Next Article in Journal
Food Safety Knowledge among Professional Food Handlers in Slovenia: The Results of a Nation-Wide Survey
Previous Article in Journal
Storage Quality Changes in Craft and Industrial Blueberry, Strawberry, Raspberry and Passion Fruit-Mango Sorbets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bacteriophages for the Targeted Control of Foodborne Pathogens

1
Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 1M4, Canada
2
Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, AB T1J 4B1, Canada
3
Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
4
Food and Human Nutritional Sciences, Faculty of Agricultural & Food Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
*
Author to whom correspondence should be addressed.
Foods 2023, 12(14), 2734; https://doi.org/10.3390/foods12142734
Submission received: 14 June 2023 / Revised: 5 July 2023 / Accepted: 14 July 2023 / Published: 18 July 2023
(This article belongs to the Section Food Quality and Safety)

Abstract

:
Foodborne illness is exacerbated by novel and emerging pathotypes, persistent contamination, antimicrobial resistance, an ever-changing environment, and the complexity of food production systems. Sporadic and outbreak events of common foodborne pathogens like Shiga toxigenic E. coli (STEC), Salmonella, Campylobacter, and Listeria monocytogenes are increasingly identified. Methods of controlling human infections linked with food products are essential to improve food safety and public health and to avoid economic losses associated with contaminated food product recalls and litigations. Bacteriophages (phages) are an attractive additional weapon in the ongoing search for preventative measures to improve food safety and public health. However, like all other antimicrobial interventions that are being employed in food production systems, phages are not a panacea to all food safety challenges. Therefore, while phage-based biocontrol can be promising in combating foodborne pathogens, their antibacterial spectrum is generally narrower than most antibiotics. The emergence of phage-insensitive single-cell variants and the formulation of effective cocktails are some of the challenges faced by phage-based biocontrol methods. This review examines phage-based applications at critical control points in food production systems with an emphasis on when and where they can be successfully applied at production and processing levels. Shortcomings associated with phage-based control measures are outlined together with strategies that can be applied to improve phage utility for current and future applications in food safety.

1. Introduction

The number of foodborne infections is ever-increasing despite legislative and microbiological prevention strategies to ensure food safety and public health. Emerging pathotypes, sanitizer-resistant microbes, and interconnections along the supply chain, such as production environment, contaminated processing equipment/surfaces, and asymptomatic food handlers, are considered major challenges contributing to an increase in foodborne infections [1]. Food-producing animals (cattle, swine, and chickens), leafy greens, soil, and water are the main bacterial sources of foodborne infections. The risks of transmission of foodborne diseases are influenced by environmental conditions such as temperature, moisture, and various agricultural and industrial practices (Figure 1).
Shiga toxigenic E. coli (STEC), Salmonella, Campylobacter, and Listeria monocytogenes are recognized as the most urgent threats in food-producing systems and are frequently associated with foodborne disease outbreaks. Recent examples include a Listeria outbreak in South Africa which recorded 728 patients and 193 mortalities [2]. The case–control and trace-back analysis suggested that polony, a ready-to-eat processed meat, was the source of infection [2]. In Canada, an E. coli O157 outbreak linked to romaine lettuce was reported and traced to the Yuma growing region in the United States [3]. This outbreak recorded eight cases of illnesses with no deaths. In China, a Salmonella outbreak was reported in Changzhi County [4]. Salmonella recovered from this outbreak was associated with a multidrug-resistant strain, resulting in 11 cases of illness but no deaths. In England, a Campylobacter outbreak was linked to the consumption of raw milk in 2016 [5], affecting sixty-nine individuals with no mortalities.
Various decontamination strategies such as the use of chemical sanitizers (sodium dichloroisocyanurate, quaternary ammonium compounds, chlorine, peracetic acid, and lactic acid), heat treatment (pasteurization), washing (water), and chilling are commonly used during food processing to reduce the risk of pathogens entering the food chain [6,7]. Bacteriophage-based interventions are being implemented as an additional hurdle or in synergy with these current interventions. For example, ListShield™ is a cocktail of bacteriophages that targets L. monocytogenes and is the first purified phage food additive used commercially to control this pathogen in ready-to-eat meat and poultry products and in food processing environments [8]. SalmoFreshTM, specific for Salmonella, can be used directly on poultry, fish and shellfish, and fresh and processed fruits and vegetables [9]. A comprehensive list of commercial bacteriophage (phage) products used for the biocontrol of foodborne pathogens in various foods has been reviewed [10]. These commercial phages are “customized combinations” that are of a narrow spectrum, given the vast genetic diversity of foodborne pathogens in different environments and food types. Regardless, the application of phages as biocontrol agents is promising in the food production system.
In this article, we review factors impacting cross-contamination, such as occurrence, prevalence levels, infective dose and persistence associated with foodborne pathogens, and the critical control points within the food production system where the risk of contamination is likely highest, with a focus on where and how bacteriophages can be successfully applied at the food production and processing levels.

2. Factors Impacting Cross-Contamination of Pathogens Occurrence, Virulence, and Pathogenicity

Shiga-toxigenic E. coli O157 (STEC) super-shedding incidents, where some cattle within a herd can shed >104 CFU/g in feces [11], can contribute to a high prevalence of the pathogen in production farms and subsequently in processing facilities. Compounded by a low infective dose (10–100 cells), STEC infections can cause life-threatening diseases such as hemolytic uremic syndrome (HUS), which can lead to kidney failure, especially in pregnant women, children, the elderly, and the immunocompromised [12]. STEC strains can survive harsh food processing conditions and serve as hot spots for persistent cross-contamination events in food processing environments. Although STEC O157:H7 has been the predominant serogroup associated with HUS worldwide, cases with non-O157 serogroups such as O26, O111, O103, O121, O45, and O145 have also been increasingly associated with foodborne illness, accounting for about 83% of all STEC infections in humans from 2000 to 2010 in the United States [13]. STEC strains can grow over a pH range of 4.4–10.0 [14] and temperatures ranging from 6 to 40 °C [15]. Some strains are heat-tolerant [16], whereas others have demonstrated strong biofilm-forming abilities on food processing surfaces like stainless steel [17,18] and polystyrene [19]. Thus, STEC O157 and a variety of non-O157 STEC strains are pathogens of priority, for which there is an urgent need to develop control strategies to reduce or completely prevent their adulteration of the food chain.
Listeria monocytogenes, the causative agent of human listeriosis, is associated with bacteremia, endocarditis, and infection of the central nervous system [20]. L. monocytogenes is a ubiquitous pathogen that thrives in diverse environments such as soil, water, various food products, and in the digestive tract of humans and animals [21,22]. It can survive and grow over a wide range of temperatures (2–45 °C) and pH (4.6–9.5) and in the presence of high salt concentrations [23,24]. Because of its ubiquitous distribution and robustness, there is a high chance that naturally persistent strains can be introduced into the food processing system through cross-contamination. Additionally, the ability of L. monocytogenes to form biofilms enables them to survive and persist on food processing surfaces [25]. The infectious dose of L. monocytogenes is not known but is likely lower in susceptible individuals like immunocompromised patients, the elderly, infants, and pregnant women than in healthy individuals [26]. In addition, L. monocytogenes may be a transient resident in the gastrointestinal tract of humans, with about 2–10% of human carriers lacking clinical symptoms [27]. These assertions are in line with L. monocytogenes outbreaks, during which the immunocompromised, neonates, and pregnant women are most likely to be infected [2,23,28].
Campylobacter jejuni is one of the causative agents of human campylobacteriosis characterized by mild diarrhea to serious conditions like bloody diarrhea, myocarditis, celiac disease, acute cholecystitis, and colorectal cancer [29]. Poultry meat contaminated with C. jejuni is the main route of infection in humans [30,31]. Unlike L. monocytogenes, which is stress resistant, C. jejuni is very susceptible to environmental stressors such as cold temperatures and acidic (pH of <4.9) conditions [31]. While these susceptibilities could be helpful in limiting the survival of C. jejuni and thus reducing the chances of cross-contamination, it has a high prevalence in poultry. C. jejuni is part of the normal microflora in the gastrointestinal tract of poultry, where prevalence can be 100% in broiler chickens at slaughter [32,33]. Furthermore, C. jejuni numbers are high (105–108 CFU/mL) in slaughterhouse carcass water [31], suggesting that cross-contamination of carcasses is inevitable. Additionally, C. jejuni may be well protected from harsh environmental conditions in biofilms [34]. The infectious dose of C. jejuni ranges from 400 to 106 cells in healthy young individuals and is strain-specific [35]. However, the infectious dose may be lower in susceptible/immunocompromised individuals with underlying conditions. Therefore, measures to reduce C. jejuni in processing plants are imperative to prevent human disease.
Salmonella Typhimurium, the causative agent of salmonellosis, is characterized by various clinical diseases such as gastroenteritis, enteric fever (typhoid fever), and bacteremia [36]. S. Typhimurium is a microorganism that thrives at a neutral pH. However, S. Typhimurium is capable of tolerating acidic (pH 3.3) conditions [37]. S. Typhimurium is widespread in nature and associated with cross-contamination in diverse food sources such as poultry [38], pork [39], beef [40], and fruits and vegetables [41]. The association with multiple food sources may enable it to circulate between different hosts, the environment, and humans through close contact or poor hygienic conditions during food handling. Salmonella can survive in low-moisture foods such as powdered milk, chocolate, peanut butter, infant foods, cereal, and bakery products over extended periods of time [42]. Salmonella is known to form biofilms on glass slides [43] and stainless steel [44,45]. The ability to form biofilms enhances the ability of Salmonella to survive in food-processing plants. The infectious dose of Salmonella is estimated to be 100 cells or higher [46] and can vary depending on the susceptibility of individuals.
Overall, it appears that high microbial load, persistence, infectious dose, and risk groups (susceptible individuals in the population) associated with the different pathogens directly correspond to cross-contamination events in the food production system. Although phenotypic variations such as the occurrence of persister cells within biofilms can impede control measures at any stage from production through to the processing plant, it is important to examine the random phenotypic diversity within a homogeneous single-cell population [47,48,49]. Phenotypic heterogeneity in a single-cell clonal population is known to confer resistance to antibiotics [50,51] as a result of the occurrence of two distinct subpopulations, a condition referred to as bi-stability [52]. These two subpopulations help ensure the survival of the population during environmental changes, a condition known as bet-hedging [53]. Therefore, to reduce the number of bacterial pathogens in the food production and processing environment, the possibility of single-cell phenotypic variants throughout the processing environment must be considered.

