv) bring the soil and the detergent
2001; Knetsch & Koole, 2011). Pre-conditioning the surface with together (at the right temperature, under the right conditions of
a surfactant has also been reported to prevent bacterial adhesion flow and turbulence, at the right chemical concentration, for the
(Chen, 2012; Choi, Park, Lee, Park, & Kim, 2011). The research of right period of time), (v) rinse away all traces of detergent and
Zeraik and Nitschke (2010) demonstrated that after conditioning soil, with the objective of achieving the standard of cleanliness
with surfactant, the surface became more hydrophilic. The data appropriate to the duty for which the equipment is destined to be
illustrated the decrease in hydrophobicity on the treated surfaces used, (vi) always undertake cleaning as soon as possible after
and showed a significant decrease in bacterial attachment. completion of the production operation, and (vii) when necessary,
However, other factors are still considered to contribute to the undertake a disinfection or sterilization process immediately
reduction of bacterial attachment. before the equipment is returned to processing or production
duties in order to reduce the level of microbiological contamina-
5.1. Cleaning and disinfection tion to one consistent with the hygienic standard required for that
duty.
In the food industry, there is debris everywhere which would Bremer, Fillery, and McQuillan (2006) mimicked the CIP of dairy
promote the accumulation of microorganisms and encourage bio- biofilms on a laboratory scale to study the effectiveness of the
film formation. Therefore, regular cleaning is required so as to different caustic and acid wash steps. According to the results, it
S. Srey et al. / Food Control 31 (2013) 572e585 579

was postulated that the efficacy of the cleaning conditions con- 5.3.2. Hydrogen peroxide (H2O2)
ducted may change according to the amount of soiling and cleaning H2O2 is one of the widely used disinfectants due to its highly
cycles, as well as the fact that the removal of bacterial biofilms on oxidizing capacity based on the production of free radicals which
surfaces in a dairy manufacturing plant can be improved by using affect the biofilm matrix and has been found to be efficient against
caustic and nitric additives. In another study, the life cycle of four biofilms (de Carvalho, 2007; de Carvalho & da Fonseca, 2007).
CIP methods (conventional alkaline/acid cleaning with hot water Moreover, many studies have shown its efficiency as a disinfectant
disinfection, one-phase alkaline cleaning with acid chemical against biofilms (Christina, Penna, Mazzola, Maria, & Martins, 2001;
disinfection, enzyme-base cleaning with acid chemical disinfection Toté et al., 2010). From the perspective of safety, H2O2 is known to be
and the conventional method with disinfection by cold nitric acid at a safe solution which does not cause allergic reactions (Rideout,
pH 2) was evaluated. It was found that the CIP methods with small Teschke, Dimich-Ward, & Kennedy, 2005). Moreover, Kim, Silva,
volumes and low temperatures, such as enzyme-based cleaning Chamul, and Chen (2000) showed that H2O2 can be used at a high
and one-phase alkaline cleaning, were the most highly recom- concentration without negatively affecting the product quality.
mended alternative methods (Eide, Homleid, & Mattsson, 2003). H2O2 was used against four strains of Vibrio spp. in seawater. It
The study on biofilm removal of bacterial isolates sampled in the was found to be very effective in inhibiting biofilm formation at
food industry by enzymes proposed that the implementation of a concentration of 0.05% (500 mg/l). It can also kill mature biofilms
enzymatic control of bacterial biofilms in the food industry would at concentrations between 0.08% and 0.2% (Shikongo-Nambabi
present a noteworthy alternative, while the conventional CIP using et al., 2010). In this study, the bacteria killing efficiency was
chemical agents is not providing satisfactory hygienic results higher than the previous study of Kim et al. (2000). It is postulated
(Lequette, Boels, Clarisse, & Faille, 2010). that the different mineral ions present in seawater are essential in
the enhancement of H2O2 reaction against microorganism
5.3. Chemical-based control (Shikongo-Nambabi et al., 2010). Likewise, it was reported to
completely eradicate biofilms at a concentration of 5% with
In the study on the effect of mechanical stress on biofilms a 15 min exposure time (Robbins, Fisher, Moltz, & Martin, 2005).
challenged by different chemicals, it was stated that chemical
agents would react with the EPS complex which would enhance the 5.3.3. Ozone
mechanical biofilm removal. The removal rate was significantly Ozone, a result of oxygen atoms exposed to high-voltage electric
improved after treating the biofilm with chemical agents (Simões, discharge, is a bluish gas with strong odor and potential oxidizing
Pereira, & Vieira, 2005). However, in another study, bacterial cells properties (Horvath, Bilitzky, & Hüttner, 1985, p. 350). It is a potent
were destroyed after being subjected to chemical agents, while the antimicrobial agent which can be used against bacteria, fungi,
EPS matrix was left unaffected. The best result was attained by viruses, protozoa, and bacterial and fungal spores (Khardre, Yousef,
applying both chemical and mechanical treatment (Exner, & Kim, 2001). The microorganisms are eradicated by the disruption
Tuschewitzki, & Scharnagel, 1987). Accordingly, it was suggested or breakdown of the cell envelope, which in turn leads to the
that mechanical treatment cannot remove bacterial cells (Jessen & leakage of the cell contents. Cell lysis is a faster inactivation
Lammert, 2003). As such, it can be postulated that chemical and mechanism than that of other antimicrobial agents where perme-
mechanical treatment has a synergistic effect and both play ation through the cell membrane is indispensable in order to
important roles in biofilm and bacterial cell removal. effectively inactivate the microbe. Due to its mechanism, it is
speculated that it cannot lead to microorganism resistance (Pascual,
5.3.1. Sodium hypochlorite Llorca, & Canut, 2007). Many researchers have demonstrated the
Sodium Hypochlorite (NaClO) is a chemical compound used for efficiency of ozone against bacterial cells and biofilms (Dosti, Guzel-
bleaching or disinfection and as such it has been used for dis- Seydim, & Greene, 2005; Lagrange, Reiprich, & Hoffmann, 2004). A
infecting surfaces. It was reported to be an effective disinfectant for study by Tachikawa, Yamanaka, and Nakamuro (2009) on the
biofilm inactivation (Ozdemir, Buzoglu, Calt, Stabholz, & Steinberg, disinfection and removal of biofilms by ozone water on P. fluorescens
2010; da Silva et al., 2011). However, it is known to be more and P. aeruginosa biofilms showed that by forming biofilms, the
effective in low pH than alkaline pH environments (Araújo, Lemos, resistibility of the microorganisms against ozone was increased by
Mergulhão, Melo, & Simões, 2011). NaClO was reported to be 3000 and 10 times, respectively. The reason behind the resistance
a potential biofilm antimicrobial agent against Staphylococcus could be the reaction between ozone and the biofilm matrix
aureus (Toté, Horemans, Vanden Berghe, Maes, & Cos, 2010), introduced into the environment by the bacteria. However, the
Prevotella intermedia, Peptostreptococcus miros, Streptococcus inter- surviving cells were found to be less than 1% after being treated
medius, Fusobacterium nucleatum, and Enterococcus faecalis when with ozone.
compared to other disinfectants used in a study by Spratt, Pratten,
Wilson, and Gulabivala (2001). In a study by Lomander, Schreuders, 5.3.4. Peracetic acid
Russek-Cohen, and Ali (2004), 50 ppm sodium hypochlorite solu- Peracetic acid is a result of the reaction between hydrogen
́
tion was able to significantly eliminate biofilm cells compared to peroxide and acetic acid, or by the oxidation of acethaldehyde. The
rinsing in water. In the comparison on detaching biofilm removal, it mixture has a strong odor and a low pH (2.8) and is usually
was not more effective than water in detaching biofilms. Six produced in concentrations between 5 and 15%. It has been used in
percent NaClO was also reported to be able to kill a significant water purification and as a disinfectant (Anonymous, n.d.-a). It is
amount of E. faecalis biofilms, which is statistically better than other known as an ideal antimicrobial agent according to its extreme
tested agents. The efficiency of NaClO was tested in the elimination oxidizing capacity. Furthermore, it cannot be deactivated by cata-
of L. monocytogenes, Pseudomonas fragi and Staphylococcus xylosus lase and peroxidasedenzymes that degrade H2O2. This agent also
(Norwood & Gilmour, 2000). Consequently, all planktonic cells decomposes into safe and environmental friendly residues in food
were eliminated with an exposure to 10 ppm free chlorine for 30 s, (acetic acid and hydrogen peroxide), hence it can be applied
while it can reduce only 2 log of a L. monocytogenes biofilm with without rinsing and its efficacy is not affected by protein residues
a concentration of 1000 ppm with an exposure time of 20 min. It (Anonymous, n.d.-b). A study showed that peracetic acid can reduce
was speculated that the co-cultured biofilm may enhance its L. monocytogenes biofilm adhered for 24 h by 5 log with a concen-
resistance to disinfectants. tration of 0.50% w/v (Cabeça, Pizzolitto, & Pizzolitto, 2008).