3. Bacteriophages

Bacteriophages (phages) are viruses that infect and kill bacteria. Some phages are host-specific, whereas others can infect multiple species or bacterial genera [54]. The host range of a phage determines the phage–host interaction events such as attachment, infection, and lysis, traits which are important in their application against target pathogen(s). For example, a narrow-spectrum phage is preferred for the control of a target pathogen in a live animal to avoid affecting commensal beneficial bacteria. In contrast, in food processing plants, a phage with a broad host range is preferred to efficiently reduce or eliminate the diversity of strains within a pathogenic species. It has been shown that under suitable conditions, a broad-spectrum phage can revert to a narrow state and vice versa [55]. Therefore, a cautious approach is required to delineate the phage host range [56], as well as employing phages with a broad or narrow host range for specific biocontrol applications.
Phages are ubiquitous in nature, have been isolated from food products like yogurt and cheese [57], and are naturally found in the human gut [58]. This suggests that humans encounter phages either directly or indirectly in daily life. Moreover, oral administration of phages is not known to cause disease in animals [59,60]. Also, because phages are ubiquitously distributed, they can provide an important diverse repertoire of control agents which can be exploited for control of pathogens in environments that possess differing selective pressures like temperature, salinity, pH, and UV radiation.
Lysogenic phages infect bacterial hosts, can integrate into the host genome, and alter the genotypic and phenotypic characteristics of the host. For example, the spread of Shiga toxin by E. coli O157:H7 is associated with a lambdoid bacteriophage [61], in which the toxin is only expressed when the lysogen becomes lytic during a stress response [49,62]. Lysogenic phages (lambdoid phage) are also being explored for phage-based DNA vaccine delivery in humans [63,64].
Lytic phages infect and kill their bacterial host through lysis. There is an ever-growing interest in lytic phages as control agents against targeted foodborne pathogens, including, Listeria, Salmonella, Campylobacter, and STEC [65,66,67,68]. The increasing interest in lytic phages as biocontrol agents follows the growing concerns with antibiotic-resistant bacteria [69,70] and an increased number of studies that are demonstrating the efficacy of phage therapy. Some of these are reflected in the recent increase in the number of phage clinical studies [71,72], along with the increased sequence-based characterization of phages through comparative genomics and proteomics [73,74,75].
Other than the biological traits of the phage, such as lysogenic or lytic capacity, various environmental factors may affect phage efficacy and viability. For example, external environmental conditions such as UV radiation, pH, temperature, and salt concentration can affect phage activity [76,77,78,79,80,81]. The level of inactivation depends on the intensity of these factors and may differ for different phages. For example, Iriarte, Balogh, Momol, Smith, Wilson, and Jones [76] found that phage populations associated with the surface of tomato leaves plummeted during the months when levels of UV radiation were the highest. Also, the activity of a lambdoid phage (λ-gt11) was reduced at extreme pHs of 11.8 or 2.0 but remained active after 24 h of exposure to pHs of 3–11 [82]. In contrast, the activity of a myovirus, JD007, was completely lost at a pH of 4 after 2 h of exposure at room temperature [83]. Some phages can survive temperature shifts; ≥75% of phages isolated from hot springs in California remained viable at 0 °C, whereas 18 to 30% remained intact at 105 °C [84]. Variations in phage viability are likely driven by the high genetic diversity among phages [81], geographic location, and experimental conditions. Therefore, getting a phage with the desired stable characteristic for target interventions requires both in vitro and field testing. Nevertheless, tailed phages such as T4 and T7 may be preferred, as shown by Ackermann et al. [85], as they remain stable over long periods (10–32 years) of refrigerated storage.
Phage–host exposure can result in phage-insensitive isolates [86]. However, it is possible that phage-insensitive strains are endemic within target pathogen populations as a result of previous exposure to environmental selective pressures. Phenotype-associated subpopulations, referred to as the phenotypic switch, have been reported in L. monocytogenes exposed to hostile environmental conditions [87]. The presence of phage-insensitive strains within a population is an important consideration for the development of targeted interventions in the phage arms race against foodborne pathogens.

4. Food-Production Animals, Leafy Green Produce, and Foodborne Pathogens

Food-production animals and plants are adapting to ever-changing environmental conditions like temperature, moisture, and availability of nutrients. The same applies to foodborne pathogens in the broader environment and in the gut of animals. Temperature, moisture, and availability of nutrients can facilitate pathogen growth, adaptation, and transmission to food-production animals through the food they eat or by wind, contaminated water, or when manure is used as a fertilizer to produce leafy greens. Unlike broad-spectrum antibiotics, which can be administered empirically [88,89] to control pathogens, no single phage preparation can control the entire range of pathogens that can potentially adulterate food. The formulation of phage cocktails has been proposed to be one possible solution to overcoming the limitation of a narrow host range [90]. However, developing a phage cocktail with consideration of the different mechanisms that can influence receptor binding to targeted cells requires an in-depth knowledge of phage diversity [91], as antagonistic interference can impact cocktail efficacy. Also, the phage latent period, the time required for a phage to induce host cell lysis [92], is important for cocktail concoction as a differential infection cycle of phages within a cocktail may select for the progeny of phages with the shortest latent period in the presence of a target pathogen defying the goal of the cocktail. As a result, even though phages offer the potential to control foodborne pathogens, optimizing phages for empiric application in livestock and food processing environments can be difficult and may require substantial research and development efforts. Therefore, target control of a pathogen would be highly preferred, with formulated cocktails possibly reducing the emergence of phage-resistant bacterial mutants as a result of their ability to target multiple receptors in pathogens [90].

4.1. Application of Phages for Target Control of Foodborne Pathogens in Food-Production Animals

The prevalence and persistence of foodborne pathogens on the hide and within the gut of food-producing animals on farms (pre-harvest) is the primary source of cross-contamination in the food chain. On-farm phage-based control strategies can successfully reduce target pathogen populations on and within livestock before slaughter [93,94]. Incidence and persistence of a target pathogen can vary among various animals, animals within a herd, or in different sites in the same animal. For example, C. jejuni is known to be prevalent in broiler chickens at slaughter, with all birds being carriers on some farms [32,33]. In cattle herds, up to 20% of the animals have been identified as E. coli O157 super-shedders [11,95]. Differentiating super-shedding cattle within a herd from transient shedders can be challenging and may require substantial sampling events over a long period [96] before treatment with phages. Various environments within the cattle gut, such as the rumen, cecum, colon, and rectum, may be colonized by a target pathogen, with the site within the digestive tract influencing the efficacy of phage-based interventions. Rumen physiological conditions such as pH can inactivate phages [97], whereas pathogen colonization and biofilm formation at the rectum may shelter targeted cells from phages [98]. Even though phage encapsulation has been shown to protect phages at a pH of 3.0 for 20 min of exposure [99], an in-depth understanding of the factors that affect phage stability from storage to delivery is required [100]. Individual phages or cocktails need to be optimized to infect targeted pathogens across various environments and in biofilms. Also, phenotypic variants which may arise within the target microbial population due to responses to environmental pressures in food-producing animals (Figure 2) can further compromise phage-based control strategies. Some subpopulations within the target population may survive phage treatment (Figure 3). The use of comparative genomics can differentiate, track, and provide valuable insight into plausible cell variants within a targeted pathogen population before phage application. This approach can also be used to select phages or concoct a phage cocktail with a broad host range to include phenotypic variants as part of the targeted pathogen population.
Several studies (n = 15) have explored the potential of phages to control E. coli O157:H7, Campylobacter, and Salmonella in food animals on farms (Table 1). In most studies, a single phage or a cocktail of phages was orally administered to the animal. In finisher broiler chickens, a ≥2 log reduction in Campylobacter or Salmonella at different phage dosages (106–1012 PFU/mL) was obtained (Table 1), which can contribute to a lower microbial load entering the processing stage. Except for the studies of Kittler et al. [101], Clavijo et al. [102], and Arthur et al. [103] that were performed in commercial farms and lairage targeting C. jejuni, Salmonella, and E. coli O157:H7, respectively, other studies (n = 12) concentrated on germ-free small-scaled in vivo controlled experiments. For example, the prevalence of bacteria among individuals in the natural environment is due to the level of natural exposure to the pathogen. In contrast, germ-free small-scaled controlled experiments deliver an equal dose to individuals at a preselected site within the digestive tract. Even though a challenge dose can be used to establish a baseline of phage-mediated pathogen reduction [104], differential pathogen colonization amongst individuals, differences in immune responses, nutrient availability, and the composition of the intestinal microbiome should be considered [105]. Second, a mid-log short-term culture (≥18 h) grown under optimal growth temperature (37 °C) on nutrient broth (Luria-Bertani or tryptic soy broth) administered in germ-free small-scaled controlled settings are likely to have traits that differ from environmental strains. Third, mid-log cultures from controlled settings may lack cell variants compared to environmental strains that are exposed to fluctuations in temperature and moisture. In this context, the efficacy of phage therapy is likely to be overestimated in controlled settings as mid-log growing cultures are known to be susceptible to phages [106] as compared to natural settings where cell variants may compromise phage efficacy.
Phage applications in either human therapeutic or agricultural sectors require a better comprehension of phage stability, viability, and survivability in diverse and hostile environments [81]. Except for three of the phages used in the experimental studies (Table 1), all were members of the Myoviridae (n = 9) or Siphoviridae (n = 3) of the order Caudovirales. Most phages were isolated from the target animal, and although their selection may be biased due to their ease of isolation and lytic potential, they have been shown to be stable and remain infective within the target animal during treatment. Some phages from the abovementioned families are known to be highly resistant to adverse environments, such as in desert surface sand exposed to high and low temperatures [107]. Extreme environments such as this may be a rich source of phages for the biocontrol of pathogens under a variety of food processing conditions.
Most studies that have used phages to control L. monocytogenes in food animals have focused on ready-to-eat beef, pork, and poultry. These foods typically undergo a period of cold storage which can enrich L. monocytogenes if it is present [20]. Moreover, a case–control study by Nightingale et al. [108] showed that L. monocytogenes can also proliferate in cattle, as levels were much higher in feces than in feed or the farm environment. Silage has been shown to be a common source of L. monocytogenes and targeting this source with phages may be an effective means of preventing on-farm transmission. Targeting feed can be more cost-effective as opposed to the direct control of L. monocytogenes in the host animal. However, the phage/cocktail should be selected with respect to its ability to withstand a wide range of temperatures and pH for a maximum efficacy that encompasses both the conditions in silage and those in the intestinal tract of the host animal.
Table 1. A summary of studies of bacteriophages used to control foodborne pathogens in/on food animals.
Table 1. A summary of studies of bacteriophages used to control foodborne pathogens in/on food animals.
Target AnimalTarget BacteriaPhage/FamilyPhage/MixturePhage DosePhage Delivery RouteSettingsEfficacyReference
24-day-old broiler chickenC. jejuniCP20 and CP30A/MyoviridaeCocktail107 PFU/mLOralIn vivo controlledA reduction of up to 2.4 log10 CFU/g 2 days post-treatment[94]
9-day-old broiler chickenC. jejuniNCTC12673, 12674, 12678, and 12672/Nd1Single and cocktail107 PFU/mLOralIn vivo controlledA 2.8 log10 CFU/g reduction 21 days post-treatment[109]
36-day-old commercial broiler chickenC. jejuniNCTC12672, 12673, 12674, and 12678/MyoviridaeCocktail7.2 and 7.9 PFU/mLOralCommercial farmA 3.2 log10 CFU/g reduction at slaughter[101]
25-day-old broiler chickenC. jejuniCP220/MyoviridaeSingle107 and 109 PFU/mLOralIn vivo controlledA 2.0 log10 CFU/g reduction 2 days post-treatment[110]
SheepE. coli O157:H7CEV1 and CEV2/SiphoviridaeSingle and cocktail1011 PFU/mLOralIn vivo controlledCocktail had 99.9% reduction compared to 99% in single 2 days post inoculation[111]
One-day-old Ross broiler chicksC. coli and C. jejuniphiCcoIBB35, phiCcoIBB37 and phiCcoIBB12/MyoviridaeCocktail106 or 107 PFU/mLOralCommercial farmA 2.0 log10 CFU/g reduction 2 days post-treatment[112]
16-month- and 8–9-year-old cattleE. coli O157:H7e11/2 and e4/1c/MyoviridaeCocktail1011 PFU/mLOralIn vivo controlledNo significant difference compared to control 2 days post inoculation[113]
≥1-year-old cattle (steer)E. coli O157:H7rV5, wV7, wV8, and wV11/Nd1Cocktail1010 PFU/bolus and 1011 PFU/feed Oral bolus or phage mixed in cattle feedIn vivo controlledThe duration of shedding was reduced by 14 days in bolus-fed steers as compared with control steers, but phage did not reduce E. coli O157:H7 shedding overall[99]
Cattle before passing through the lairageE. coli O157:H7Finalyse®/Nd1Cocktail 1010 PFU/gallon of waterSprayed on hide Commercial farmNo significant reduction after 3 days of application compared to control[103]
CattleE. coli O157:H7rV5, wV7, wV8, and wV11/MyoviridaeCocktail1011 PFU/mLOral and rectalIn vivo controlledNo significant difference compared to control over 83 days post inoculation[114]
Six-month-old Holstein steersE. coli O157:H7KH1 and SH1/Nd1Cocktail1011 PFU/mLRecto-anal junctionIn vivo controlledReduction in the average number of E. coli O157:H7 among phage-treated steers compared to control steers[115]
38-day-old broiler chickenS. enterica serotypes Enteritidis, Typhimurium, and Hadarϕ151/Myoviridae, ϕ10, and ϕ25/SiphoviridaeSingle109 or 1011 PFU/mLOralIn vivo controlledPhage ϕ151 had a 4.2 log10 CFU/g reduction 1 day post-treatment for both S. Enteritidis and Typhimurium. Phage ϕ10, a 2.19 log10 CFU/g reduction for S. Typhimurium.
No reduction by ϕ25 on Hadar
[116]
18-day-old commercial broiler chickenSalmonellaSalmoFREE® (φ San15, φ San23, φ San24, and φ San25)/MyoviridaeCocktail108 PFU/mLOralIn vivo controlled100% reduction on day 33 post-treatment compared to control[102]
One-day-old broiler chickenS. TyphimuriumΦst1/SiphoviridaeSingle1010 or 1012 PFU/mLIntracloacalIn vivo controlled100% reduction after 1 day post-treatment compared to control[117]
4-day-old broiler chickenS. enterica serotype EnteritidisCNPSA1, CNPSA3, and CNPSA4/Nd1Single1011 PFU/mLOralIn vivo controlledA reduction of 3.5 orders of magnitude of CFU/g 5 days post treatment[118]
Nd1 = not determined in the studies analyzed.