580 S. Srey et al. / Food Control 31 (2013) 572e585

According to Frank, Ehlers, and Wicker (2003), peracetic acid can (Carmen et al., 2004). Baumann, Martin, and Feng (2009) also
reduce more than 6 log of L. monocytogenes biofilms on stainless showed a significant effect on biofilm removal on stainless steel
steel in the presence of fat and protein soil with a concentration of food contact surfaces by combining the use of ozonation and
2.0 ml/l and an exposure time of 10 min. Others studies also sonication.
showed the effectiveness of peracetic acid against various micro-
organisms (Marques et al., 2007; Salvia, Teodoro, Balducci, Koga- 5.4.2. Enzymes
Ito, & Oliveira, 2011). According to Toté et al. (2010), peracetic Enzymes are proteins with catalytic activity on a specific
acid eliminated approximately 98% and 99% of viable S. aureus and chemical molecule. It can be an important alternative for biofilm
P. aeruginosa, respectively, with only 1 min of contact time but not removal in the food industry. However, EPS is a heterogenic matrix
the biofilm matrix. However, there are also several other studies so, in order to degrade the complex, a combination of enzymes is
which showed that peracetic acid is inefficient or less effective than required (Simões et al. 2010). Lequette et al. (2010) showed that the
other disinfectants against biofilms (Królasik, Zakowska, Krepska, & enzymes efficiency on biofilm removal may vary according to the
Klimek, 2010; Rossoni & Gaylarde, 2000). It is suggested that species of bacteria and it can also be enhanced by combining with
aldehydes do not degrade the biofilm matrix, and instead improve surfactants; this suggests that proteins also contribute to the
the stability. The biofilm complex needs to be eradicated before adhesion of biofilms as proposed by Hinsa and O’Toole (2006). The
chemical agents can be used effectively (Exner et al., 1987). research of Molobela, Cloete, and Beukes (2010) indicated that
protease enzymes were effective in the degradation of P. fluorescens
5.4. Other approaches of biofilm control biofilm’s EPS, while amylase enzymes were less effective. It was
suggested that the structural composition of EPS varies even among
The conventional control strategies are chemical-based, bacteria of the same species, with the way they were formulated
however, it is possible that microorganisms hold a certain degree and their mode of action, are the reasons for the enzymes ineffi-
of resistance to such strategies, or may acquire it later through ciency. It is well known that a mixture of proteases and amylases is
mutation or genetic exchange. These processes enable the bacteria commonly used to respond to the variety present in a single
to survive and proliferate under higher concentration of disinfec- biofilm. While proteases hydrolyze the proteins, amylases break the
tants (Gilbert & Mcbain, 2003; McBain, Rickard, & Gilbert, 2002). bond of carbohydrates associated with the complex. Among all
Therefore, new approaches to controlling biofilms in the food 1,4-glycosidic bond cleaving amylases, a-Amylases are frequently
industry have been introduced. Simões et al. (2010) reviewed used because of their thermostability; however, they will not
potential green strategies, including control using enzyme, phage, stay active for long as they are calcium metalloenzymes
and microbial interactions and metabolite molecules. (Craigen, Dashiff, & Kadouri, 2011). Similarly, a combination of
polysaccharide-hydrolyzing enzymes and oxidoreductases were
5.4.1. Ultrasonication recommended for bacteria biofilm removal due to the wide range of
Ultrasonication is a well known technique used in various food polysaccharide-hydrolyzing enzymes activities which make it
industry processes namely freezing, cutting, drying, tempering, useful for the degradation of biofilms matrix; the bactericidal effect
bleaching, sterilization, and extraction (Chemat, Zill-e-Huma, & of oxidoreductases (Johansen, Falholt, & Gram, 1997). In another
Khan, 2011). It was reported to also be used as an efficient biofilm study, individual biofilm cleaning efficiencies of pectin esterase,
removal method (Oulahal-Lagsir, Martial-Gros, Boistier, Blum, & pectin lyase, and cellulose were shown to be not as effective against
Bonneau, 2000a). Oulahal-Lagsir, Martial-Gros, Bonneau, et al. P. fluorescens mature biofilms. However, the efficacy was enhanced
(2000b) investigated the use of an ultrasonic apparatus on biofilm when pronase was used in the treatment process, and after being
removal from stainless steel and polypropylene surfaces. The treated with the aforementioned enzymes and the detachment-
apparatus was demonstrated to be able to remove twice as much of promoting-agent, pronase, to simultaneously treat with others
the industrial milk biofilm as the swabbing method on poly- enzymes (Orgaz, Neufeld, & Sanjose, 2007). As can be seen, enzy-
propylene sheets. However, it has been documented that even matic control against biofilms could be used as a new and improved
though lower-frequency in sonation is remarkably more efficient environmental friendly alternative strategy according to its
than higher frequency for reducing biofilm cells viability, bacteria nontoxic characteristics and instability.
in food industries cannot be solely eliminated using the present
ultrasonic technologies thus combining techniques of ultrasound 5.4.3. Phages
with other treatment techniques were recommended (Piyasena, The first application of phages was in the early 20th century for
Mohareb, & McKellar, 2003; Qian, Sagers, & Piti, 1997). Accord- bacterial infections treatment in Eastern Europe, and have been
ingly, the combination of ultrasound and ethylenediaminetetra- shown to decrease biofilm formation (Curtin & Donlan, 2006;
acetic acid (EDTA), and ultrasound and enzymes showed a higher Merril, Scholl, & Adhya, 2003). Unlike chemical-based antimicrobial
efficacy in removing biofilms. The results were more promising agents that can cause corrosion, phages can be a suitable substitute
against S. aureus than E. coli biofilms in the study. The authors (Goldman, Starosvetsky, & Armon, 2009), thus it has also been
stated that this method was in agreement with an industrial control proposed as a biofilm control method (Curtin & Donlan, 2006).
method, i.e. a combined treatment of ultrasound generation in Bacteriophage spray treatment was suggested to be an alternative
enzymes preparation restricted to an active chamber area with to dipping, brushing, or sponging of the spinach harvester blade on
a fast and good reproducible recovery compared to other a chlorine solution according to its compatibility with harvest
approaches (Oulahal, Martial-Gros, Bonneau, & Blum, 2007). sanitation practices (Patel, Sharma, Millner, Calaway, & Singh,
Ultrasound was also reported to increase the effectiveness of 2011). It was documented that phage components and their
antibiotics against biofilm cells (Peterson & Pitt, 2000). Two assembly synthesis vary according to the host bacterial growth rate
processes were observed to be account for the increased efficiency: and their amount of protein-synthesizing during the time of
i), ultrasound improves the diffusion of oxygen into the biofilm infection; however, it was also reported that even under a glucose-
matrix which allows biofilm cells to become active, and therefore, limited chemostat, T4 phage can effect E. coli biofilms (Corbin,
affected by antibiotics and ii), the high ratedenhanced by McLean, & Aron, 2001). In a study on the effect of the phage
ultrasounddof antibiotics transported into the complex may philBB-PF7A on P. fluorescens biofilms, the important role of
destroy the bacteria before they gain resistance to the agents convection mechanism was discussed. It was said that biofilm cells
S. Srey et al. / Food Control 31 (2013) 572e585 581

lysis is more efficient under static conditions than dynamic when a bacteriophage is used with an antibiotic (Verma, Harjai, &
conditions. It was also found that fIBB-PF7A could be a remarkable Chhibber, 2010). Hurdle technology is a very promising new
biological agent according to its biofilm cells lysing capability in approach in controlling biofilms in the food industry.
a markedly rapid time (Sillankorva, Neubauer, & Azeredo, 2008).
The same phage was also used to control the dual species biofilm of
P. fluorescens and Staphylococcus lentus, and accounted for the 6. Conclusion
dramatic decrease in the target bacteria cells (P. fluorescens).
Surprisingly, it was proved that phages can be used to efficiently Pathogenic microorganisms in biofilms formed in different food
reach and lyse their target bacterium in both single and dual industries settings are a source of food contamination. As the
species biofilms notwithstanding the presence of a non-susceptible demand for fresh, RTE and processed foods increases, many studies
host (Sillankorva, Neubauer, & Azeredo, 2010). are needed to address biofilm removal and disinfectant efficacy in
Cerca, Oliveira, and Azeredo (2007) studied the susceptibility of food industries. Even though up to now the conventional control
S. epidermidis planktonic cells and biofilms according to the lytic strategies are still used and still being developed, a more
action of staphylococcus bacteriophage K. The phage K lysis effi- economical and environmental friendly control strategy is indis-
ciency depends upon the bacteria growth phase. This was also pensible to satisfy the need of industrial food safety. It should be
found to be true in the research of Sillankorva, Oliveira, Vieira, taken into account that disinfection method shall provide a desir-
Sutherland, and Azeredo (2004). Fluorescence Correlation Spec- able cost-effective result and not causing any adverse effect on
troscopy was used to study the diffusion and reaction of bacterio- human health as well as the environment. Therefore, we highly
phages inside biofilms; the results indicated that bacteriophages recommended hurdle technology since it gives synergistic effect
can infiltrate different biofilm complexes, and that generally, they which reduces the materials and energy consumption. Moreover,
are immobilized, amplified, and released by a lytic cycle in the inhibiting biofilms and quorum sensing by natural antimicrobials
biofilm and interact with their specific binding sites on the hosts would also be an alternative to combat the biofilms problems.
although the lytic activity was not observed (Briandet et al., 2008).