4.2. Application of Phages for Controlling Foodborne Pathogens in Leafy Green Vegetables at Pre-Harvest Level

Vegetables are the main sources of plant fiber, minerals, and vitamins necessary for a nutritious and healthy diet. Like food-producing animals, leafy green vegetables (LGVs) such as spinach, lettuce, cabbage, and Swiss chard are potential vehicles of foodborne pathogens. Most LGVs are consumed fresh, hence increasing the risk that they may be a transmission source of pathogens involved in foodborne outbreaks [119]. STEC O157, Salmonella, Campylobacter, and L. monocytogenes are often associated with foodborne disease outbreaks through LGVs [120,121,122,123,124,125]. Contaminated farmland, manure, and irrigation water contaminated with feces from livestock and wildlife are the primary sources of LGV contamination at the farm [126,127]. Phage-based control targeting these sources of contamination has the potential to reduce the risk of LGVs being contaminated with pathogens.
To our knowledge, there is no evidence of a commercial field-scale experiment using phages for the pre-harvest control of STEC O157, Salmonella, Campylobacter, or L. monocytogenes in LGVs. LGVs are usually grown in large areas and are prone to various plausible sources of contamination, like contaminated farmland, irrigation water, and animal feces through runoff waters. Also, identifying a contaminated area of the farm and pathogen location on or in the LGVs can be challenging for localized and target application of phages [128]. Finally, spraying the entire farm with phages using irrigation water as a carrier is presently economically infeasible due to the large quantities of phages that would be required.

5. Food-Processing Environment

According to Rosenquist et al. [129], the incidence of Campylobacter-related illnesses linked with the consumption of contaminated food could be reduced 30 times if the number of pathogens on food-animal carcasses was decreased by 2 log during processing. In addition to the potential contamination of meat and LVGs during harvest, pathogens can also originate from the food processing environment, poor food handling, and contaminated food contact surfaces. Using phage/cocktails to kill pathogens in the food processing environment and on food contact surfaces before and after food processing could substantially reduce the risk of food contamination.
Several studies have used phages to control STEC O157 and non-O157, Salmonella, Campylobacter, and L. monocytogenes in food processing environments. Reinhard et al. [130] used a Listeria-specific phage to successfully reduce Listeria populations in a processing plant (Table 2). To our knowledge, data using this approach to control resident environmental foodborne pathogens are limited. It is possible that resident foodborne pathogens in the processing plant on non-food contact surfaces can contaminate food products through the generation of aerosols [131]. Control measures that target resident pathogen strains are necessary because they can lower the pathogen heterogeneity at the processing level and subsequent contaminations. Although controlling foodborne pathogens on non-food contact surfaces can be promising, it can be laborious and challenging as to where and how the phage/cocktail can be applied. This is because the applied phage/cocktail needs to encounter all surfaces within the processing plant. Also, differential colonization and growth state of pathogens within the plant may require different concentrations of phages or a cocktail for effective control. For example, it is known that actively growing bacterial cells in small numbers are eliminated by phages, whereas larger colonies survive and retain a mixture of sensitive and resistant variants [106]. Likewise, a better understanding of the host density and the extent of host–phage interaction that influences the degree of post-treatment phage proliferation is required. A chemotactic-based, target-specific engineered phage could be a means of overcoming conditions where phage efficacy is dependent on random bacteria–phage contact.
Experimentally contaminated food/food surface studies indicate that foodborne pathogens can form mono-species biofilms on food contact surfaces and that a population reduction of 1–5.4 log10 CFU/cm2 at varied temperature conditions is achievable using a single phage type or cocktails (Table 2). These findings indicate the potential of using phage/cocktails to reduce mono-species biofilm cells on food contact surfaces. However, biofilm formation in an experimentally controlled setting may not necessarily reflect that in processing plants. Also, incompletely eradicated biofilm cells can regrow after treatment [132], resulting in downstream contamination. Multiple bacterial species [133], nutrient availability [134], and temperature [135] are factors that can affect biofilm formation and vary between experimentally controlled studies and the natural environment. Therefore, their role in biofilm formation by a foodborne pathogen on food surfaces in processing plants needs to be clearly understood before phage control measures are employed. Also, a biofilm architecture consisting of an exopolysaccharide matrix and multiple bacterial layers can confer resistance [136] by affecting phage diffusion within the biofilms. This is contrary to the expectation that because phages are non-motile, they will be more efficient killers of localized than dispersed bacteria cells, as phage progeny can easily infect nearby cells. One way that phages can gain access to cells within biofilms is through the production of depolymerases [137,138] that can degrade the biofilm matrix. Another challenge to phage efficacy in biofilms is that persister cells, which are metabolically inactive within biofilms, lack the resources like amino acids, ATP, polymerases, and ribosomes that phages require for replication [139]. In this context, phages can infect these non-growing cells but cannot produce progeny for further infection of additional target cells. In a scenario where non-growing cells are infected by phages, it can be dubbed ‘held at gunpoint’, as the reactivation of cell growth is on approach to triggering phage activity [140]. Though phage-based application is a potential method for controlling foodborne pathogens, identifying and optimizing phages for target eradication of biofilms may call for substantial research and development efforts. Genetically engineered phages that are specifically developed to combat biofilms may be integral to the success of phage therapy in food production environments [141].
Table 2. Summarized studies on bacteriophages used to control foodborne pathogens in food processing environments and on food contact surfaces.
Table 2. Summarized studies on bacteriophages used to control foodborne pathogens in food processing environments and on food contact surfaces.
Environment or Surface TypeTarget BacteriaPhage/FamilyPhage/MixturePhage DoseBacteria DoseMode of ApplicationTemperature ConditionEfficacyReference
Ready-to-eat food manufacturing area (door frames/seals, floors/curbing, wheels/casters, walls/windows/curbing, drains, catch pan, water pipe, freezer doors/door seals, etc.)L. monocytogenesPhageGuard Listex™Cocktail 107 and 108 PFU/mLNot indicatedSpraying4 and 20 °CModerate application of 107 resulted in a 66% reduction in listeria prevalence at both 4 and 20 °C, whereas at concentration 108, a reduction of 43 and 32% was obtained at 4 and 20 °C, respectively.[130]
GlassC. jejuni (NCTC 11168 and PT14)CP8 and CP30Single106 or 109 PFU/mL105 CFU/mL (initial cells for biofilm formation)Spot inoculation37 °CA 1 to 3 log10 CFU/cm2 reduction 24 h after phage treatment compared with control.[142]
Stainless steel and polyurethane thermoplastic beltingCocktail of L. monocytogenes and L. innocuaP100Cocktail107 and 108 PFU/cm2104–105 CFU/cm2Spot inoculation4 and 20 °COverall, a reduction of 1.27–3.33 and 1.17–2.76 log10 CFU/cm2 on stainless steel and polyurethane thermoplastic belting, respectively, with a higher reduction at a high phage dilution of 108.[143]
Spinach harvester bladeCocktail of E. coli O157:H7Phages not specifiedCocktail108 PFU/mL105–106 CFU/mLSpraying22 °CReduction in biofilm populations by 4.5 log10 CFU on blades after 2 h of phage treatment.[144]
Stainless steelL. monocytogenes (19CO9, 19DO3 and 19EO3)LiMN4L, LiMN4p, and LiMN17Single or cocktail109 PFU/mL108 CFU/mLImmersion15 °CSingle phages reduced biofilm cells by 3–4.5 log units and cocktail by 3.8–5.4 and log10 CFU/cm2.[65]
Stainless steel, rubber, and MBEC biofilm devicesS. Enteritidis (ATCC13076) and S. Typhimurium (ATCC14028)BP 1369 and BP 1370/Myoviridae and Podoviridae, respectively.Single108 PFU/mLInitial inoculum of 105 CFU/mLImmersion10 and 30 °CA reduction in biofilm cells by 3.0, 2.0, and 3.0 log CFU/cm2 on stainless steel, rubber, and an MBEC device.[145]
Stainless steel chips, ceramic tile chips, and high-density polyethylene chipsCocktail of O157:H7 (EK27, ATCC 43895, and 472)BEC8Cocktail106 PFU/mL106, 105, and 104 CFU/chipSpot inoculation4, 12, 23, and 37 °CNo biofilm survivors were detected (detection limit 10 CFU/chip) after 1 h of treatment at 12, 23, and 37 °C.[146]
Polystyrene and stainless steelS. EnteritidisPVP-SESingleMOIs (0.1, 1, and 10)104 CFU/mLImmersion4
and 22
°C
A 2–5 log10 CFU/cm2 reduction with a higher killing efficiency at room temperature.[147]
Stainless steelE. coli O113:H21 and O154:H10SA21RBSingle1013 PFU/mL106 and 105 CFU/mL, respectivelyImmersion22
°C
A reduction in biofilm cells by 2.5 and 2.1 log10 CFU/cm2 for O113:H21 and O154:H10, respectively, for 24 h biofilm after 3 h of phage treatment.[68]
Polystyrene microplateS. EnteritidisCW1, CW11, M4, and M10CocktailNot indicated102 CFU/mL for developing biofilm. Mature biofilm (48 h) number not indicatedImmersion37 °CA reduction in cells in the developing biofilm and mature biofilm by 0.79 and 0.4 log10 CFU/cm2, respectively.[148]
Food product types, available nutrients, moisture content, and temperature abuse during food processing can promote pathogen proliferation, increasing the risk of further pathogen presence in ready-to-eat (RTE) foods. Control interventions on food products can reduce colonization and the downstream flow of foodborne pathogens. Chlorine is commonly used to reduce pathogens during the wash stage in food processing plants. However, there are concerns that chlorine can inactivate phages [143]. In contrast, Ding et al. [149] found that residual chlorine on lettuce leaves did not reduce the lytic potential of E. coli O157:H7 phages. This may suggest that the effect of residual chlorine on phages needs to be evaluated before both approaches are employed within food processing environments. The isolation and identification of sanitizer-resistant phages could facilitate the synergistic application of chlorine and phages. Also, the application of phages on food/food contact surfaces before the wash step is important for the desired outcome [130].
A number of experimental studies have used phages to control STEC O157 and non-O157, Salmonella, Campylobacter, and L. monocytogenes in food products and leafy green vegetables (Table 3). These results from both commercial and non-commercial phages are promising, with log reduction (0.39–4.54) after phage applications at different temperatures (−20 to 30 °C), dosages (107–1010 PFU/mL), bacterial densities (low level 102–104 CFU/mL, and high level 105–107 CFU/mL), time points, application method, and duration of phage exposure (5 min to 7 days). These are important factors for consideration during phage treatment. For example, phage-resistant mutant strains can develop after a longer exposure time [150], whereas phage effectiveness can also be affected by shorter exposure times if it has a long infection cycle. Spraying phages on food is generally less effective than if the food is immersed in a phage solution [151]. This response has been attributed to increased Brownian motion with immersion than spraying. Second, phage efficacy is influenced by the host–concentration [106] and is also temperature-dependent [152], with a greater reduction in the numbers of target bacteria when they are present at lower concentrations and at 21 °C than at 4 °C. However, phage activity has been reported to be active against Salmonella associated with chicken meat at −20 °C within the first 24 h of storage [153]. This suggests that phages need to be isolated and characterized for use at a specific temperature to optimize their efficacy against targeted pathogens. Additional factors, including experimental design, food type, bacterial host, and phage type, may influence the efficacy of the phage, requiring a full understanding of these factors in relation to the targeted pathogen.

6. Future Perspective and Conclusions

For a successful and sustainable strategy, phages should be used for customized treatments, as suggested by Torres-Barceló [169]. Here we propose the ‘niche-specific’ use of phages in the food processing system for effective control of pathogens, bearing in mind that a phage/cocktail against a target pathogen across the food chain may not be suitable in different niches such as the animal, plant, or food processing environments. With the knowledge of the targeted pathogen and the nature of its variants in specific environments, niche-specific phages or cocktails could be formulated for spraying or immersion and oral or topical application. More commercial field-scale studies are required, even though they are more difficult and costly than in vivo laboratory experiments, to affirm the efficacy of phages in more real-world food production systems. Such a strategy could pave the way for effective target-specific phage formulation as an additional processing aid to the ongoing efforts to further improve food safety.

Author Contributions

Conceptualization, E.W.B., K.S., R.Z., D.N., T.A., C.N.-B. and T.A.M.; writing—original draft preparation, E.W.B.; writing—review and editing, K.S., R.Z., D.N., T.A., C.N.-B. and T.A.M.; supervision, K.S., R.Z., D.N., T.A., C.N.-B. and T.A.M.; funding acquisition, K.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Results-Driven Agricultural Research (RDAR) project 2021R1014R.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Newell, D.G.; Koopmans, M.; Verhoef, L.; Duizer, E.; Aidara-Kane, A.; Sprong, H.; Opsteegh, M.; Langelaar, M.; Threfall, J.; Scheutz, F. Food-borne diseases—The challenges of 20 years ago still persist while new ones continue to emerge. Int. J. Food Microbiol. 2010, 139, S3–S15. [Google Scholar] [CrossRef] [PubMed]
  2. Thomas, J.; Govender, N.; McCarthy, K.M.; Erasmus, L.K.; Doyle, T.J.; Allam, M.; Ismail, A.; Ramalwa, N.; Sekwadi, P.; Ntshoe, G. Outbreak of listeriosis in South Africa associated with processed meat. N. Engl. J. Med. 2020, 382, 632–643. [Google Scholar] [CrossRef]
  3. Health Canada. Outbreak of E. coli Infections Linked to Romaine Lettuce. 2018. Available online: https://www.canada.ca/en/public-health/services/public-health-notices/2018/public-health-notice-outbreak-e-coli-infections-linked-romaine-lettuce.html (accessed on 1 April 2023).
  4. Xiang, Y.; Li, F.; Dong, N.; Tian, S.; Zhang, H.; Du, X.; Zhou, X.; Xu, X.; Yang, H.; Xie, J. Investigation of a salmonellosis outbreak caused by multidrug resistant Salmonella Typhimurium in China. Front. Microbiol. 2020, 11, 801. [Google Scholar] [CrossRef] [PubMed]
  5. Kenyon, J.; Inns, T.; Aird, H.; Swift, C.; Astbury, J.; Forester, E.; Decraene, V. Campylobacter outbreak associated with raw drinking milk, North West England, 2016. Epidemiol. Infect. 2020, 148, e13. [Google Scholar] [CrossRef] [Green Version]
  6. Acuff, G. Chemical decontamination strategies for meat. In Improving the Safety of Fresh Meat; Sofos, J.N., Ed.; CRC Press: Boca Raton, FL, USA, 2005; pp. 350–363. [Google Scholar]
  7. Anderson, N.M. Recent advances in low moisture food pasteurization. Curr. Opin. Food Sci. 2019, 29, 109–115. [Google Scholar] [CrossRef]
  8. FDA. Food additives permitted for direct addition to food for human consumption: Bacteriophage preparation. Fed. Regist. 2006, 71, 47729–47732. [Google Scholar]
  9. FDA. Intralytix Wins Regulatory Approval For Phage-Based Food Safety Product Effective Against Salmonella. 2013. Available online: http://www.intralytix.com (accessed on 17 April 2023).
  10. Alomari, M.M.M.; Dec, M.; Urban-Chmiel, R. Bacteriophages as an alternative method for control of zoonotic and foodborne pathogens. Viruses 2021, 13, 2348. [Google Scholar] [CrossRef]
  11. Matthews, L.; Low, J.; Gally, D.; Pearce, M.; Mellor, D.; Heesterbeek, J.; Chase-Topping, M.; Naylor, S.; Shaw, D.; Reid, S. Heterogeneous shedding of Escherichia coli O157 in cattle and its implications for control. Proc. Natl. Acad. Sci. USA 2006, 103, 547–552. [Google Scholar] [CrossRef]
  12. Kaper, J.B.; Nataro, J.P.; Mobley, H.L. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2004, 2, 123–140. [Google Scholar] [CrossRef]
  13. Gould, L.H.; Mody, R.K.; Ong, K.L.; Clogher, P.; Cronquist, A.B.; Garman, K.N.; Lathrop, S.; Medus, C.; Spina, N.L.; Webb, T.H. Increased recognition of non-O157 Shiga toxin–producing Escherichia coli infections in the United States during 2000–2010: Epidemiologic features and comparison with E. coli O157 infections. Foodborne Pathog. Dis. 2013, 10, 453–460. [Google Scholar] [CrossRef]
  14. Desmarchelier, P.; Fegan, N. Enteropathogenic Escherichia coli. In Foodborne Microorganisms of Public Health Signiicance, 6th ed.; Hocking, A.D., Ed.; Australian Institute of Food Science and Technology (NSW Branch): Sydney, Australia, 2003; pp. 267–310. [Google Scholar]
  15. Kim, C.; Ndegwa, E. Influence of pH and temperature on growth characteristics of leading foodborne pathogens in a laboratory medium and select food beverages. Austin Food Sci. 2018, 3, 1031. [Google Scholar]
  16. Zhang, P.; Tran, F.; Stanford, K.; Yang, X. Are antimicrobial interventions associated with heat-resistant Escherichia coli on meat? Appl. Environ. Microbiol. 2020, 86, e00512–e00520. [Google Scholar] [CrossRef] [PubMed]
  17. Ma, Z.; Bumunang, E.W.; Stanford, K.; Bie, X.; Niu, Y.D.; McAllister, T.A. Biofilm Formation by Shiga Toxin-Producing Escherichia coli on Stainless Steel Coupons as Affected by Temperature and Incubation Time. Microorganisms 2019, 7, 95. [Google Scholar] [CrossRef] [Green Version]
  18. Bumunang, E.; Ateba, C.; Stanford, K.; McAllister, T.A.; Niu, Y.D. Biofilm formation by South-African Shiga toxigenic non-O157 Escherichia coli on stainless steel coupons. Can. J. Microbiol. 2020, 66, 328–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Bumunang, E.W.; McAllister, T.A.; King, R.; Ortega Polo, R.; Stanford, K.; Zaheer, R.; Niu, Y.D.; Ateba, C.N. Characterization of Non-O157 Escherichia coli from Cattle Faecal Samples in the North-West Province of South Africa. Microorganisms 2019, 7, 272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Farber, J.M.; Peterkin, P. Listeria monocytogenes, a food-borne pathogen. Microbiol. Rev. 1991, 55, 476–511. [Google Scholar] [CrossRef]
  21. Vivant, A.-L.; Garmyn, D.; Piveteau, P. Listeria monocytogenes, a down-to-earth pathogen. Front. Cell. Infect. Microbiol. 2013, 3, 87. [Google Scholar] [CrossRef] [Green Version]
  22. Sleator, R.D.; Watson, D.; Hill, C.; Gahan, C.G. The interaction between Listeria monocytogenes and the host gastrointestinal tract. Microbiology 2009, 155, 2463–2475. [Google Scholar] [CrossRef]
  23. Swaminathan, B.; Gerner-Smidt, P. The epidemiology of human listeriosis. Microbes Infect. 2007, 9, 1236–1243. [Google Scholar] [CrossRef] [Green Version]
  24. Gandhi, M.; Chikindas, M.L. Listeria: A foodborne pathogen that knows how to survive. Int. J. Food Microbiol. 2007, 113, 1–15. [Google Scholar] [CrossRef]
  25. Dygico, L.K.; Gahan, C.G.; Grogan, H.; Burgess, C.M. The ability of Listeria monocytogenes to form biofilm on surfaces relevant to the mushroom production environment. Int. J. Food Microbiol. 2020, 317, 108385. [Google Scholar] [CrossRef] [PubMed]
  26. Pouillot, R.; Klontz, K.C.; Chen, Y.; Burall, L.S.; Macarisin, D.; Doyle, M.; Bally, K.M.; Strain, E.; Datta, A.R.; Hammack, T.S. Infectious dose of Listeria monocytogenes in outbreak linked to ice cream, United States, 2015. Emerg. Infect. Dis. 2016, 22, 2113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Buchanan, R.L.; Gorris, L.G.; Hayman, M.M.; Jackson, T.C.; Whiting, R.C. A review of Listeria monocytogenes: An update on outbreaks, virulence, dose-response, ecology, and risk assessments. Food Control 2017, 75, 1–13. [Google Scholar] [CrossRef]
  28. Goulet, V.; Hebert, M.; Hedberg, C.; Laurent, E.; Vaillant, V.; De Valk, H.; Desenclos, J.-C. Incidence of listeriosis and related mortality among groups at risk of acquiring listeriosis. Clin. Infect. Dis. 2012, 54, 652–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Kaakoush, N.O.; Castaño-Rodríguez, N.; Mitchell, H.M.; Man, S.M. Global epidemiology of Campylobacter infection. Clin. Microbiol. Rev. 2015, 28, 687–720. [Google Scholar] [CrossRef] [Green Version]
  30. Acheson, D.; Allos, B.M. Campylobacter jejuni infections: Update on emerging issues and trends. Clin. Infect. Dis. 2001, 32, 1201–1206. [Google Scholar] [CrossRef]
  31. Keener, K.; Bashor, M.; Curtis, P.; Sheldon, B.; Kathariou, S. Comprehensive review of Campylobacter and poultry processing. Compr. Rev. Food Sci. Food Saf. 2004, 3, 105–116. [Google Scholar] [CrossRef]
  32. Newell, D.; Fearnley, C. Sources of Campylobacter colonization in broiler chickens. Appl. Environ. Microbiol. 2003, 69, 4343–4351. [Google Scholar] [CrossRef] [Green Version]
  33. Sahin, O.; Kassem, I.I.; Shen, Z.; Lin, J.; Rajashekara, G.; Zhang, Q. Campylobacter in poultry: Ecology and potential interventions. Avian Dis. 2015, 59, 185–200. [Google Scholar] [CrossRef]
  34. Teh, A.H.T.; Lee, S.M.; Dykes, G.A. Does Campylobacter jejuni form biofilms in food-related environments? Appl. Environ. Microbiol. 2014, 80, 5154–5160. [Google Scholar] [CrossRef] [Green Version]
  35. Black, R.E.; Levine, M.M.; Clements, M.L.; Hughes, T.P.; Blaser, M.J. Experimental Campylobacter jejuni infection in humans. J. Infect. Dis. 1988, 157, 472–479. [Google Scholar] [CrossRef]
  36. Foley, S.L.; Johnson, T.J.; Ricke, S.C.; Nayak, R.; Danzeisen, J. Salmonella pathogenicity and host adaptation in chicken-associated serovars. Microbiol. Mol. Biol. Rev. 2013, 77, 582–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Foster, J.W.; Hall, H.K. Adaptive acidification tolerance response of Salmonella typhimurium. J. Bacteriol. 1990, 172, 771–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Humphrey, T. Public-health aspects of Salmonella infection. Salmonella Domest. Anim. 2000, 1, 245–263. [Google Scholar]
  39. Bonardi, S. Salmonella in the pork production chain and its impact on human health in the European Union. Epidemiol. Infect. 2017, 145, 1513–1526. [Google Scholar] [CrossRef] [Green Version]
  40. Besser, T.; Goldoft, M.; Pritchett, L.; Khakhria, R.; Hancock, D.; Rice, D.; Gay, J.; Johnson, W.; Gay, C. Multiresistant Salmonella Typhimurium DT104 infections of humans and domestic animals in the Pacific Northwest of the United States. Epidemiol. Infect. 2000, 124, 193–200. [Google Scholar] [CrossRef]
  41. Krishnasamy, V.P.; Marshall, K.; Dewey-Mattia, D.; Wise, M. Outbreak characteristics and epidemic curves for multistate outbreaks of Salmonella infections associated with produce: United States, 2009–2015. Foodborne Pathog. Dis. 2020, 17, 15–22. [Google Scholar] [CrossRef]
  42. Podolak, R.; Enache, E.; Stone, W.; Black, D.G.; Elliott, P.H. Sources and risk factors for contamination, survival, persistence, and heat resistance of Salmonella in low-moisture foods. J. Food Prot. 2010, 73, 1919–1936. [Google Scholar] [CrossRef]