According to Tait, Skillman, and Sutherland (2002), phages and
References
bacteria can steadily co-exist in biofilms, thus a mixture of phages
and polysaccharide depolymerases and disinfectant was suggested Aarnisalo, K., Lundén, J., Korkeala, H., & Wirtanen, G. (2007). Susceptibility of Listeria
for a better biofilm control. To address this challenge, bacterio- monocytogenes strains to disinfectants and chlorinated alkaline cleaners at cold
temperatures. LWT - Food Science and Technology, 40(6), 1041e1048.
phages can be engineered to be able to express a biofilm-degrading
Abdallah, F. B., Chaieb, K., Zmantar, T., Kallel, H., & Bakhrouf, A. (2009). Adherence
enzyme, which would be a good asset in overcoming the challenges assays and slime production of Vibrio alginolyticus and Vibrio parahaemolyticus.
in controlling biofilms. The engineered phages were reported to be Brazilian Journal of Microbiology, 394e398.
able to noticeably eradicate bacterial cells in biofilms (approxi- Adetunji, V. O., & Isola, T. O. (2011). Crystal violet binding assay for assessment of
biofilm formation by Listeria monocytogenes and Listeria spp on wood, steel and
mately 99.997% removal), as well as the biofilm complex (Lu & glass surfaces. Global Veterinarian, 6(1), 6e10.
Collins, 2007). Alliance For Food and Farming. (2010). Analysis of produce related foodborne illness
outbreaks. Retrieved 21.11.12, from. http://www.foodandfarming.info/docs/
386Produce_Analysis_2010_Final.pdf.
5.5. Hurdle technology Anonymous. (2011). Raw milk cheddar recalled due to Listeria concerns. Food Safety
News, Retrieved 26.04.12, from. http://www.foodsafetynews.com/2011/11/raw-
Hurdle technology is a combination of two or more different milk-cheddar-recalled-due-to-listeria-concerns/.
Anonymous. (n.d.-a). Peracetic acid as a disinfectant. Retrieved 02.05.12, from http://
control techniques which have been proven to be effective. www.lenntech.com/processes/disinfection/chemical/disinfectants-peracetic-
However, in order to achieve an effective treatment, the right acid.htm.
combination is needed. It was shown that the combination treat- Anonymous. (n.d.-b). Peracetic acid. Retrieved 03.05.12, from http://www.tmsplc.
com/news5009/news-details.htm.
ment of NaClO with UV irradiation had a better reduction of food-
Anwar, H., Dasgupta, M. K., & Costerton, J. W. (1990). Testing the susceptibility of
borne pathogens in food than single treatment (Ha & Ha, 2011). bacteria in biofilms to antibacterial agents. Antimicrobial Agents and
DeQueiroz and Day (2007) studied the antimicrobial activity and Chemotherapy, 34(11), 2043e2046.
Araújo, P., Lemos, M., Mergulhão, F., Melo, L., & Simões, M. (2011). Antimicrobial
effectiveness of a combination of NaClO and hydrogen peroxide
resistance to disinfectants in biofilms. Science Against Microbial Pathogens:
(H2O2) in killing and removing P. aeruginosa biofilms from surfaces. Communicating Current Research and Technological Advances, 826e834.
It showed an increased reduction in the number of cells with only Arnold, J. W., Boothe, D. H., Suzuki, O., & Bailey, G. W. (2004). Multiple imaging
a short exposure time. The synergistic effect of a combined treat- techniques demonstrate the manipulation of. Journal of Microscopy, 216(3),
215e221.
ment of biofilms with H2O2 and UV was also shown to be more Augustin, M., Ali-Vehmas, T., & Atroshi, F. (2004). Assessment of enzymatic cleaning
effective than the single use of H2O2 by 10 fold, which could agents and disinfectants against bacterial biofilms. Journal of Pharmacy &
contribute to a more environmental-friendly treatment Pharmaceutical sciences: a Publication of the Canadian Society for Pharmaceutical
Sciences, Société canadienne des sciences pharmaceutiques, 7(1), 55e64.
(Vankerckhoven et al., 2011). Schurman, Sumner, and Marcy (2001) Baumann, A. R., Martin, S. E., & Feng, H. (2009). Removal of Listeria monocytogenes
tested the effectiveness of H2O2 and demonstrated that its efficacy biofilms from stainless steel by use of ultrasound and ozone. Journal of food
was enhanced when there was an increase in temperature. More- Protection, 72(6), 1306e1309.
Borucki, M. K., Peppin, J. D., & White, D. (2003). Variation in biofilm formation
over, an increase in organic acid concentration could also increase among strains of Listeria monocytogenes. Applied and Environmental Microbi-
its efficiency. Additionally, the synergistic effect of ozone and ology, 69(12), 7336e7342.
ultrasound was also shown to be efficient for biofilm cell reduction. Boulange-Petermann, L., Jullien, C., Dubois, P. E., Benezech, T., & Faille, C. (2004).
Influence of surface chemistry on the hygienic status of industrial stainless
However, the amount of cells eradicated was only slightly more steel. Biofouling, 20(1), 25e33.
compared to that when ozone was used alone. It was suggested that Bower, C. K., McGuire, J., & Daeschel, M. A. (1996). The adhesion and detachment of
the energy efficiency of the combined process shall be studied in bacteria and spores on food-contact surfaces. Trends in Food Science &
Technology, 7(5), 152e157.
order to have a cost-effective treatment system (Patil, 2010).
Bremer, P. J., Fillery, S., & McQuillan, A. J. (2006). Laboratory scale Clean-In-Place
Moreover, the efficiency of antibiotics alone and synergizing with (CIP) studies on the effectiveness of different caustic and acid wash steps on
a lytic bacteriophage in the removal of old Klebsiella pneumoniae the removal of dairy biofilms. International Journal of Food Microbiology, 106(3),
biofilms was examined. The findings revealed that phages can be 254e262.
Bremer, P. J., Monk, I., & Butler, R. (2002). Inactivation of Listeria monocytogenes/
surprisingly capable in the removal of older biofilms on account of Flavobacterium spp. biofilms using chlorine: impact of substrate, pH, time and
depolymerase. Then again, the eradication of biofilms was improved concentration. Letters in Applied Microbiology, 35(4), 321e325.
582 S. Srey et al. / Food Control 31 (2013) 572e585

Briandet, R., Lacroix-Gueu, P., Renault, M., Lecart, S., Meylheuc, T., Bidnenko, E., et al. Corbin, B. D., McLean, R. J. C., & Aron, G. M. (2001). Bacteriophage T4 multiplication
(2008). Fluorescence correlation spectroscopy to study diffusion and reaction of in a glucose-limited Escherichia coli biofilm. Canadian Journal of Microbiology,
bacteriophages inside biofilms. Applied and Environmental Microbiology, 74(7), 47(7), 680e684.
2135e2143. Costerton, J. W., Lewandowski, Z., Debeer, D., Caldwell, D., & Korber, D. (1994).
Brooks, J. D., & Flint, S. H. (2008). Biofilms in the food industry: problems and Biofilms, the customized microniche. Journal of Bacteriology, 176(8), 2137e2142.
potential solutions. International Journal of Food Science & Technology, 43(12), Costerton, J. W., & Stewart, P. S. (2001). Battling biofilms. Scientific American, 285(1),
2163e2176. 74e81.
Burmølle, M., Webb, J. S., Rao, D., Hansen, L. H., Sørensen, S. J., & Kjelleberg, S. Costerton, J. W., Stewart, P. S., & Greenberg, E. P. (1999). Bacterial biofilms:
(2006). Enhanced biofilm formation and increased resistance to antimicrobial a common cause of persistent infections. Science, 284(5418), 1318e1322.
agents and bacterial invasion are caused by synergistic interactions in multi- Craigen, B., Dashiff, A., & Kadouri, D. E. (2011). The use of commercially available
species biofilms. Applied and Environmental Microbiology, 72(6), 3916e3923. alpha-amylase compounds to inhibit and remove Staphylococcus aureus bio-
Cabeça, T. K., Pizzolitto, A. C., & Pizzolitto, E. L. (2008). Assessment of action of films. The Open Microbiology Journal, 5, 21e31.
disinfectants against Listeria monocytogenes biofilms. Journal of Food and Curtin, J. J., & Donlan, R. M. (2006). Using bacteriophages to reduce formation of
Nutrition, 17(2), 121e125. catheter-associated biofilms by Staphylococcus epidermidis. Antimicrobial Agents
Carmen, J. C., Nelson, J. L., Beckstead, B. L., Runyan, C. M., Robison, R. a, and Chemotherapy, 50(4), 1268e1275.
Schaalje, G. B., et al. (2004). Ultrasonic-enhanced gentamicin transport through Davey, M. E., & O’Toole, G. A. (2000). Microbial biofilms: from ecology to molecular
colony biofilms of Pseudomonas aeruginosa and Escherichia coli. Journal of genetics. Microbiology and Molecular Biology Reviews: MMBR, 64(4), 847e867.
Infection and Chemotherapy: Official Journal of the Japan Society of Chemotherapy, Deming, M. S., Tauxe, R. V., Blake, P. A., Dixon, S. E., Fowler, B. S., Jones, T. S., et al.
10(4), 193e199. (1987). Campylobacter enteritis at a university: transmission from eating
Caron, C. (2011). CDC: Cantaloupe Listeria outbreak deadliest in a decade e ABC News. chicken and from cats. American Journal of Epidemiology, 126(3), 526e534,
Retrieved 21.11.12, from. http://abcnews.go.com/Health/cdc-cantaloupe- Oxford University Press.
listeria-outbreak-deadliest-decade/story?id¼14622507#.UKx7K-SE3pw. DeQueiroz, G. A., & Day, D. F. (2007). Antimicrobial activity and effectiveness of
Carpentier, B., & Chassaing, D. (2004). Interactions in biofilms between Listeria a combination of sodium hypochlorite and hydrogen peroxide in killing and
monocytogenes and resident microorganisms from food industry premises. removing Pseudomonas aeruginosa biofilms from surfaces. Journal of Applied
International Journal of Food Microbiology, 97(2), 111e122. Microbiology, 103, 794e802.
de Carvalho, C. C. C. R. (2007). Biofilms: recent developments on an old battle. Di Pinto, A., Novello, L., Montemurro, F., Bonerba, E., & Tantillo, G. (2010). Occur-
Recent Patents on Biotechnology, 1(1), 49e57. rence of Listeria monocytogenes in ready-to-eat foods from supermarkets in
de Carvalho, C. C. C. R., & da Fonseca, M. M. (2007). Assessment of Southern Italy. The New Microbiologica, 33(3), 249e252.
three-dimensional biofilm structure using an optical microscope. BioTechniques, Donlan, R. M. (2002). Biofilms: microbial life on surfaces. Emerging Infectious
42(5), 616e620. Diseases, 8(9), 881e890, Centers for Disease Control and Prevention.
Castonguay, M.-H., van der Schaaf, S., Koester, W., Krooneman, J., van der Meer, W., Dosti, B., Guzel-Seydim, Z., & Greene, A. K. (2005). Effectiveness of ozone, heat and
Harmsen, H., et al. (2006). Biofilm formation by Escherichia coli is stimulated by chlorine for destroying common food spoilage bacteria in synthetic media and
synergistic interactions and co-adhesion mechanisms with adherence- biofilms. International Journal of Dairy Technology, 58(1), 19e24.
proficient bacteria. Research in Microbiology, 157(5), 471e478. Dourou, D., Beauchamp, C. S., Yoon, Y., Geornaras, I., Belk, K. E., Smith, G. C., et al.
CDC. (2011). Vital signs: Incidence and trends of infection with pathogens transmitted (2011). Attachment and biofilm formation by Escherichia coli O157:H7 at different
commonly through food e foodborne diseases active surveillance network, 10 U.S. temperatures, on various food-contact surfaces encountered in beef processing.
Sites, 1996e2010. Morbidity and Mortality Weekly Report (MMWR), Retrieved International Journal of Food Microbiology, 149(3), 262e268, Elsevier B.V.
28.03.12, from. http://www.cdc.gov/mmwr/preview/mmwrhtml/mm6022a5. Dykes, G. A., Sampathkumar, B., & Korber, D. R. (2003). Planktonic or biofilm
htm?s_cid¼mm6022a5_w. growth affects survival, hydrophobicity and protein expression patterns of
Cerca, N., Oliveira, R., & Azeredo, J. (2007). Susceptibility of Staphylococcus epi- a pathogenic Campylobacter jejuni strain. International Journal of Food Microbi-
dermidis planktonic cells and biofilms to the lytic action of staphylococcus ology, 89(1), 1e10.
bacteriophage K. Letters in Applied Microbiology, 45(3), 313e317. Eide, M. H., Homleid, J. P., & Mattsson, B. (2003). Life cycle assessment (LCA) of
Chae, M. S., & Schraft, H. (2000). Comparative evaluation of adhesion and biofilm cleaning-in-place processes in dairies. LWT - Food Science and Technology, 36(3),
formation of different Listeria monocytogenes strains. International Journal of 303e314.
Food Microbiology, 62(1e2), 103e111. Enos-Berlage, J. L., Guvener, Z. T., Keenan, C. E., & McCarter, L. L. (2005). Genetic
Chan, K. F., Tran, H. L., Kanenaka, R. Y., & Kathariou, S. (2001). Survival of clinical and determinants of biofilm development of opaque and translucent Vibrio para-
poultry-derived isolates of Campylobacter jejuni at a low temperature ( 4  C ). haemolyticus. Molecular Microbiology, 55(4), 1160e1182.
Applied and Environmental Microbiology, 67(9), 4186e4191. Exner, M., Tuschewitzki, G. J., & Scharnagel, J. (1987). Influence of biofilms by
Chandra, J., Patel, J. D., Li, J., Zhou, G., Mukherjee, P. K., McCormick, T. S., et al. (2005). chemical disinfectants and mechanical cleaning. Zentralblatt für Bakteriologie,
Modification of surface properties of biomaterials influences the ability of Mikrobiologie und Hygiene. Serie B, Umwelthygiene, Krankenhaushygiene,
Candida albicans to form biofilms. Applied and Environmental Microbiology, Arbeitshygiene, präventive Medizin, 183(5e6), 549e563.
71(12), 8795e8801. FAO/WHO [Food and Agriculture Organization of the United Nations/World Health
Changani, S. D., Belmar-Beiny, M. T., & Fryer, P. J. (1997). Engineering and chemical Organization]. (2008). Microbiological hazards in fresh leafy vegetables and herbs:
factors associated with fouling and cleaning in milk processing. Experimental Meeting Report. Rome: Microbiological Risk Assessment Series No. 14.
Thermal and Fluid Science, 14(4), 392e406. Faruque, S. M., Biswas, K., Udden, S. M. N., Ahmad, Q. S., Sack, D. A., Nair, G. B., et al.
Chaturongkasumrit, Y., Takahashi, H., Keeratipibul, S., Kuda, T., & Kimura, B. (2011). (2006). Transmissibility of cholera: in vivo-formed biofilms and their
The effect of polyesterurethane belt surface roughness on Listeria mono- relationship to infectivity and persistence in the environment. Proceedings
cytogenes biofilm formation and its cleaning efficiency. Food Control, 22(12), of the National Academy of Sciences of the United States of America, 103(16),
1893e1899, Elsevier Ltd. 6350e6355.
Chemat, F., Zill-e-Huma, & Khan, M. K. (2011). Applications of ultrasound in food Ferreira, C., Pereira, A. M., & Melo, L. F. (2010). Advances in industrial biofilm control
technology: processing, preservation and extraction. Ultrasonics Sonochemistry, with micro-nanotechnology. Applied Microbiology, 845e854.
18(4), 813e835, Elsevier B.V. Ferreira, C., Pereira, A. M., & Pereira, M. C. (2011). Biofilm control with new
Chen, G. (2012). Escherichia coli adhesion to abiotic surfaces in the presence of non- microparticles with new immobilized biocide. In M. R. Malayeri, H. Müller-
ionic surfactants. Journal of Adhesion Science and Technology, 17(16), 2131e2139. Steinhagen, & A. P. Watkinson (Eds.). Proceedings of international conference on
Chia, T. W. R., Goulter, R. M., McMeekin, T., Dykes, G. A., & Fegan, N. (2009). heat exchanger fouling and cleaning, Vol. 2011 (pp. 174e179), Crete Island, Greece.
Attachment of different Salmonella serovars to materials commonly used in Flemming, H.-C., & Ridgway, H. (2009). Biofilm control: conventional and alterna-
a poultry processing plant. Food Microbiology, 26(8), 853e859, Elsevier Ltd. tive approaches. Marine and Industrial Biofouling, 4(1), 103e117.
Chmielewski, R. A. N., & Frank, J. F. (2003). Biofilm formation and control in food Flemming, H.-C., & Schaule, G. (1988). Biofouling on membranes e a microbiological
processing facilities. Comprehensive Reviews in Food Science and Food Safety, approach. Desalination, 70(1e3), 95e119.
2(1), 22e32. Flint, S. H., Bremer, P. J., & Brooks, J. D. (1997). Biofilms in dairy manufacturing
Chmielewski, R. A. N., & Frank, J. F. (2006). A predictive model for heat inactivation plant-description, current concerns and methods of control. Biofouling: The
of Listeria monocytogenes biofilm on buna-N rubber. LWT - Food Science and Journal of Bioadhesion and Biofilm Research, 11(1), 81e97.
Technology, 39(1), 11e19. Forsythe, S. J., & Hayes, P. R. (1998). Food hygiene, microbiology and HACCP (3rd ed.).
Chmielewski, R. A. N., & Frank, J. F. (2007). Inactivation of Listeria monocytogenes Aspen.
biofilms using chemical sanitizers and heat. In H. P. Blaschek, H. H. Wang, & Frank, J. F., Ehlers, J., & Wicker, L. (2003). Removal of Listeria monocytogenes and
M. E. Agle (Eds.), Biofilms in the food environment (pp. 73e104). Ames: Blackwell poultry soil-containing biofilms using chemical cleaning and sanitizing agents
Publishing. under static condions. Food Protection Trends, 23, 654e663.
Choi, N.-C., Park, S.-J., Lee, C.-G., Park, J.-A., & Kim, S.-B. (2011). Influence of Fransisca, L., Zhou, B., Park, H., & Feng, H. (2011). The effect of calcinated calcium
surfactants on bacterial adhesion to metal oxide-coated surfaces. Environmental and chlorine treatments on Escherichia coli O157:H7 87-23 population reduc-
Engineering Research, 16(4), 219e225. tion in radish sprouts. Journal of Food Science, 76(6), M404eM412.
Christina, T., Penna, V., Mazzola, P. G., Maria, A., & Martins, S. (2001). The efficacy of Gandhi, M., Golding, S., Yaron, S., & Matthews, K. R. (2001). Use of green fluorescent
chemical agents in cleaning and disinfection programs. BMC Infectious Deseases, protein expressing Salmonella stanley to investigate survival, spatial location,
1(16). and control on alfalfa sprouts. Journal of Food Protection, 64(12), 1891e1898.