  43. Niu, T.-X.; Wang, X.-N.; Wu, H.-Y.; Bi, J.-R.; Hao, H.-S.; Hou, H.-M.; Zhang, G.-L. Transcriptomic analysis, motility and biofilm formation characteristics of Salmonella Typhimurium exposed to benzyl isothiocyanate treatment. Int. J. Mol. Sci. 2020, 21, 1025. [Google Scholar] [CrossRef] [Green Version]
  44. Nguyen, H.; Yang, Y.; Yuk, H. Biofilm formation of Salmonella Typhimurium on stainless steel and acrylic surfaces as affected by temperature and pH level. LWT-Food Sci. Technol. 2014, 55, 383–388. [Google Scholar] [CrossRef]
  45. Ma, Z.; Tang, X.; Stanford, K.; Chen, X.; McAllister, T.A.; Niu, Y.D. Single-and Dual-Species Biofilm Formation by Shiga Toxin-Producing Escherichia coli and Salmonella, and Their Susceptibility to an Engineered Peptide WK2. Microorganisms 2021, 9, 2510. [Google Scholar] [CrossRef] [PubMed]
  46. Teunis, P.F.; Kasuga, F.; Fazil, A.; Ogden, I.D.; Rotariu, O.; Strachan, N.J. Dose–response modeling of Salmonella using outbreak data. Int. J. Food Microbiol. 2010, 144, 243–249. [Google Scholar] [CrossRef] [PubMed]
  47. Ackermann, M. A functional perspective on phenotypic heterogeneity in microorganisms. Nat. Rev. Microbiol. 2015, 13, 497–508. [Google Scholar] [CrossRef]
  48. Abee, T.; Koomen, J.; Metselaar, K.; Zwietering, M.; Den Besten, H. Impact of pathogen population heterogeneity and stress-resistant variants on food safety. Annu. Rev. Food Sci. Technol. 2016, 7, 439–456. [Google Scholar] [CrossRef] [PubMed]
  49. Weigel, W.; Dersch, P. Phenotypic heterogeneity: A bacterial virulence strategy. Microbes Infect. 2018, 20, 570–577. [Google Scholar] [CrossRef]
  50. Wakamoto, Y.; Dhar, N.; Chait, R.; Schneider, K.; Signorino-Gelo, F.; Leibler, S.; McKinney, J.D. Dynamic persistence of antibiotic-stressed mycobacteria. Science 2013, 339, 91–95. [Google Scholar] [CrossRef] [PubMed]
  51. Arnoldini, M.; Vizcarra, I.A.; Peña-Miller, R.; Stocker, N.; Diard, M.; Vogel, V.; Beardmore, R.E.; Hardt, W.-D.; Ackermann, M. Bistable expression of virulence genes in Salmonella leads to the formation of an antibiotic-tolerant subpopulation. PLoS Biol. 2014, 12, e1001928. [Google Scholar] [CrossRef]
  52. Dubnau, D.; Losick, R. Bistability in bacteria. Mol. Microbiol. 2006, 61, 564–572. [Google Scholar] [CrossRef]
  53. Veening, J.-W.; Smits, W.K.; Kuipers, O.P. Bistability, epigenetics, and bet-hedging in bacteria. Annu. Rev. Microbiol. 2008, 62, 193–210. [Google Scholar] [CrossRef] [Green Version]
  54. Ross, A.; Ward, S.; Hyman, P. More is better: Selecting for broad host range bacteriophages. Front Microbiol. 2016, 7, 1352. [Google Scholar] [CrossRef] [Green Version]
  55. Meyer, J.R.; Dobias, D.T.; Medina, S.J.; Servilio, L.; Gupta, A.; Lenski, R.E. Ecological speciation of bacteriophage lambda in allopatry and sympatry. Science 2016, 354, 1301–1304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. de Jonge, P.A.; Nobrega, F.L.; Brouns, S.J.; Dutilh, B.E. Molecular and evolutionary determinants of bacteriophage host range. Trends Microbiol. 2019, 27, 51–63. [Google Scholar] [CrossRef] [PubMed]
  57. Suárez, V.; Quiberoni, A.; Binetti, A.; Reinheimer, J. Thermophilic lactic acid bacteria phages isolated from Argentinian dairy industries. J. Food Prot. 2002, 65, 1597–1604. [Google Scholar] [CrossRef] [PubMed]
  58. Shkoporov, A.N.; Hill, C. Bacteriophages of the human gut: The “known unknown” of the microbiome. Cell Host Microbe 2019, 25, 195–209. [Google Scholar] [CrossRef] [Green Version]
  59. Mirzaei, M.K.; Maurice, C.F. Ménage à trois in the human gut: Interactions between host, bacteria and phages. Nat. Rev. Microbiol. 2017, 15, 397–408. [Google Scholar] [CrossRef] [PubMed]
  60. Gangwar, M.; Rastogi, S.; Singh, D.; Shukla, A.; Dhameja, N.; Kumar, D.; Kumar, R.; Nath, G. Study on the effect of oral administration of bacteriophages in charles foster rats with special reference to immunological and adverse effects. Front. Pharmacol. 2021, 12, 615445. [Google Scholar] [CrossRef]
  61. Hayashi, T.; Makino, K.; Ohnishi, M.; Kurokawa, K.; Ishii, K.; Yokoyama, K.; Han, C.-G.; Ohtsubo, E.; Nakayama, K.; Murata, T. Complete genome sequence of enterohemorrhagic Eschelichia coli O157: H7 and genomic comparison with a laboratory strain K-12. DNA Res. 2001, 8, 11–22. [Google Scholar] [CrossRef] [Green Version]
  62. Mauro, S.A.; Koudelka, G.B. Shiga toxin: Expression, distribution, and its role in the environment. Toxins 2011, 3, 608–625. [Google Scholar] [CrossRef]
  63. Ghaemi, A.; Soleimanjahi, H.; Gill, P.; Hassan, Z.; Jahromi, S.R.M.; Roohvand, F. Recombinant λ-phage nanobioparticles for tumor therapy in mice models. Genet. Vaccines Ther. 2010, 8, 3. [Google Scholar] [CrossRef] [Green Version]
  64. Clark, J.R.; Bartley, K.; Jepson, C.D.; Craik, V.; March, J.B. Comparison of a bacteriophage-delivered DNA vaccine and a commercially available recombinant protein vaccine against hepatitis B. FEMS Immunol. Med. Microbiol. 2011, 61, 197–204. [Google Scholar] [CrossRef] [Green Version]
  65. Arachchi, G.J.G.; Cridge, A.G.; Dias-Wanigasekera, B.M.; Cruz, C.D.; McIntyre, L.; Liu, R.; Flint, S.H.; Mutukumira, A.N. Effectiveness of phages in the decontamination of Listeria monocytogenes adhered to clean stainless steel, stainless steel coated with fish protein, and as a biofilm. J. Ind. Microbiol. Biotechnol. 2013, 40, 1105–1116. [Google Scholar] [CrossRef] [PubMed]
  66. Yeh, Y.; Purushothaman, P.; Gupta, N.; Ragnone, M.; Verma, S.; De Mello, A. Bacteriophage application on red meats and poultry: Effects on Salmonella population in final ground products. Meat Sci. 2017, 127, 30–34. [Google Scholar] [CrossRef] [PubMed]
  67. Zampara, A.; Sørensen, M.C.H.; Elsser-Gravesen, A.; Brøndsted, L. Significance of phage-host interactions for biocontrol of Campylobacter jejuni in food. Food Control 2017, 73, 1169–1175. [Google Scholar] [CrossRef]
  68. Bumunang, E.W.; Ateba, C.N.; Stanford, K.; Niu, Y.D.; Wang, Y.; McAllister, T.A. Activity of bacteriophage and complex tannins against biofilm-forming shiga toxin-producing Escherichia coli from Canada and South Africa. Antibiotics 2020, 9, 257. [Google Scholar] [CrossRef] [PubMed]
  69. Kutter, E.M.; Kuhl, S.J.; Abedon, S.T. Re-establishing a place for phage therapy in western medicine. Future Microbiol. 2015, 10, 685–688. [Google Scholar] [CrossRef] [Green Version]
  70. Vikram, A.; Woolston, J.; Sulakvelidze, A. Phage biocontrol applications in food production and processing. Curr. Issues. Mol. Biol. 2021, 40, 267–302. [Google Scholar] [CrossRef]
  71. McCallin, S.; Sacher, J.C.; Zheng, J.; Chan, B.K. Current state of compassionate phage therapy. Viruses 2019, 11, 343. [Google Scholar] [CrossRef] [Green Version]
  72. Sacher, J.C.; Zheng, J. Phage therapy collaboration and compassionate use. In Bacteriophages: Biology, Technology, Therapy; Harper, D.R., Abedon, S.T., Burrowes, B.H., McConville, M., Eds.; Springer Nature Switzerland AG: New York, NY, USA, 2021; pp. 1069–1098. [Google Scholar]
  73. Niu, Y.D.; McAllister, T.A.; Nash, J.H.; Kropinski, A.M.; Stanford, K. Four Escherichia coli O157: H7 phages: A new bacteriophage genus and taxonomic classification of T1-like phages. PLoS ONE 2014, 9, e100426. [Google Scholar] [CrossRef] [Green Version]
  74. Korf, I.H.; Meier-Kolthoff, J.P.; Adriaenssens, E.M.; Kropinski, A.M.; Nimtz, M.; Rohde, M.; van Raaij, M.J.; Wittmann, J. Still Something to Discover: Novel Insights into Escherichia coli Phage Diversity and Taxonomy. Viruses 2019, 11, 454. [Google Scholar] [CrossRef] [Green Version]
  75. Bumunang, E.W.; McAllister, T.A.; Polo, R.O.; Ateba, C.N.; Stanford, K.; Schlechte, J.; Walker, M.; MacLean, K.; Niu, Y.D. Genomic Profiling of Non-O157 Shiga Toxigenic Escherichia coli-Infecting Bacteriophages from South Africa. PHAGE Ther. Appl. Res. 2022, 3, 221–230. [Google Scholar] [CrossRef]
  76. Iriarte, F.; Balogh, B.; Momol, M.; Smith, L.; Wilson, M.; Jones, J. Factors affecting survival of bacteriophage on tomato leaf surfaces. Appl. Environ. Microbiol. 2007, 73, 1704–1711. [Google Scholar] [CrossRef] [Green Version]
  77. Sinton, L.W.; Hall, C.H.; Lynch, P.A.; Davies-Colley, R.J. Sunlight inactivation of fecal indicator bacteria and bacteriophages from waste stabilization pond effluent in fresh and saline waters. Appl. Environ. Microbiol. 2002, 68, 1122–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Silva, Y.J.; Costa, L.; Pereira, C.; Cunha, Â.; Calado, R.; Gomes, N.C.; Almeida, A. Influence of environmental variables in the efficiency of phage therapy in aquaculture. Microb. Biotechnol. 2014, 7, 401–413. [Google Scholar] [CrossRef] [PubMed]
  79. Branston, S.D.; Stanley, E.C.; Ward, J.M.; Keshavarz-Moore, E. Determination of the survival of bacteriophage M13 from chemical and physical challenges to assist in its sustainable bioprocessing. Biotechnol. Bioprocess Eng. 2013, 18, 560–566. [Google Scholar] [CrossRef]
  80. Yamaki, S.; Omachi, T.; Kawai, Y.; Yamazaki, K. Characterization of a novel Morganella morganii bacteriophage FSP1 isolated from river water. FEMS Microbiol. Lett. 2014, 359, 166–172. [Google Scholar] [CrossRef] [Green Version]
  81. Jończyk, E.; Kłak, M.; Międzybrodzki, R.; Górski, A. The influence of external factors on bacteriophages. Folia Microbiol. 2011, 56, 191–200. [Google Scholar] [CrossRef] [Green Version]
  82. Jepson, C.D.; March, J.B. Bacteriophage lambda is a highly stable DNA vaccine delivery vehicle. Vaccine 2004, 22, 2413–2419. [Google Scholar] [CrossRef]
  83. Cui, Z.; Feng, T.; Gu, F.; Li, Q.; Dong, K.; Zhang, Y.; Zhu, Y.; Han, L.; Qin, J.; Guo, X. Characterization and complete genome of the virulent Myoviridae phage JD007 active against a variety of Staphylococcus aureus isolates from different hospitals in Shanghai, China. Virol. J. 2017, 14, 1–8. [Google Scholar] [CrossRef] [Green Version]
  84. Breitbart, M.; Wegley, L.; Leeds, S.; Schoenfeld, T.; Rohwer, F. Phage community dynamics in hot springs. Appl. Environ. Microbiol. 2004, 70, 1633–1640. [Google Scholar] [CrossRef] [Green Version]