Cloete, T. E., Jacobs, L., & Brözel, V. S. (1998). The chemical control of biofouling in Garrido, V., Vitas, A. I., & García-Jalón, I. (2009). Survey of Listeria monocytogenes in
industrial water systems. Biodegradation, 9(1), 23e37. ready-to-eat products: prevalence by brands and retail establishments
S. Srey et al. / Food Control 31 (2013) 572e585 583

for exposure assessment of listeriosis in Northern Spain. Food Control, 20(11), Kim, H., Ryu, J.-H., & Beuchat, L. R. (2006). Attachment of and biofilm formation by
986e991, Elsevier Ltd. Enterobacter sakazakii on stainless steel and enteral feeding tubes. Applied and
Gerke, C., Kraft, A., Süssmuth, R., Schweitzer, O., & Götz, F. (1998). Characterization Environmental Microbiology, 72(9), 5846e5856.
of the N-acetylglucosaminyltransferase activity involved in the biosynthesis of Kim, T. J., Silva, J. L., Chamul, R. S., & Chen, T. C. (2000). Influence of ozone, hydrogen
the Staphylococcus epidermidis polysaccharide intercellular adhesin. The Journal peroxide, or Salt on microbial Profile, TBARS and Color of channel Catfish fillets.
of Biological Chemistry, 273(29), 18586e18593. Journal of Food Science, 65(7), 1210e1213.
Gerstel, U., & Römling, U. (2001). Oxygen tension and nutrient starvation are major Kim, S.-H., & Wei, C.-I. (2009). Molecular characterization of biofilm formation and
signals that regulate agfD promoter activity and expression of the multicellular attachment of Salmonella enterica serovar typhimurium DT104 on food contact
morphotype in Salmonella typhimurium. Environmental Microbiology, 3(10), surfaces. Journal of Food Protection, 72(9), 1841e1847.
638e648. Klausen, M., Heydorn, A., Ragas, P., Lambertsen, L., Aaes-Jørgensen, A., Molin, S.,
Gilbert, P., & Mcbain, A. J. (2003). Potential impact of increased use of biocides in et al. (2003). Biofilm formation by Pseudomonas aeruginosa wild type, flagella
consumer products on prevalence of antibiotic resistance. Clinical Microbiology and type IV pili mutants. Molecular Microbiology, 48(6), 1511e1524.
Reviews, 16(2), 189e208. Klayman, B. J., Volden, P. A., Stewart, P. S., & Camper, A. K. (2009). Escherichia coli
Goldman, G., Starosvetsky, J., & Armon, R. (2009). Inhibition of biofilm formation on O157:H7 requires colonizing partner to adhere and persist in a capillary flow
UF membrane by use of specific bacteriophages. Journal of Membrane Science, cell. Environmental Science & Technology, 43(6), 2105e2111.
342(1e2), 145e152. Knetsch, M. L. W., & Koole, L. H. (2011). New strategies in the development of
Gram, L., Bagge-Ravn, D., Ng, Y. Y., Gymoese, P., & Vogel, B. F. (2007). Influence of antimicrobial coatings: the example of increasing usage of silver and silver
food soiling matrix on cleaning and disinfection efficiency on surface attached nanoparticles. Polymers, 3(1), 340e366.
Listeria monocytogenes. Food Control, 18(10), 1165e1171. Kogure, K., Ikemoto, E., & Morisaki, H. (1998). Attachment of Vibrio alginolyticus to
Gram, L., & Huss, H. H. (1996). Microbiological spoilage of fish and fish products. glass surfaces is dependent on swimming speed. Journal of Bacteriology, 180(4),
International Journal of Food Microbiology, 33(1), 121e137. 932e937.
Guðbjörnsdóttir, B., Einarsson, H., & Thorkelsson, G. (2005). Microbial adhesion to Koutzayiotis, C. (1992). Bacterial biofilms in milk pipelines. African Journal of Dairy
processing lines for fish fillets and cooked shrimp: influence of stainless steel Science, 24, 19e22.
surface finish and presence of gram-negative bacteria on the attachment of Królasik, J., Zakowska, Z., Krepska, M., & Klimek, L. (2010). Resistance of bacterial
Listeria monocytogenes. Food Technology and Biotechnology, 43(1), 55e61. biofilms formed on stainless steel surface to disinfecting agent. Polish Journal of
Ha, J.-H., & Ha, S.-D. (2011). Synergistic effects of sodium hypochlorite and ultra- Microbiology, 59(4), 281e287.
violet radiation in reducing the levels of selected foodborne pathogenic Kuda, T., Yano, T., & Kuda, M. T. (2008). Resistances to benzalkonium chloride of
bacteria. Foodborne Pathogens and Disease, 8(5), 587e591. bacteria dried with food elements on stainless steel surface. LWT - Food Science
Habimana, O., Heir, E., Langsrud, S., Asli, A. W., & Møretrø, T. (2010). Enhanced and Technology, 41(6), 988e993.
surface colonization by Escherichia coli O157:H7 in biofilms formed by an Kumar, C. G., & Anand, S. K. (1998). Significance of microbial biofilms in food
Acinetobacter calcoaceticus isolate from meat-processing environments. Applied industry: a review. International Journal of Food Microbiology, 42(1e2), 9e27.
and Environmental Microbiology, 76(13), 4557e4559. Lagrange, F., Reiprich, W., & Hoffmann, M. (2004). CIP-cleaning and disinfection
Han, Y., Sherman, D. M., Linton, R. H., Nielsen, S. S., & Nelson, P. E. (2000). The effects with ozone water. Fleischwirtschaft, 84(2), 112e114, Deutscher Fachverlag.
of washing and chlorine dioxide gas on survival and attachment of Escherichia Lapidot, A., Romling, U., & Yaron, S. (2006). Biofilm formation and the survival of
coli O157: H7 to green pepper surfaces. Food Microbiology, 17(5), 521e533. Salmonella typhimurium on parsley. International Journal of Food Microbiology,
Hanning, I., Jarquin, R., & Slavik, M. (2008). Campylobacter jejuni as a secondary 109(3), 229e233.
colonizer of poultry biofilms. Journal of Applied Microbiology, 105(4), 1199e1208. Latorre, A. A., Van Kessel, J. S., Karns, J. S., Zurakowski, M. J., Pradhan, A. K., Boor, K. J.,
Harris, N. V., Weiss, N. S., & Nolan, C. M. (1986). The role of poultry and meats in the et al. (2010). Biofilm in milking equipment on a dairy farm as a potential source
etiology of Campylobacter jejuni/coli Enteritis. American Journal of Public Health, of bulk tank milk contamination with Listeria monocytogenes. Journal of Dairy
76(4), 407e411. Science, 93(6), 2792e2802, Elsevier.
Harvey, J., Keenan, K. P., & Gilmour, A. (2007). Assessing biofilm formation by Lis- Lelièvre, C., Antonini, G., Faille, C., & Bénézech, T. (2002). Cleaning-in-Place:
teria monocytogenes strains. Food Microbiology, 24(4), 380e392. modelling of cleaning kinetics of pipes soiled by Bacillus spores assuming
Hashimoto, H. (2001). Evaluation of the anti-biofilm effect of a new anti-bacterial a process combining removal and deposition. Food and Bioproducts, 80,
silver citrate/lecithin coating in an in-vitro experimental system using a modi- 305e312.
fied Robbins device. Kansensho  gaku zasshi. The Journal of the Japanese Lelièvre, C., Faile, C., & Bénézech, T. (2001). Removal kinetics of Bacillus cereus spores
Association for Infectious Diseases, 75(8), 678e685. from stainless steel pipes under CIP procedure: influence of soiling and cleaning
Helke, D. M., Somers, E. B., & Wong, A. C. L. (1993). Attachment of Listeria mono- conditions. Journal of Food Process, 24(6), 359e379.
cytogenes and Salmonella typhimurium to stainless steel and buna-N in the Lelièvre, C., Legentilhomme, P., Gaucher, C., Legrand, J., Faille, C., & Bénézech, T.
presence of milk and individual milk components. Journal of Food Protection, (2002). Cleaning in place: effect of local wall shear stress variation on bacterial
56(6), 479e484. removal from stainless steel equipment. Chemical Engineering Science, 57(8),
Helke, D. M., & Wong, A. C. L. (1994). Survival and growth characteristics of Listeria 1287e1297.
monocytogenes and Salmonella typhimurium on stainless steel and buna-n Lequette, Y., Boels, G., Clarisse, M., & Faille, C. (2010). Using enzymes to remove bio-
rubber. Journal of Food Protection, 57(6), 963e968, International Association films of bacterial isolates sampled in the food-industry. Biofouling, 26(4), 421e431.
for Food Protection. Liao, C.-H., & Cooke, P. H. (2001). Response to trisodium phosphate treatment of
Hinsa, S. M., & O’Toole, G. A. (2006). Biofilm formation by Pseudomonas fluorescens Salmonella Chester attached to fresh-cut green pepper slices. Canadian Journal
WCS365: a role for LapD. Microbiology, 152(5), 1375e1383. of Microbiology, 47(1), 25e32.
Hopkins, R. S., & Scott, A. S. (1983). Handling raw chicken as a source for sporadic Lomander, A., Schreuders, P., Russek-Cohen, E., & Ali, L. (2004). Evaluation of
Campylobacter jejuni infections. The Journal of Infectious Diseases, 148(4), 770. chlorines’ impact on biofilms on scratched stainless steel surfaces. Bioresource
Horvath, M., Bilitzky, L., & Hüttner, J. (1985). Ozone. Amsterdam; New York: Elsevier. Technology, 94(3), 275e283.
Howell, D., & Behrends, B. (2006). A review of surface roughness in antifouling coatings López-Gálvez, F., Gil, M. I., Truchado, P., Selma, M. V., & Allende, A. (2010). Cross-
illustrating the importance of cutoff length. Biofouling, 22(5e6), 401e410. contamination of fresh-cut lettuce after a short-term exposure during pre-
Janssens, J. C. A., Steenackers, H., Robijns, S., Gellens, E., Levin, J., Zhao, H., et al. washing cannot be controlled after subsequent washing with chlorine
(2008). Brominated furanones inhibit biofilm formation by Salmonella dioxide or sodium hypochlorite. Food Microbiology, 27(2), 199e204.
enterica serovar typhimurium. Applied and Environmental Microbiology, 74(21), Lu, T. K., & Collins, J. J. (2007). Dispersing biofilms with engineered enzymatic
6639e6648. bacteriophage. Proceedings of the National Academy of Sciences of the United
Jessen, B., & Lammert, L. (2003). Biofilm and disinfection in meat processing plants. States of America, 104(27), 11197e11202.
International Biodeterioration & Biodegradation, 51(4), 265e269. McBain, A. J., Rickard, A. H., & Gilbert, P. (2002). Possible implications of biocide
Johansen, C., Falholt, P., & Gram, L. (1997). Enzymatic removal and disinfection of accumulation in the environment on the prevalence of bacterial antibiotic
bacterial biofilms. Applied and Environmental Microbiology, 63(9), 3724e3728. resistance. Journal of Industrial Microbiology & Biotechnology, 29(6), 326e330.
Joseph, L. A., & Wright, A. C. (2004). Expression of Vibrio vulnificus capsular poly- Mafu, A. A., Roy, D., Goulet, J., & Magny, P. (1990). Attachment of Listeria mono-
saccharide inhibits biofilm formation. Journal of Bacteriology, 186(3), 889e893. cytogenes to stainless steel, glass, polypropylene, and rubber surfaces after short
Kaplan, J. B., Ragunath, C., Ramasubbu, N., & Fine, D. H. (2003). Detachment of contact times. Journal of Food Protection, 53(9), 742e746.
Actinobacillus actinomycetemcomitans biofilm cells by an endogenous b-hexos- McLandsborough, L., Rodriguez, A., Pérez-Conesa, D., & Weiss, J. (2006). Biofilms:
aminidase activity. Journal of Bacteriology, 185(16), 4693e4698. at the interface between biophysics and microbiology. Food Biophysics, 1(2),
Kaplan, J. B., Ragunath, C., Velliyagounder, K., Fine, D. H., & Ramasubbu, N. (2004). 94e114.
Enzymatic detachment of Staphylococcus epidermidis biofilms. Antimicrobial McLean, R. J. C., Whiteley, M., Stickler, D. J., & Fuqua, W. C. (1997). Evidence of
Agents and Chemotherapy, 48(7), 2633e2636. autoinducer activity in naturally occurring biofilms. FEMS Microbiology Letters,
Karunasagar, I., Otta, S. K., & Karunasagar, I. (1996). Biofilm formation by Vibrio 154, 259e263.
harveyi on surfaces. Aquaculture, 140(3), 241e245. Mariani, C., Oulahal, N., Chamba, J.-F., Dubois-Brissonnet, F., Notz, E., & Briandet, R.
Keskinen, L. A., Burke, A., & Annous, B. A. (2009). Efficacy of chlorine, acidic elec- (2011). Inhibition of Listeria monocytogenes by resident biofilms present on
trolyzed water and aqueous chlorine dioxide solutions to decontaminate wooden shelves used for cheese ripening. Food Control, 22(8), 1357e1362,
Escherichia coli O157:H7 from lettuce leaves. International Journal of Food Elsevier Ltd.
Microbiology, 132(2e3), 134e140, Elsevier B.V. Marin, C., Hernandiz, A., & Lainez, M. (2009). Biofilm development capacity of
Khardre, M. A., Yousef, A. E., & Kim, J.-G. (2001). Microbiological aspects of ozone Salmonella strains isolated in poultry risk factors and their resistance against
́
applications in food: a review. Journal of Food Science, 66(9), 1242e1252. disinfectants. Poultry Science, 88(2), 424e431.
584 S. Srey et al. / Food Control 31 (2013) 572e585

Marouani-Gadri, N., Augier, G., & Carpentier, B. (2009). Characterization of bacterial Patel, J., & Sharma, M. (2010). Differences in attachment of Salmonella enterica
strains isolated from a beef-processing plant following cleaning and disinfec- serovars to cabbage and lettuce leaves. International Journal of Food Microbi-
tion e influence of isolated strains on biofilm formation by Sakaï and EDL 933 E. ology, 139(1e2), 41e47, Elsevier B.V.
coli O157:H7. International Journal of Food Microbiology, 133(1e2), 62e67, Patel, J., Sharma, M., Millner, P., Calaway, T., & Singh, M. (2011). Inactivation of
Elsevier B.V. Escherichia coli O157:H7 attached to spinach harvester blade using bacterio-
Marques, S. C., Rezende, J. D. G. O. S., Alves, L. A. D. F., Silva, B. C., Alves, E., phage. Foodborne Pathogens and Disease, 8(4), 541e546.
Abreu, L. R. D., et al. (2007). Formation of biofilms by Staphylococcus aureus on Patil, S. (2010). Efficacy of ozone and ultrasound for microbial reduction in fruit juice.
stainless steel and glass surfaces and its resistance to some selected chemical Dublin Institute of Technology.
sanitizers. Brazilian Journal of Microbiology, 38(3), 538e543. Pesci, E. C., Milbank, J. B., Pearson, J. P., McKnight, S., Kende, A. S., Greenberg, E. P.,
Maukonen, J., Mättö, J., Wirtanen, G., Raaska, L., Mattila-Sandholm, T., & Saarela, M. et al. (1999). Quinolone signaling in the cell-to-cell communication system of
(2003). Methodologies for the characterization of microbes in industrial Pseudomonas aeruginosa. Proceedings of the National Academy of Sciences of the
environments: a review. Journal of Industrial Microbiology and Biotechnology, United States of America, 96(20), 11229e11234.
30(6), 327e356. Peterson, R. V., & Pitt, W. G. (2000). The effect of frequency and power density on
Merril, C. R., Scholl, D., & Adhya, S. L. (2003). The prospect for bacteriophage therapy the ultrasonically-enhanced killing of biofilm-sequestered Escherichia coli.
in western medicine. Nature Reviews. Drug Discovery, 2(6), 489e497. Colloids and Surfaces B: Biointerfaces, 17(4), 219e227.
Meyer, B. (2003). Approaches to prevention, removal and killing of biofilms. Pillai, S. K., Sakoulas, G., Eliopoulos, G. M., Moellering, R. C., Murray, B. E., &
International Biodeterioration & Biodegradation, 51(4), 249e253. Inouye, R. T. (2004). Effects of glucose on fsr-mediated biofilm formation in
Midelet, G., & Carpentier, B. (2004). Impact of cleaning and disinfection agents on Enterococcus faecalis. The Journal of Infectious Diseases, 190(5), 967e970.
biofilm structure and on microbial transfer to a solid model food. Journal of Piyasena, P., Mohareb, E., & McKellar, R. C. (2003). Inactivation of microbes
Applied Microbiology, 97(2), 262e270. using ultrasound: a review. International Journal of Food Microbiology, 87(3),
Molobela, I. P., Cloete, T. E., & Beukes, M. (2010). Protease and amylase enzymes for 207e216.
biofilm removal and degradation of extracellular polymeric substances (EPS) Qian, Z., Sagers, R. D., & Piti, W. G. (1997). The effect of ultrasonic frequency upon
produced by Pseudomonas fluorescens bacteria. Journal of Microbiology, 4(14), enhanced killing of P. aeruginosa biofilms. Annals of Biomedical Engineering,
1515e1524. 25(1), 69e76.
Momba, M. N. B., & Binda, M. A. (2002). Combining chlorination and chloramination Rajkowski, K. T. (2009). Biofilms in fish processing. In P. M. Fratamico, B. A. Annous,
processes for the inhibition of biofilm formation in drinking surface water & N. W. Gunther (Eds.), Biofilms in the food and beverage industries (pp. 499e
system models. Journal of Applied Microbiology, 92(4), 641e648. 516). Woodhead Publishing.
Momba, M. N. B., & Makala, N. (2004). Comparing the effect of various pipe Ramesh, N., Joseph, S. W., Carr, L. E., Douglass, L. W., & Wheaton, F. W. (2002).
materials on biofilm formation in chlorinated and combined chlorine- Evaluation of chemical disinfectants for the elimination of Salmonella biofilms
chloraminated water systems. WaterSA, 30(2), 175e182. from poultry transport containers. Poultry Science, 81(6), 904e910.