  85. Ackermann, H.-W.; Tremblay, D.; Moineau, S. Long-term bacteriophage preservation. WFCC Newsl. 2004, 38, 35–40. [Google Scholar]
  86. Fister, S.; Stessl, B.; Fuchs, S.; Schoder, D.; Wagner, M.; Rossmanith, P. Screening and characterisation of bacteriophage P100 insensitive Listeria monocytogenes isolates in Austrian dairy plants. Food Control 2016, 59, 108–117. [Google Scholar] [CrossRef]
  87. Metselaar, K.I.; den Besten, H.M.; Abee, T.; Moezelaar, R.; Zwietering, M.H. Isolation and quantification of highly acid resistant variants of Listeria monocytogenes. Int. J. Food Microbiol. 2013, 166, 508–514. [Google Scholar] [CrossRef]
  88. Hatchette, T.F.; Farina, D. Infectious diarrhea: When to test and when to treat. Can. Med Assoc. J. 2011, 183, 339–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. DuPont, H.L. Persistent diarrhea: A clinical review. JAMA 2016, 315, 2712–2723. [Google Scholar] [CrossRef] [PubMed]
  90. Malik, D.J.; Sokolov, I.J.; Vinner, G.K.; Mancuso, F.; Cinquerrui, S.; Vladisavljevic, G.T.; Clokie, M.R.; Garton, N.J.; Stapley, A.G.; Kirpichnikova, A. Formulation, stabilisation and encapsulation of bacteriophage for phage therapy. Adv. Colloid Interface Sci. 2017, 249, 100–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Niu, Y.D.; Liu, H.; Du, H.; Meng, R.; Sayed Mahmoud, E.; Wang, G.; McAllister, T.A.; Stanford, K. Efficacy of individual bacteriophages does not predict efficacy of bacteriophage cocktails for control of Escherichia coli O157. Front. Microbiol. 2021, 12, 616712. [Google Scholar] [CrossRef]
  92. Abedon, S.T.; Herschler, T.D.; Stopar, D. Bacteriophage latent-period evolution as a response to resource availability. Appl. Environ. Microbiol. 2001, 67, 4233–4241. [Google Scholar] [CrossRef] [Green Version]
  93. Newell, D.; Elvers, K.; Dopfer, D.; Hansson, I.; Jones, P.; James, S.; Gittins, J.; Stern, N.; Davies, R.; Connerton, I. Biosecurity-based interventions and strategies to reduce Campylobacter spp. on poultry farms. Appl. Environ. Microbiol. 2011, 77, 8605–8614. [Google Scholar] [CrossRef] [Green Version]
  94. Richards, P.J.; Connerton, P.L.; Connerton, I.F. Phage biocontrol of Campylobacter jejuni in chickens does not produce collateral effects on the gut microbiota. Front. Microbiol. 2019, 10, 476. [Google Scholar] [CrossRef]
  95. Chen, S.; Sanderson, M.; Lanzas, C. Investigating effects of between-and within-host variability on Escherichia coli O157 shedding pattern and transmission. Prev. Vet. Med. 2013, 109, 47–57. [Google Scholar] [CrossRef]
  96. Williams, K.; Ward, M.; Dhungyel, O.; Hall, E.; Van Breda, L. A longitudinal study of the prevalence and super-shedding of Escherichia coli O157 in dairy heifers. Vet. Microbiol. 2014, 173, 101–109. [Google Scholar] [CrossRef]
  97. Smith, H.W.; Huggins, M.B.; Shaw, K.M. Factors influencing the survival and multiplication of bacteriophages in calves and in their environment. Microbiology 1987, 133, 1127–1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Naylor, S.W.; Low, J.C.; Besser, T.E.; Mahajan, A.; Gunn, G.J.; Pearce, M.C.; McKendrick, I.J.; Smith, D.G.; Gally, D.L. Lymphoid follicle-dense mucosa at the terminal rectum is the principal site of colonization of enterohemorrhagic Escherichia coli O157: H7 in the bovine host. Infect. Immun. 2003, 71, 1505–1512. [Google Scholar] [CrossRef] [Green Version]
  99. Stanford, K.; McAllister, T.; Niu, Y.; Stephens, T.; Mazzocco, A.; Waddell, T.; Johnson, R. Oral delivery systems for encapsulated bacteriophages targeted at Escherichia coli O157: H7 in feedlot cattle. J. Food Prot. 2010, 73, 1304–1312. [Google Scholar] [CrossRef] [PubMed]
  100. Yang, Y.; Du, H.; Zou, G.; Song, Z.; Zhou, Y.; Li, H.; Tan, C.; Chen, H.; Fischetti, V.A.; Li, J. Encapsulation and delivery of phage as a novel method for gut flora manipulation in situ: A review. J. Control. Release 2023, 353, 634–649. [Google Scholar] [CrossRef] [PubMed]
  101. Kittler, S.; Fischer, S.; Abdulmawjood, A.; Glünder, G.; Klein, G. Effect of bacteriophage application on Campylobacter jejuni loads in commercial broiler flocks. Appl. Environ. Microbiol. 2013, 79, 7525–7533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Clavijo, V.; Baquero, D.; Hernandez, S.; Farfan, J.; Arias, J.; Arévalo, A.; Donado-Godoy, P.; Vives-Flores, M. Phage cocktail SalmoFREE® reduces Salmonella on a commercial broiler farm. Poult. Sci. 2019, 98, 5054–5063. [Google Scholar] [CrossRef]
  103. Arthur, T.M.; Kalchayanand, N.; Agga, G.E.; Wheeler, T.L.; Koohmaraie, M. Evaluation of bacteriophage application to cattle in lairage at beef processing plants to reduce Escherichia coli O157: H7 prevalence on hides and carcasses. Foodborne Pathog. Dis. 2017, 14, 17–22. [Google Scholar] [CrossRef] [Green Version]
  104. Grant, A.Q.; Parveen, S.; Schwarz, J.; Hashem, F.; Vimini, B. Reduction of Salmonella in ground chicken using a bacteriophage. Poult. Sci. 2017, 96, 2845–2852. [Google Scholar] [CrossRef]
  105. Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Núñez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [Google Scholar] [CrossRef]
  106. Eriksen, R.S.; Svenningsen, S.L.; Sneppen, K.; Mitarai, N. A growing microcolony can survive and support persistent propagation of virulent phages. Proc. Natl. Acad. Sci. USA 2018, 115, 337–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Prigent, M.; Leroy, M.; Confalonieri, F.; Dutertre, M.; DuBow, M.S. A diversity of bacteriophage forms and genomes can be isolated from the surface sands of the Sahara Desert. Extremophiles 2005, 9, 289–296. [Google Scholar] [CrossRef] [PubMed]
  108. Nightingale, K.; Schukken, Y.; Nightingale, C.; Fortes, E.; Ho, A.; Her, Z.; Grohn, Y.; McDonough, P.; Wiedmann, M. Ecology and transmission of Listeria monocytogenes infecting ruminants and in the farm environment. Appl. Environ. Microbiol. 2004, 70, 4458–4467. [Google Scholar] [CrossRef] [Green Version]
  109. Fischer, S.; Kittler, S.; Klein, G.; Glünder, G. Impact of a single phage and a phage cocktail application in broilers on reduction of Campylobacter jejuni and development of resistance. PLoS ONE 2013, 8, e78543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. El-Shibiny, A.; Scott, A.; Timms, A.; Metawea, Y.; Connerton, P.; Connerton, I. Application of a group II Campylobacter bacteriophage to reduce strains of Campylobacter jejuni and Campylobacter coli colonizing broiler chickens. J. Food Prot. 2009, 72, 733–740. [Google Scholar] [CrossRef] [Green Version]
  111. Raya, R.R.; Oot, R.A.; Moore-Maley, B.; Wieland, S.; Callaway, T.R.; Kutter, E.M.; Brabban, A.D. Naturally resident and exogenously applied T4-like and T5-like bacteriophages can reduce Escherichia coli O157: H7 levels in sheep guts. Bacteriophage 2011, 1, 15–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Carvalho, C.M.; Gannon, B.W.; Halfhide, D.E.; Santos, S.B.; Hayes, C.M.; Roe, J.M.; Azeredo, J. The in vivo efficacy of two administration routes of a phage cocktail to reduce numbers of Campylobacter coli and Campylobacter jejuni in chickens. BMC Microbiol. 2010, 10, 232. [Google Scholar] [CrossRef] [Green Version]
  113. Rivas, L.; Coffey, B.; McAuliffe, O.; McDonnell, M.J.; Burgess, C.M.; Coffey, A.; Ross, R.P.; Duffy, G. In vivo and ex vivo evaluations of bacteriophages e11/2 and e4/1c for use in the control of Escherichia coli O157: H7. Appl. Environ. Microbiol. 2010, 76, 7210–7216. [Google Scholar] [CrossRef] [Green Version]
  114. Rozema, E.A.; Stephens, T.P.; Bach, S.J.; Okine, E.K.; Johnson, R.P.; Stanford, K.; McAllister, T.A. Oral and rectal administration of bacteriophages for control of Escherichia coli O157: H7 in feedlot cattle. J. Food Prot. 2009, 72, 241–250. [Google Scholar] [CrossRef]
  115. Sheng, H.; Knecht, H.J.; Kudva, I.T.; Hovde, C.J. Application of bacteriophages to control intestinal Escherichia coli O157: H7 levels in ruminants. Appl. Environ. Microbiol. 2006, 72, 5359–5366. [Google Scholar] [CrossRef] [Green Version]
  116. Atterbury, R.J.; Van Bergen, M.; Ortiz, F.; Lovell, M.; Harris, J.; De Boer, A.; Wagenaar, J.; Allen, V.; Barrow, P. Bacteriophage therapy to reduce Salmonella colonization of broiler chickens. Appl. Environ. Microbiol. 2007, 73, 4543–4549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Wong, C.L.; Sieo, C.C.; Tan, W.S.; Abdullah, N.; Hair-Bejo, M.; Abu, J.; Ho, Y.W. Evaluation of a lytic bacteriophage, Φ st1, for biocontrol of Salmonella enterica serovar Typhimurium in chickens. Int. J. Food Microbiol. 2014, 172, 92–101. [Google Scholar] [CrossRef]
  118. Fiorentin, L.; Vieira, N.D.; Barioni, W., Jr. Oral treatment with bacteriophages reduces the concentration of Salmonella Enteritidis PT4 in caecal contents of broilers. Avian Pathol. 2005, 34, 258–263. [Google Scholar] [CrossRef] [PubMed]
  119. Mogren, L.; Windstam, S.; Boqvist, S.; Vågsholm, I.; Söderqvist, K.; Rosberg, A.K.; Lindén, J.; Mulaosmanovic, E.; Karlsson, M.; Uhlig, E. The hurdle approach–A holistic concept for controlling food safety risks associated with pathogenic bacterial contamination of leafy green vegetables. A review. Front Microbiol. 2018, 9, 1965. [Google Scholar] [CrossRef] [Green Version]
  120. Heiman, K.E.; Mody, R.K.; Johnson, S.D.; Griffin, P.M.; Gould, L.H. Escherichia coli O157 outbreaks in the United States, 2003–2012. Emerg. Infect. Dis. 2015, 21, 1293. [Google Scholar] [CrossRef] [PubMed]
  121. Bennett, S.; Sodha, S.; Ayers, T.; Lynch, M.; Gould, L.; Tauxe, R. Produce-associated foodborne disease outbreaks, USA, 1998–2013. Epidemiol. Infect. 2018, 146, 1397–1406. [Google Scholar] [CrossRef] [Green Version]
  122. Vestrheim, D.; Lange, H.; Nygård, K.; Borgen, K.; Wester, A.; Kvarme, M.; Vold, L. Are ready-to-eat salads ready to eat? An outbreak of Salmonella Coeln linked to imported, mixed, pre-washed and bagged salad, Norway, November 2013. Epidemiol. Infect. 2016, 144, 1756–1760. [Google Scholar] [CrossRef] [Green Version]
  123. Self, J.L.; Conrad, A.; Stroika, S.; Jackson, A.; Burnworth, L.; Beal, J.; Wellman, A.; Jackson, K.A.; Bidol, S.; Gerhardt, T. Outbreak of listeriosis associated with consumption of packaged salad—United States and Canada, 2015–2016. MMWR Morb. Mortal. Wkly. Rep. 2016, 65, 879–881. [Google Scholar] [CrossRef] [Green Version]