Møretrø, T., Hermansen, L., Holck, A. L., Sidhu, M. S., Rudi, K., & Langsrud, S. (2003). Rideout, K., Teschke, K., Dimich-Ward, H., & Kennedy, S. M. (2005). Considering risks
Biofilm formation and the presence of the intercellular adhesion Locus ica to healthcare workers from glutaraldehyde alternatives in high-level disinfec-
among Staphylococci from food and food processing environments. Applied and tion. The Journal of Hospital Infection, 59(1), 4e11.
Environmental Microbiology, 69(9), 5648e5655. Robbins, J. B., Fisher, C. W., Moltz, A. G., & Martin, S. E. (2005). Elimination of Listeria
Neuman, W. (2011). Listeria outbreak traced to Colorado cantaloupe packing shed e monocytogenes biofilms by ozone, chlorine, and hydrogen peroxide. Journal of
NYTimes. Retrieved 21.11.2012, from. http://www.nytimes.com/2011/10/20/ Food Protection, 68(3), 494e498.
business/listeria-outbreak-traced-to-colorado-cantaloupe-packing-shed.html? Romney, A. J. D. (1990). CIP: Cleaning in place. Cambridgeshire, UK: Huntingdon.
pagewanted¼all. Ronner, A. B., & Wong, A. C. L. (1993). Biofilm development and sanitizer inactiva-
Nilsson, R. E., Ross, T., & Bowman, J. P. (2011). Variability in biofilm production by tion of Listeria monocytogenes and Salmonella typhimurium on stainless steel
Listeria monocytogenes correlated to strain origin and growth conditions. and buna-n rubber. Journal of Food Protection, 56(9), 750e758.
International Journal of Food Microbiology, 150(1), 14e24, Elsevier B.V. Rosmaninho, R., Santos, O., Nylander, T., Paulsson, M., Beuf, M., Benezech, T., et al.
Norwood, D. E., & Gilmour, A. (2000). The growth and resistance to sodium hypo- (2007). Modified stainless steel surfaces targeted to reduce fouling e evaluation
chlorite of Listeria monocytogenes in a steady-state multispecies biofilm. Journal of fouling by milk components. Journal of Food Engineering, 80(4), 1176e1187.
of Applied Microbiology, 88(3), 512e520. Rossoni, E. M., & Gaylarde, C. C. (2000). Comparison of sodium hypochlorite and
Ölmez, H., & Temur, S. D. (2010). Effects of different sanitizing treatments on bio- peracetic acid as sanitising agents for stainless steel food processing surfaces
films and attachment of Escherichia coli and Listeria monocytogenes on green using epifluorescence microscopy. International Journal of Food Microbiology,
leaf lettuce. LWT - Food Science and Technology, 43(6), 964e970, Elsevier Ltd. 61(1), 81e85.
Orgaz, B., Neufeld, R. J., & Sanjose, C. (2007). Single-step biofilm removal with Salo, S., Ehavald, H., Raaska, L., Vokk, R., & Wirtanen, G. (2006). Microbial surveys in
delayed release encapsulated Pronase mixed with soluble enzymes. Enzyme, 40, Estonian dairies. LWT - Food Science and Technology, 39(5), 460e471.
1045e1051. Salvia, A. C. R. D., Teodoro, G. R., Balducci, I., Koga-Ito, C. Y., & Oliveira, S. H. G. D.
Osaili, T. M., Alaboudi, A. R., & Nesiar, E. A. (2011). Prevalence of Listeria spp. and (2011). Effectiveness of 2% peracetic acid for the disinfection of gutta-percha
antibiotic susceptibility of Listeria monocytogenes isolated from raw chicken and cones. Brazilian Oral Research, 25(1), 23e27.
ready-to-eat chicken products in Jordan. Food Control, 22(3e4), 586e590, Sanders, S. Q., Boothe, D. H., Frank, J. F., & Arnold, J. W. (2007). Culture and detection
Elsevier Ltd. of Campyloabaeter jejuni within mixed microbial populations of biofilms on
O’Toole, G. A., & Kaplan, H. B. (2000). Biofilm formation as microbial development. stainless steel. Journal of Food Protection, 70(6), 1379e1385.
Annual Reviews in Microbiology, 49e79. Sapers, G. M. (2001). Efficacy of washing and sanitizing methods for disinfection of
O’Toole, G. A., & Kolter, R. (1998). Flagellar and twitching motility are necessary for fresh fruit and vegetable products. Food Technology and Biotechnology, 39(4),
Pseudomonas aeruginosa biofilm development. Molecular Microbiology, 30(2), 305e311.
295e304. Sapers, G. M., & Sites, J. E. (2003). Efficacy of 1% hydrogen peroxide wash in
Oulahal-Lagsir, N., Martial-Gros, A., Boistier, E., Blum, L. J., & Bonneau, M. (2000a). decontaminating apples. Journal of Food Science, 68(5), 1793e1797.
The development of an ultrasonic apparatus for the non-invasive and repeat- Sauer, K., Camper, A. K., Ehrlich, G. D., Costerton, J. W., & Davies, D. G. (2002).
able removal of fouling in food processing equipment. Letters in Applied Pseudomonas aeruginosa displays multiple phenotypes during development as
Microbiology, 30(1), 47e52. a biofilm. Bacteriology, 184(4), 1140e1154.
Oulahal-Lagsir, N., Martial-Gros, A., Bonneau, M., & Blum, L. J. (2000b). Ultrasonic Scallan, E. (2011). Foodborne illness acquired in the United Statesdunspecified
methodology coupled to ATP bioluminescence for the non-invasive detection of agents. Emerging Infectious Diseases, 17(1), 16e22.
fouling in food processing equipmentevalidation and application to a dairy Scallan, E., Hoekstra, R. M., Angulo, F. J., Tauxe, R. V., Widdowson, M.-A., Roy, S. L.,
factory. Journal of Applied Microbiology, 89(3), 433e441. et al. (2011). Foodborne illness acquired in the United Statesdmajor pathogens.
Oulahal, N., Martial-Gros, A., Bonneau, M., & Blum, L. J. (2007). Removal Emerging Infectious Disease Journal - CDC, 17(1).
of meat biofilms from surfaces by ultrasounds combined with enzymes Scardino, A. J., Harvey, E., & De Nys, R. (2006). Testing attachment point theory:
and/or a chelating agent. Innovative Food Science & Emerging Technologies, 8(2), diatom attachment on microtextured polyimide biomimics. Biofouling, 22(1e2),
192e196. 55e60.
Ozdemir, H. O., Buzoglu, H. D., Calt, S., Stabholz, A., & Steinberg, D. (2010). Effect of Schurman, J. J., Sumner, S. S., & Marcy, J. E. (2001). Antibacterial activity of hydrogen
ethylenediaminetetraacetic acid and sodium hypochlorite irrigation on peroxide against Escherichia coli O157:H7 and Salmonella spp. in friut juices, both
Enterococcus faecalis biofilm colonization in young and old human root canal alone and in combination with organic acids. Virginia Polytechnic Institute and
dentin: in vitro study. Journal of Endodontics, 36(5), 842e846. State University.
Park, S.-I., Daeschel, M. A., & Zhao, Y. (2004). Functional properties of antimicrobial September, S. M., Els, F. A., Venter, S. N., & Brözel, V. S. (2007). Prevalence of bacterial
lysozyme-chitosan composite films. Journal of Food Science, 69(8), M215eM221. pathogens in biofilms of drinking water distribution systems. Journal of Water
Park, S. H., Jarquin, R., Hanning, I., Almeida, G., & Ricke, S. C. (2011). Detection of and Health, 05(2), 219e227.
Salmonella spp. survival and virulence in poultry feed by targeting the hilA gene. Sharma, M., & Anand, S. K. (2002). Characterization of constitutive microflora of
Journal of Applied Microbiology, 111(2), 426e432. biofilms in dairy processing lines. Food Microbiology, 19, 627e636.
Parsek, M. R., & Singh, P. K. (2003). Bacterial biofilms: an emerging link to disease Shikongo-Nambabi, M. (2011). Control of bacterial contamination during marine
pathogenesis. Annual Review of Microbiology, 57, 677e701. fish processing. Journal of Biology, 3(1), 1e17.
Pascual, A., Llorca, I., & Canut, A. (2007). Use of ozone in food industries for reducing Shikongo-Nambabi, M., Kachigunda, B., & Venter, S. N. (2010). Evaluation of
the environmental impact of cleaning and disinfection activities. Trends in Food oxidising disinfectants to control Vibrio biofilms in treated seawater used for
Science & Technology, 18, S29eS35. fish processing. WaterSA, 36(3), 215e220.
S. Srey et al. / Food Control 31 (2013) 572e585 585

Sidhu, M. S., Langsrud, S., & Holck, A. (2001). Disinfectant and antibiotic resistance Tait, K., Skillman, L. C., & Sutherland, I. W. (2002). The efficacy of bacteriophage as
of lactic acid bacteria isolated from the food industry. Microbial Drug Resistance a method of biofilm eradication. Biofouling, 18(4), 305e311.
(Larchmont, N.Y.), 7(1), 73e83. Tang, X., Flint, S. H., Bennett, R. J., Brooks, J. D., & Morton, R. H. (2009). Biofilm
Silagyi, K., Kim, S.-H., Lo, Y. M., & Wei, C.-i (2009). Production of biofilm and quorum growth of individual and dual strains of Klebsiella oxytoca from the dairy
sensing by Escherichia coli O157:H7 and its transfer from contact surfaces to industry on ultrafiltration membranes. Journal of Industrial Microbiology &
meat, poultry, ready-to-eat deli, and produce products. Food Microbiology, 26(5), Biotechnology, 36(12), 1491e1497.