  124. Callejón, R.M.; Rodríguez-Naranjo, M.I.; Ubeda, C.; Hornedo-Ortega, R.; Garcia-Parrilla, M.C.; Troncoso, A.M. Reported foodborne outbreaks due to fresh produce in the United States and European Union: Trends and causes. Foodborne Pathog. Dis. 2015, 12, 32–38. [Google Scholar] [CrossRef]
  125. Gardner, T.J.; Fitzgerald, C.; Xavier, C.; Klein, R.; Pruckler, J.; Stroika, S.; McLaughlin, J.B. Outbreak of campylobacteriosis associated with consumption of raw peas. Clin. Infect. Dis. 2011, 53, 26–32. [Google Scholar] [CrossRef] [Green Version]
  126. Liu, C.; Hofstra, N.; Franz, E. Impacts of climate change on the microbial safety of pre-harvest leafy green vegetables as indicated by Escherichia coli O157 and Salmonella spp. Int. J. Food Microbiol. 2013, 163, 119–128. [Google Scholar] [CrossRef]
  127. Garcia, B.C.B.; Dimasupil, M.A.Z.; Vital, P.G.; Widmer, K.W.; Rivera, W.L. Fecal contamination in irrigation water and microbial quality of vegetable primary production in urban farms of Metro Manila, Philippines. J. Environ. Sci. Health Part B 2015, 50, 734–743. [Google Scholar] [CrossRef] [PubMed]
  128. Delaquis, P.; Bach, S.; Dinu, L.-D. Behavior of Escherichia coli O157: H7 in leafy vegetables. J. Food Prot. 2007, 70, 1966–1974. [Google Scholar] [CrossRef] [PubMed]
  129. Rosenquist, H.; Nielsen, N.L.; Sommer, H.M.; Nørrung, B.; Christensen, B.B. Quantitative risk assessment of human campylobacteriosis associated with thermophilic Campylobacter species in chickens. Int. J. Food Microbiol. 2003, 83, 87–103. [Google Scholar] [CrossRef] [PubMed]
  130. Reinhard, R.G.; Kalinowski, R.M.; Bodnaruk, P.W.; Eifert, J.D.; Boyer, R.R.; Duncan, S.E.; Bailey, R.H. Practical application of bacteriophage in food manufacturing facilities for the control of Listeria sp. J. Food Saf. 2020, 43, e12871. [Google Scholar] [CrossRef]
  131. Brown, K.; Wray, S. Control of airborne contamination in food processing. In Hygiene in Food Processing; Elsevier: Amsterdam, The Netherlands, 2014; pp. 174–202. [Google Scholar]
  132. Fauvart, M.; De Groote, V.N.; Michiels, J. Role of persister cells in chronic infections: Clinical relevance and perspectives on anti-persister therapies. J. Med. Microbiol. 2011, 60, 699–709. [Google Scholar] [CrossRef]
  133. Raghupathi, P.K.; Liu, W.; Sabbe, K.; Houf, K.; Burmølle, M.; Sørensen, S.J. Synergistic interactions within a multispecies biofilm enhance individual species protection against grazing by a pelagic protozoan. Front. Microbiol. 2018, 8, 2649. [Google Scholar] [CrossRef] [Green Version]
  134. Klausen, M.; Heydorn, A.; Ragas, P.; Lambertsen, L.; Aaes-Jørgensen, A.; Molin, S.; Tolker-Nielsen, T. Biofilm formation by Pseudomonas aeruginosa wild type, flagella and type IV pili mutants. Mol. Microbiol. 2003, 48, 1511–1524. [Google Scholar] [CrossRef]
  135. Wang, R.; Bono, J.L.; Kalchayanand, N.; Shackelford, S.; Harhay, D.M. Biofilm formation by Shiga toxin–producing Escherichia coli O157: H7 and Non-O157 strains and their tolerance to sanitizers commonly used in the food processing environment. J. Food Prot. 2012, 75, 1418–1428. [Google Scholar] [CrossRef] [Green Version]
  136. Nwodo, U.U.; Green, E.; Okoh, A.I. Bacterial exopolysaccharides: Functionality and prospects. Int. J. Mol. Sci. 2012, 13, 14002–14015. [Google Scholar] [CrossRef] [Green Version]
  137. Carson, L.; Gorman, S.P.; Gilmore, B.F. The use of lytic bacteriophages in the prevention and eradication of biofilms of Proteus mirabilis and Escherichia coli. FEMS Immunol Med. Microbiol. 2010, 59, 447–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Gutiérrez, D.; Briers, Y.; Rodríguez-Rubio, L.; Martínez, B.; Rodríguez, A.; Lavigne, R.; García, P. Role of the pre-neck appendage protein (Dpo7) from phage vB_SepiS-phiIPLA7 as an anti-biofilm agent in Staphylococcal species. Front. Microbiol. 2015, 6, 1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Wang, I.-N.; Dykhuizen, D.E.; Slobodkin, L.B. The evolution of phage lysis timing. Evol. Ecol. 1996, 10, 545–558. [Google Scholar] [CrossRef]
  140. Harper, D.R.; Parracho, H.M.; Walker, J.; Sharp, R.; Hughes, G.; Werthén, M.; Lehman, S.; Morales, S. Bacteriophages and biofilms. Antibiotics 2014, 3, 270–284. [Google Scholar] [CrossRef] [Green Version]
  141. Azeredo, J.; Sutherland, I.W. The use of phages for the removal of infectious biofilms. Curr. Pharm. Biotechnol. 2008, 9, 261–266. [Google Scholar] [CrossRef] [PubMed]
  142. Siringan, P.; Connerton, P.L.; Payne, R.J.; Connerton, I.F. Bacteriophage-mediated dispersal of Campylobacter jejuni biofilms. Appl. Environ. Microbiol. 2011, 77, 3320–3326. [Google Scholar] [CrossRef] [Green Version]
  143. Reinhard, R.G.; Kalinowski, R.M.; Bodnaruk, P.W.; Eifert, J.D.; Boyer, R.R.; Duncan, S.E.; Bailey, R.H. Fate of Listeria on various food contact and noncontact surfaces when treated with bacteriophage. J. Food Saf. 2020, 40, e12775. [Google Scholar] [CrossRef]
  144. Patel, J.; Sharma, M.; Millner, P.; Calaway, T.; Singh, M. Inactivation of Escherichia coli O157: H7 attached to spinach harvester blade using bacteriophage. Foodborne Pathog. Dis. 2011, 8, 541–546. [Google Scholar] [CrossRef] [Green Version]
  145. Sadekuzzaman, M.; Mizan, M.F.R.; Yang, S.; Kim, H.-S.; Ha, S.-D. Application of bacteriophages for the inactivation of Salmonella spp. in biofilms. Food Sci. Technol. Int. 2018, 24, 424–433. [Google Scholar] [CrossRef]
  146. Viazis, S.; Akhtar, M.; Feirtag, J.; Diez-Gonzalez, F. Reduction of Escherichia coli O157: H7 viability on hard surfaces by treatment with a bacteriophage mixture. Int. J. Food Microbiol. 2011, 145, 37–42. [Google Scholar] [CrossRef]
  147. Milho, C.; Silva, M.D.; Melo, L.; Santos, S.; Azeredo, J.; Sillankorva, S. Control of Salmonella Enteritidis on food contact surfaces with bacteriophage PVP-SE2. Biofouling 2018, 34, 753–768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Ning, Z.; Zhang, L.; Cai, L.; Xu, X.; Chen, Y.; Wang, H. Biofilm removal mediated by Salmonella phages from chicken-related sources. Food Sci. Hum. Wellness 2023, 12, 1799–1808. [Google Scholar] [CrossRef]
  149. Ding, Y.; Nan, Y.; Qiu, Y.; Niu, D.; Stanford, K.; Holley, R.; McAllister, T.; Narváez-Bravo, C. Use of a phage cocktail to reduce the numbers of seven Escherichia coli strains belonging to different STEC serogroups applied to fresh produce and seeds. J. Food Saf. 2023; early view. [Google Scholar] [CrossRef]
  150. Sillankorva, S.; Neubauer, P.; Azeredo, J. Pseudomonas fluorescens biofilms subjected to phage phiIBB-PF7A. BMC Biotechnol. 2008, 8, 79. [Google Scholar] [CrossRef] [Green Version]
  151. Jassim, S.; Abdulamir, A.; Abu Bakar, F. Novel phage-based bio-processing of pathogenic Escherichia coli and its biofilms. World J. Microbiol. Biotechnol. 2012, 28, 47–60. [Google Scholar] [CrossRef]
  152. Hong, Y.; Schmidt, K.; Marks, D.; Hatter, S.; Marshall, A.; Albino, L.; Ebner, P. Treatment of Salmonella-contaminated eggs and pork with a broad-spectrum, single bacteriophage: Assessment of efficacy and resistance development. Foodborne Pathog. Dis. 2016, 13, 679–688. [Google Scholar] [CrossRef]
  153. Abhisingha, M.; Dumnil, J.; Pitaksutheepong, C. Efficiency of phage cocktail to reduce Salmonella Typhimurium on chicken meat during low temperature storage. LWT-Food Sci. Technol. 2020, 129, 109580. [Google Scholar] [CrossRef]
  154. Witte, S.; Huijboom, L.; Klamert, S.; van de Straat, L.; Hagens, S.; Fieseler, L.; de Vegt, B.T.; van Mierlo, J.T. Application of bacteriophages EP75 and EP335 efficiently reduces viable cell counts of Escherichia coli O157 on beef and vegetables. Food Microbiol. 2022, 104, 103978. [Google Scholar] [CrossRef] [PubMed]
  155. Gencay, Y.E.; Ayaz, N.D.; Copuroglu, G.; Erol, I. Biocontrol of Shiga Toxigenic Escherichia coli O157: H7 in Turkish raw meatball by bacteriophage. J. Food Saf. 2016, 36, 120–131. [Google Scholar] [CrossRef]
  156. Carter, C.D.; Parks, A.; Abuladze, T.; Li, M.; Woolston, J.; Magnone, J.; Senecal, A.; Kropinski, A.M.; Sulakvelidze, A. Bacteriophage cocktail significantly reduces Escherichia coli O157: H7 contamination of lettuce and beef, but does not protect against recontamination. Bacteriophage 2012, 2, 178–185. [Google Scholar] [CrossRef] [Green Version]
  157. Duc, H.M.; Son, H.M.; Yi, H.P.S.; Sato, J.; Ngan, P.H.; Masuda, Y.; Honjoh, K.; Miyamoto, T. Isolation, characterization and application of a polyvalent phage capable of controlling Salmonella and Escherichia coli O157: H7 in different food matrices. Food Res. Int. 2020, 131, 108977. [Google Scholar] [CrossRef]
  158. Shebs-Maurine, E.; Giotto, F.; Laidler, S.; de Mello, A. Effects of bacteriophages and peroxyacetic acid applications on beef contaminated with Salmonella during different grinding stages. Meat Sci. 2021, 173, 108407. [Google Scholar] [CrossRef] [PubMed]
  159. Sukumaran, A.T.; Nannapaneni, R.; Kiess, A.; Sharma, C.S. Reduction of Salmonella on chicken meat and chicken skin by combined or sequential application of lytic bacteriophage with chemical antimicrobials. Int. J. Food Microbiol. 2015, 207, 8–15. [Google Scholar] [CrossRef] [PubMed]
  160. Ishaq, A.; Ebner, P.D.; Syed, Q.A.; ur Rahman, H.U. Employing list-shield bacteriophage as a bio-control intervention for Listeria monocytogenes from raw beef surface and maintain meat quality during refrigeration storage. LWT-Food Sci. Technol. 2020, 132, 109784. [Google Scholar] [CrossRef]
  161. Zhou, C.; Zhu, M.; Wang, Y.; Yang, Z.; Ye, M.; Wu, L.; Bao, H.; Pang, M.; Zhou, Y.; Wang, R. Broad host range phage vB-LmoM-SH3-3 reduces the risk of Listeria contamination in two types of ready-to-eat food. Food Control 2020, 108, 106830. [Google Scholar] [CrossRef]
  162. Yang, S.; Sadekuzzaman, M.; Ha, S.-D. Reduction of Listeria monocytogenes on chicken breasts by combined treatment with UV-C light and bacteriophage ListShield. LWT-Food Sci. Technol. 2017, 86, 193–200. [Google Scholar] [CrossRef]