514e519, Elsevier Ltd. Thouvenin, M., Langlois, V., Briandet, R., Langlois, J. Y., Guerin, P. H., Peron, J. J., et al.
Sillankorva, S., Neubauer, P., & Azeredo, J. (2008). Pseudomonas fluorescens biofilms (2003). Study of erodable paint properties involved in antifouling activity.
subjected to phage phiIBB-PF7A. BMC Biotechnology, 8, 79. Biofouling, 19(3), 177e186.
Sillankorva, S., Neubauer, P., & Azeredo, J. (2010). Phage control of dual species Toté, K., Horemans, T., Vanden Berghe, D., Maes, L., & Cos, P. (2010). Inhibitory effect
biofilms of Pseudomonas fluorescens and Staphylococcus lentus. Biofouling, 26(5), of biocides on the viable masses and matrices of Staphylococcus aureus and
567e575. Pseudomonas aeruginosa biofilms. Applied and Environmental Microbiology,
Sillankorva, S., Oliveira, R., Vieira, M. J., Sutherland, I., & Azeredo, J. (2004). Pseu- 76(10), 3135e3142.
domonas fluorescens infection by bacteriophage PhiS1: the influence of Trachoo, N., Frank, J. F., & Stern, N. J. (2002). Survival of Campylobacter
temperature, host growth phase and media. FEMS Microbiology Letters, 241(1), jejuni in biofilms isolated from chicken houses. Journal of Food Protection, 65(7),
13e20. 1110e1116.
da Silva, P. M. B., Acosta, E. J. T. R., de Rezende Pinto, L., Graeff, M., Vanhaecke, E., Remon, J. P., Moors, M., Raes, F., De Rudder, D., & Van Peteghem, A.
Spolidorio, D. M. P., Almeida, R. S., et al. (2011). Microscopical analysis of (1990). Kinetics of Pseudomonas aeruginosa adhesion to 304 and 316-L stainless
Candida albicans biofilms on heat-polymerised acrylic resin after chlorhexidine steel: role of cell surface hydrophobicity. Applied and Environmental Microbi-
gluconate and sodium hypochlorite treatments. Mycoses, 54(6), e712ee717. ology, 56(3), 788e795.
Simões, M., Pereira, M. O., & Vieira, M. J. (2005). Effect of mechanical stress on Van Houdt, R., & Michiels, C. W. (2005). Role of bacterial cell surface structures in
biofilms challenged by different chemicals. Water Research, 39(20), 5142e5152. Escherichia coli biofilm formation. Research in Microbiology, 156(5e6), 626e633.
Simões, M., Simões, L. C., Machado, I., Pereira, M. O., & Vieira, M. J. (2006). Control of Vankerckhoven, E., Verbessem, B., Crauwels, S., Declerck, P., Muylaert, K.,
flow-generated biofilms with surfactants: evidence of resistance and recovery. Willems, K. A., et al. (2011). Exploring the potential synergistic effects of chemical
Food and Bioproducts, 84(4), 338e345. disinfectants and UV on the inactivation of free-living bacteria and treatment of
Simões, M., Simões, L. C., & Vieira, M. J. (2010). A review of current and emergent biofilms in a pilot-scale system. Water Science & Technology, 64(6), 1247e1253.
biofilm control strategies. LWT - Food Science and Technology, 43(4), 573e583. Verma, V., Harjai, K., & Chhibber, S. (2010). Structural changes induced by a lytic
Sinde, E., & Carballo, J. (2000). Attachment of Salmonella spp. and Listeria bacteriophage make ciprofloxacin effective against older biofilm of Klebsiella
monocytogenes to stainless steel, rubber and polytetrafluorethylene: the influ- pneumoniae. Biofouling, 26(6), 729e737.
ence of free energy and the effect of commercial sanitizers. Food Microbiology, Vestby, L. K., Møretrø, T., Langsrud, S., Heir, E., & Nesse, L. L. (2009). Biofilm forming
17, 439e447. abilities of Salmonella are correlated with persistence in fish meal- and feed
Skerker, J. M., & Berg, H. C. (2001). Direct observation of extension and retraction of factories. BMC Veterinary Research, 5, 20.
type IV pili. Proceedings of the National Academy of Sciences of the United States of Waak, E., Tham, W., & Danielsson-Tham, M.-L. (2002). Prevalence and fingerprinting
America, 98(12), 6901e6904. of Listeria monocytogenes strains isolated from raw whole milk in farm bulk
Sofos, J. N., & Geornaras, I. (2010). Overview of current meat hygiene and safety tanks and in dairy plant receiving tanks. Applied and Environmental Microbi-
risks and summary of recent studies on biofilms, and control of Escherichia coli ology, 68(7), 3366e3370.
O157:H7 in nonintact, and Listeria monocytogenes in ready-to-eat, meat prod- Wagner, M., Auer, B., Trittremmel, C., Hein, I., & Schoder, D. (2007). Survey on the
ucts. Meat Science, 86(1), 2e14, Elsevier Ltd. Listeria contamination of ready-to-eat food products and household environ-
Spratt, D. A., Pratten, J., Wilson, M., & Gulabivala, K. (2001). An in vitro evaluation of ments in Vienna, Austria. Zoonoses and Public Health, 54(1), 16e22.
the antimicrobial efficacy of irrigants on biofilms of root canal isolates. Inter- Walton, M. (2008). Principles of cleaning-in-place (CIP). In A. Tamime (Ed.),
national Endodontic Journal, 34(4), 300e307. Cleaning-in-Place: Dairy, food and beverage operations (pp. 1e9). Oxford: Black-
Stepanovi c, S., Cirkovi
c, I., Ranin, L., & Svabi
c-Vlahovi
c, M. (2004). Biofilm formation well Publishing, Third.
by Salmonella spp. and Listeria monocytogenes on plastic surface. Letters in Whipps, J. M., Hand, P., Pink, D. A. C., & Bending, G. D. (2008). Chapter 7: human
Applied Microbiology, 38(5), 428e432. pathogens and the phyllosphere. Advances in Applied Microbiology, 64, 183e221.
Stepanovi 
c, S., Cirkovic, I., Mija 
c, V., & Svabi 
c-Vlahovi
c, M. (2003). Influence of the Whiteley, M., Bangera, M. G., Bumgarner, R. E., Parsek, M. R., Teitzel, G. M., Lory, S.,
incubation temperature, atmosphere and dynamic conditions on biofilm et al. (2001). Gene expression in Pseudomonas aeruginosa biofilms. Letter to
formation by Salmonella spp. Food Microbiology, 20, 339e343. Nature, 413, 860e864.
Stewart, P. S., & Franklin, M. J. (2008). Physiological heterogeneity in biofilms. Nature WHO. (2007). WHO Food safety and foodborne illness. World Health Organiza-
Reviews. Microbiology, 6(3), 199e210. tion, Retrieved 26.03.12, from. http://www.who.int/mediacentre/factsheets/
Stoodley, P., Cargo, R., Rupp, C. J., Wilson, S., & Klapper, I. (2002). Biofilm fs237/en/.
material properties as related to shear-induced deformation and detachment WHO. (n.d) WHO Foodborne diseases. World Health Organization. Retrieved
phenomena. Journal of Industrial Microbiology & Biotechnology, 29(6), 361e367. 26.03.12, from http://www.who.int/topics/foodborne_diseases/en/.
Stoodley, P., Sauer, K., Davies, D. G., & Costerton, J. W. (2002). Biofilms as complex Wiedmann, M., Weilmeier, D., Dineen, S. S., Ralyea, R., & Boor, K. J. (2000). Molecular
differentiated communities. Annual Review of Microbiology, 56, 187e209. and phenotypic characterization of Pseudomonas spp. isolated from milk.
Storgards, E., Simola, H., Sjöberg, A.-M., & Wirtanen, G. (1999). Hygiene of gasket Applied and Environmental Microbiology, 66(5), 2085e2095.
materials used in food processing equipment part 1: new materials. Food and Wijman, J. G. E., de Leeuw, P. P. L. A., Moezelaar, R., Zwietering, M. H., & Abee, T.
Bioproducts Processing, 77(2), 137e145. (2007). Air-liquid interface biofilms of Bacillus cereus: formation, sporulation,
Suslow, T. V. (2001). Water disinfection: A practical approach to calculating dose and dispersion. Applied and Environmental Microbiology, 73(5), 1481e1488.
values for preharvest and postharvest applications. Publication 7256. Agriculture Wong, A. C. L. (1998). Biofilms in food processing environments. Journal of Dairy
and Natural Resources, University of California. http://vric.ucdavis.edu. Science, 81(10), 2765e2770, Elsevier.
Tachikawa, M., Yamanaka, K., & Nakamuro, K. (2009). Studies on the disinfection Zeraik, A. E., & Nitschke, M. (2010). Biosurfactants as agents to reduce adhesion of
and removal of biofilms by ozone water using an artificial microbial biofilm pathogenic bacteria to polystyrene surfaces: effect of temperature and hydro-
system. Ozone: Science & Engineering, 31(1), 3e9. phobicity. Current Microbiology, 61(6), 554e559.

You might also like