  163. Chibeu, A.; Agius, L.; Gao, A.; Sabour, P.M.; Kropinski, A.M.; Balamurugan, S. Efficacy of bacteriophage LISTEX™ P100 combined with chemical antimicrobials in reducing Listeria monocytogenes in cooked turkey and roast beef. Int. J. Food Microbiol. 2013, 167, 208–214. [Google Scholar] [CrossRef]
  164. Orquera, S.; Gölz, G.; Hertwig, S.; Hammerl, J.; Sparborth, D.; Joldic, A.; Alter, T. Control of Campylobacter spp. and Yersinia enterocolitica by virulent bacteriophages. J. Mol. Genet. Med. 2012, 6, 273. [Google Scholar] [CrossRef] [Green Version]
  165. Bigwood, T.; Hudson, J.; Billington, C.; Carey-Smith, G.; Heinemann, J. Phage inactivation of foodborne pathogens on cooked and raw meat. Food Microbiol. 2008, 25, 400–406. [Google Scholar] [CrossRef]
  166. Firlieyanti, A.S.; Connerton, P.L.; Connerton, I.F. Campylobacters and their bacteriophages from chicken liver: The prospect for phage biocontrol. Int. J. Food Microbiol. 2016, 237, 121–127. [Google Scholar] [CrossRef]
  167. Lu, Y.T.; Ma, Y.; Wong, C.W.; Wang, S. Characterization and application of bacteriophages for the biocontrol of Shiga-toxin producing Escherichia coli in Romaine lettuce. Food Control 2022, 140, 109109. [Google Scholar] [CrossRef]
  168. Zhang, X.; Niu, Y.D.; Nan, Y.; Stanford, K.; Holley, R.; McAllister, T.; Narváez-Bravo, C. SalmoFresh™ effectiveness in controlling Salmonella on romaine lettuce, mung bean sprouts and seeds. Int. J. Food Microbiol. 2019, 305, 108250. [Google Scholar] [CrossRef] [PubMed]
  169. Torres-Barceló, C. Phage therapy faces evolutionary challenges. Viruses 2018, 10, 323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Factors that influence the risk of transmission of foodborne pathogens into the food chain.
Figure 1. Factors that influence the risk of transmission of foodborne pathogens into the food chain.
Foods 12 02734 g001
Figure 2. (A) Single-cell heterogeneity within food-production animal/produce (B) and a diverse pathogen population in processing environments. These diverse populations may arise due to response to environmental selective pressures and from cross-contamination events.
Figure 2. (A) Single-cell heterogeneity within food-production animal/produce (B) and a diverse pathogen population in processing environments. These diverse populations may arise due to response to environmental selective pressures and from cross-contamination events.
Foods 12 02734 g002
Figure 3. (A) Phage interaction with phenotypic variants of a pathogen within food-production animal/produce; (B) phage treatment of a pathogen population in a processing environment; and (C) combining pathogen populations on animals/crops with that in food processing environments after phage treatment may result in phage-insensitive strains.
Figure 3. (A) Phage interaction with phenotypic variants of a pathogen within food-production animal/produce; (B) phage treatment of a pathogen population in a processing environment; and (C) combining pathogen populations on animals/crops with that in food processing environments after phage treatment may result in phage-insensitive strains.
Foods 12 02734 g003
Table 3. Summarized studies on bacteriophages used to control foodborne pathogens in food products and on leafy green vegetables.
Table 3. Summarized studies on bacteriophages used to control foodborne pathogens in food products and on leafy green vegetables.
Food TypeTarget BacteriaPhage/FamilyPhage/MixturePhage DoseBacteria DoseMode of ApplicationTemperature ConditionEfficacyReference
BeefE. coli O157EP75 and EP335Cocktail 107 or 108 PFU/cm2105 CFU/cm2Spot inoculation4 °CReductions of 0.8–1.1 log10 CFU/cm2 and 0.9–1.3 log10 CFU/cm2, respectively.[154]
Raw meatballE. coli O157:H7M8AEC16Single 1010 PFU/mL102, 104 and 106 CFU/gImmersion 4 °CA reduction of 0.69–2.09 log10 CFU/g after 5 h of application.[155]
Beef and lettuceE. coli O157:H7EcoShield™Cocktail109 PFU/mL103 CFU/gSpray4 °CReduced the level of bacteria by ≥94% and 87% after 5 min contact time in meet and lettuce, respectively.[156]
Beef E. coli O157:H7PS5/MyoviridaeSingle 1010 PFU/mL107 CFU/mLSpot inoculation4 and 24 °CA 2.4 log10 CFU/piece after 24 h post application at 4 °C, whereas a 3.5 log10 CFU/piece after 6 h post application at 24 °C. [157]
Chicken S. TyphimuriumPS5/MyoviridaeSingle1010 PFU/mL107 CFU/mLSpot inoculation4 and 24 °CA 1.2 log10 CFU/piece after 24 h post application at 4 °C and a 1.6 log10 CFU/piece after 6 h post application at 24 °C.[157]
Beef (coarse and fine ground)S. enterica (ATCC 51741), S. Heidelberg (ATCC 8326), S. Newport (ATCC 27869), and S. Enteritidis C (Se 13)Salmonelex™ (S16 and the FO1a)/MyoviridaeCocktail 108 and 109104 CFU/gSpot inoculation5 °COverall, a reduction of 1.6 log10 CFU/g was observed after the application of 109 phage.[158]
Ground red meat trim and poultryS. Infantis (ATCC 51741), S. Heidelberg (ATCC 8326), S. Newport (ATCC 27869), and S. Enteritidis (SE13)Salmonelex™ (S16 and the FO1a)/MyoviridaeCocktail107 and 108107 CFU/gTumbling4 °COverall, phage application on trim reduced 0.8 and 1 log10 CFU/g of Salmonella in ground pork and beef, respectively, whereas a reduction of 0.9 and 1.1 log10 CFU/g occurred in ground turkey and chicken, respectively.[66]
Chicken skinCocktail of S. Typhimurium, S. Heidelberg, and S. EnteritidisSalmoFresh™Cocktail109 PFU/mL103 CFU/gImmersion in water followed by spot inoculation and in chlorine (30 ppm) followed by phage treatment4 °CA reduction of 0.9–1 log10 CFU/cm2 with phage only. Whereas a greater reduction of 1.6 and 1.8 log10 CFU/cm2 after 2 and 24 h. after chlorine and phage treatment.[159]
Chicken S. Typhimurium, S. Newport, S., and ThompsonSalmonelex™Cocktail107 PFU/cm2104 CFU/cm2Spread in sterile filtered water or sterile tap water4 °CA reduction of 0.39 log10 CFU/cm2 and 0.67 log10 CFU/cm2 after 30 min and 8 h post-inoculation, respectively.[104]
MeatL. monocytogenesHalal-certified List-shield 109 PFU/mLConcentration not indicatedSpot inoculation4 °CA reduction of 2.3 log10 was recorded in phage-treated beef samples during the storage period of 15 days.[160]
Fresh salmon meatL. monocytogenesSH3-3/MyoviridaeSingle 108 CFU/mL105 CFU/gSpot inoculation4 °CA reduction of 2.67, 4.14, and 4.54 log10 after 24, 48, and 72 h of phage addition, respectively. [161]
Chicken Cocktail of L. monocytogenes strains ATCC 19113, ATCC19115, and ATCC 13932ListShieldCocktail 108 log CFU/g104 CFU/gSpraying4 °CA mean reduction of 0.56, 0.84, 0.46, and 0.10 log cycles in viable counts was observed at 0, 24, 48, and 72 h after phage treatment, respectively.[162]
Cooked turkey and roast beefA cocktail of L. monocytogenes (serotypes; 1/2a, 1/2b, and 4b)LISTEX™P100Cocktail107 PFU/cm2103 CFU/cm2Smearing 4 and 10 °CAn initial reduction of 2.1 and 1.7 log10 CFU/cm2, respectively, for cooked turkey and roast beef at 4 °C, while an initial reduction of 1.5 and 1.7 log10 CFU/cm2, at 10 °C.[163]
Raw chicken and pork meatC. jejuni (NCTC 11168) and C. coli (NCTC 12668)NCTC group II phage 12684 or CP81Single MOI of 10 or 100106 CFU/mLSpot inoculation4 and 37 °CNo reduction at 4 °C after 7 days of inoculation.[164]
Raw and cooked beefC. jejuniCj6/MyoviridaeSingleMOI of 10 or 10,000Low cell density of <100/cm2 or high cell density of 104 CFU/cm2Spot inoculation5 and 24 °CNo reduction at 5 °C compared to control with low MOI. However, a 2 log10 CFU/cm2 reduction on raw and cooked meat at high host density and a high MOI of 10,000.[165]
Chicken C. jejuni (NCTC12662 or RM1221)F356 and F357Cocktail107 PFU104 CFU/cm2Spot inoculation5 °CA 0.73 log10 reduction at 5 °C after 24 h post-treatment.[67]
Chicken liverC. jejuni (HPC5 and 81–176)Phages ϕ3 or ϕ15/MyoviridaeSingle 108 PFU/g103 or 105 CFU/gPhage added to liver stomachates containing C. jejuni4 °CA 0.2 to 0.7 log10 CFU/g reduction 48 h post-treatment.[166]
LettuceSalmonella ser. Enteritidis (ATCC13076) and Salmonella ser. Typhimurium (ATCC14028)BP 1369 and BP 1370/Myoviridae and Podoviridae, respectivelySingle108 PFU/mL106 CFU/mLImmersion 10, 20, and 30 °CA reduction of >1.0 log10 CFU/cm2 after 2 h of post-treatment.[145]
Romaine lettuce Individual strains of STEC (EDL933; O157:H7, SN061; O26: H11, SN576; O111:NM and SN608; and O103:H2)VE04, VE05, and VE07Single108 PFU/mL107 CFU/mLSpot inoculation and spreading with pipet10 °CA reduction of 2.6–6 log10 CFU/cm2 after 3 days of storage at a temperature of 10 °C.[167]
Romaine lettuce, mung bean sprouts, and seedsCocktail of Salmonella strains (Newport, Braenderup, Typhimurium, Kentucky, and HeidelbergSalmoFresh™/MyoviridaeCocktail108 PFU/mL105 CFU/mLSpraying or immersion2, 10, and 25 °COverall reduction by spraying SalmoFresh™ onto lettuce and sprouts reduced Salmonella by 0.76 and 0.83 log10 CFU/g, respectively, whereas a reduction of 2.43 and 2.16 log10 CFU/g by immersion was observed on lettuce and sprouts, respectively.[168]
Romaine and iceberg lettuceE. coli O157:H7AYO26, AXO111, AXO121, AYO145A/Myoviridae, AXO103, AKFV33/Siphoviridae, and AXO45BCocktail>108 PFU/mLHigh (105 CFU/g) and low (103 CFU/g)Immersion 2 °CA reduction of 2.6–3.2 and 1.7–2.3 log10 CFU/g for low and high contamination, respectively.[149]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bumunang, E.W.; Zaheer, R.; Niu, D.; Narvaez-Bravo, C.; Alexander, T.; McAllister, T.A.; Stanford, K. Bacteriophages for the Targeted Control of Foodborne Pathogens. Foods 2023, 12, 2734. https://doi.org/10.3390/foods12142734

AMA Style

Bumunang EW, Zaheer R, Niu D, Narvaez-Bravo C, Alexander T, McAllister TA, Stanford K. Bacteriophages for the Targeted Control of Foodborne Pathogens. Foods. 2023; 12(14):2734. https://doi.org/10.3390/foods12142734

Chicago/Turabian Style

Bumunang, Emmanuel W., Rahat Zaheer, Dongyan Niu, Claudia Narvaez-Bravo, Trevor Alexander, Tim A. McAllister, and Kim Stanford. 2023. "Bacteriophages for the Targeted Control of Foodborne Pathogens" Foods 12, no. 14: 2734. https://doi.org/10.3390/foods12142734

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop