Phage therapy applications: Efficacy and impact on the ...

164
Purdue University Purdue e-Pubs Open Access Dissertations eses and Dissertations January 2015 Phage therapy applications: Efficacy and impact on the animal intestinal microbiome Yingying Hong Purdue University Follow this and additional works at: hps://docs.lib.purdue.edu/open_access_dissertations is document has been made available through Purdue e-Pubs, a service of the Purdue University Libraries. Please contact [email protected] for additional information. Recommended Citation Hong, Yingying, "Phage therapy applications: Efficacy and impact on the animal intestinal microbiome" (2015). Open Access Dissertations. 1115. hps://docs.lib.purdue.edu/open_access_dissertations/1115

Transcript of Phage therapy applications: Efficacy and impact on the ...

Page 1: Phage therapy applications: Efficacy and impact on the ...

Purdue UniversityPurdue e-Pubs

Open Access Dissertations Theses and Dissertations

January 2015

Phage therapy applications: Efficacy and impact onthe animal intestinal microbiomeYingying HongPurdue University

Follow this and additional works at: https://docs.lib.purdue.edu/open_access_dissertations

This document has been made available through Purdue e-Pubs, a service of the Purdue University Libraries. Please contact [email protected] foradditional information.

Recommended CitationHong, Yingying, "Phage therapy applications: Efficacy and impact on the animal intestinal microbiome" (2015). Open AccessDissertations. 1115.https://docs.lib.purdue.edu/open_access_dissertations/1115

Page 2: Phage therapy applications: Efficacy and impact on the ...

Graduate School Form 30 Updated 1/15/2015

PURDUE UNIVERSITY GRADUATE SCHOOL

Thesis/Dissertation Acceptance

This is to certify that the thesis/dissertation prepared

By

Entitled

For the degree of

Is approved by the final examining committee:

To the best of my knowledge and as understood by the student in the Thesis/Dissertation Agreement, Publication Delay, and Certification Disclaimer (Graduate School Form 32), this thesis/dissertation adheres to the provisions of Purdue University’s “Policy of Integrity in Research” and the use of copyright material.

Approved by Major Professor(s):

Approved by: Head of the Departmental Graduate Program Date

Yingying Hong

Phage Therapy Applications: Efficacy and Impact on the Animal Intestinal Microbiome

Doctor of Philosophy

Alan MathewCo-chair

Paul Ebner

Co-chair

Haley Oliver

Marcos Rostagno

Alan Mathew

Ryan Cabot Dec 8, 2015

Page 3: Phage therapy applications: Efficacy and impact on the ...

i

i

PHAGE THERAPY APPLICATIONS: EFFICACY AND IMPACT ON THE ANIMAL

INTESTINAL MICROBIOME

A Dissertation

Submitted to the Faculty

of

Purdue University

by

Yingying Hong

In Partial Fulfillment of the

Requirements for the Degree

of

Doctor of Philosophy

December 2015

Purdue University

West Lafayette, Indiana

Page 4: Phage therapy applications: Efficacy and impact on the ...

ii

ii

For my parents

Page 5: Phage therapy applications: Efficacy and impact on the ...

iii

iii

ACKNOWLEDGEMENTS

I would like to express my sincere appreciation to everyone who contributed to

the completion of my dissertation. Firstly, I would like to thank my PhD committee, Drs.

Alan Mathew, Paul Ebner, Haley Oliver and Marcos Rostagno, for sharing their immense

knowledge and insightful ideas, and for providing me with constructive suggestions and

continuous support. I would like to especially thank Dr. Ebner, who I have known since

2007 when I was participating the Undergraduate Research Abroad Program at Purdue. I

feel really fortunate to have the chance (thank you Dr. Mathew for offering me the

opportunity to continue my PhD) to continue learn from him for my PhD study. Thank

you very much for leading me to the interesting area of phage therapy, thank you for

teaching me not just science but many “life-saving” people skills, thank you for all your

words of encouragement, thank you for laughing at my bad jokes, and of course, thank

you for keeping faith in my ability to talk about feelings. It has been a delightful

experience working in your lab.

I am also very fortunate to be friends with many talented people at Purdue.

Special thanks go to Jiayi Zhang (Jay) and Yanying Pan (Yani), my labmates and best

friends, for your help with my experiments as well as my job searching. Jay, your

futuristic and entrepreneur mind is a constant inspiration to me; and Yani, I admire your

optimistic life style and artistic view of life. Thank you also goes to Liting Xu (Lily) and

Page 6: Phage therapy applications: Efficacy and impact on the ...

iv

iv

Hang Lu for being wonderful friends since college, and to Luiz Albino, Archana Singh,

Kyle Schmidt, Anne Marshall, Danielle Marks, Samantha Hatter, Tianhui Zhou, Junyi

Tao and many others, for helping with my projects, and to Ms. JoAnn Galyon for always

having the magic to squeeze me into Dr. Mathew’s calendar.

Last, but most importantly, my deepest gratitude and love to my parents, for their

unconditional and everlasting understanding, support, and love.

Page 7: Phage therapy applications: Efficacy and impact on the ...

v

v

TABLE OF CONTENTS

Page

ABSTRACT ....................................................................................................................... ix!CHAPTER 1.! LITERATURE REVIEW ......................................................................... 1!

1.1! Foodborne Pathogens And Foodborne Diseases ................................................... 1!1.1.1! Pathogenesis of Major Foodborne Pathogens .................................................. 2!1.1.2! The Transmission and Prevalence of Major Foodborne Pathogens ................ 4!1.1.3! Challenges of Antibiotic Resistance ................................................................ 6!1.1.4! Food Safety Interventions ................................................................................ 7!

1.2! Phage Therapy ..................................................................................................... 11!1.2.1! Pre-harvest Phage Applications ..................................................................... 12!1.2.2! Post-harvest Phage Applications ................................................................... 15!1.2.3! Phage Genome Characterization .................................................................... 17!1.2.4! Phage Resistance ........................................................................................... 18!1.2.5! Animal Gut Microbiome ................................................................................ 21!1.2.6! Phage Commercialization and Its Hurdles .................................................... 24!

1.3! Goal of The Projects ............................................................................................ 26!List of References ........................................................................................................ 27!

CHAPTER 2.! DEVELOPMENT OF BACTERIOPHAGE TREATMENTS TO

REDUCE ESCHERICHIA COLI O157:H7 CONTAMINATION OF BEEF PRODUCTS

AND PRODUCE .............................................................................................................. 40!2.1! Abstract ............................................................................................................... 40!2.2! Introduction ......................................................................................................... 42!2.3! Materials And Methods ....................................................................................... 44!

2.3.1! Bacteria and Bacteriophages .......................................................................... 44!

Page 8: Phage therapy applications: Efficacy and impact on the ...

vi

vi

Page

2.3.2! Bacteriophage Kinetics .................................................................................. 44!2.3.3! Transmission Electron Microscopy (TEM) ................................................... 45!2.3.4! Phage Treatment of E. coli O157:H7 Contaminated Food Products ............. 45!2.3.5! Isolation of Phage Resistant E. coli O157:H7 Mutants ................................. 47!2.3.6! Phage Adsorption in Phage Resistant E. coli O157:H7 Mutants ................... 47!2.3.7! In Vitro Stability of Phage Resistance ........................................................... 48!2.3.8! Fitness of Phage Resistant E. coli O157:H7 Mutants .................................... 48!2.3.9! Statistical Analysis ......................................................................................... 49!

2.4! Results ................................................................................................................. 50!2.4.1! Phage Characterization .................................................................................. 50!2.4.2! Phage Treatment of E. coli O157:H7 Contaminated Food Products ............. 50!2.4.3! Kinetics of Phage Infection of Phage Resistant Mutants ............................... 51!2.4.4! Adsorption of Phage to Phage Resistant E. coli O157:H7 Mutants .............. 52!2.4.5! In Vitro Stability of Phage Resistance ........................................................... 53!2.4.6! Fitness of Phage Resistant E. coli O157:H7 Isolates ..................................... 53!

2.5! Discussion ........................................................................................................... 55!2.6! Summary and Conclusions .................................................................................. 62!2.7! Note ..................................................................................................................... 63!List of References ........................................................................................................ 73!

CHAPTER 3.! COMPLETE GENOME SEQUENCES OF TWO ESCHERICHIA COLI

O157:H7 PHAGES EFFECTIVE IN LIMITING CONTAMINATION OF FOOD

PRODUCTS …. ................................................................................................................ 78!3.1! Abstract ............................................................................................................... 78!3.2! Genome Annoucement ........................................................................................ 78!List of References ........................................................................................................ 81!

CHAPTER 4.! TREATMENT OF SALMONELLA CONTAMINATED EGGS AND

PORK WITH A BROAD-SPECTRUM, SINGLE BACTERIOPHAGE: ASSESSMENT

OF EFFICACY AND RESISTANCE DEVELOPMENT ................................................ 83!4.1! Abstract ............................................................................................................... 83!

Page 9: Phage therapy applications: Efficacy and impact on the ...

vii

vii

Page

4.2! Introduction ......................................................................................................... 85!4.3! Materials And Methods ....................................................................................... 87!

4.3.1! Bacteria and Bacteriophage ........................................................................... 87!4.3.2! Phage Treatment of Salmonella Contaminated Food Products ..................... 87!4.3.3! Phage Susceptibility of Salmonella After Phage Treatment .......................... 89!4.3.4! Statistical Analysis ......................................................................................... 89!

4.4! Results ................................................................................................................. 90!4.4.1! Phage Treatment of Salmonella Contaminated Ground Pork ........................ 90!4.4.2! Phage Treatment of Salmonella Contaminated Liquid Egg .......................... 91!4.4.3! Development of Phage Resistant Salmonella ................................................ 91!

4.5! Discussion ........................................................................................................... 93!List of References ...................................................................................................... 104!

CHAPTER 5.! THE IMPACT OF ORAL ADMINISTERED PHAGES ON HOST:

ACUTE IMMUNE RESPONSES AND GUT MICROBIOME .................................... 108!5.1! Abstract ............................................................................................................. 108!5.2! Introduction ....................................................................................................... 110!5.3! Materials And Methods ..................................................................................... 112!

5.3.1! Bacteriophages ............................................................................................. 112!5.3.2! Mice Immune Responses to Phage Treatment ............................................. 112!5.3.3! Impact of Phage Therapy on Pig Gut Microbiome ...................................... 113!5.3.4! Statistical Analysis ....................................................................................... 116!

5.4! Results ............................................................................................................... 117!5.4.1! Immune Responses of Mice to Phage Treatment ........................................ 117!5.4.2! Pig Growth Performance ............................................................................. 118!5.4.3! Gut Microbiome Analysis and Comparison ................................................ 118!

5.5! Discussion ......................................................................................................... 120!5.6! Note ................................................................................................................... 126!List of References ...................................................................................................... 135!

Page 10: Phage therapy applications: Efficacy and impact on the ...

viii

viii

Page

CHAPTER 6.! GENERAL CONCLUSIONS ............................................................... 139!List of Reference ........................................................................................................ 144!

VITA ............................................................................................................................... 145!

Page 11: Phage therapy applications: Efficacy and impact on the ...

ix

ix

ABSTRACT

Hong, Yingying. Ph.D., Purdue University, December 2015. Phage Therapy Applications: Efficacy and Impact On the Gut Microbiome. Major Professors: Alan Mathew and Paul Ebner.

Phage therapy is a promising antibacterial intervention that has received renewed

interest in recent years. In this study, the antibacterial efficacy of phage treatments was

tested in different food matrices, under different conditions, as well as in both phage

cocktail and single phage forms. Additionally, this study also examined phage genome

sequences, bacterial phage resistance, and the impact of orally administered phages on

acute immune response and gut microbiome of the host in effort to more clearly assess

the safety and long-lasting efficacy of phage-based treatments.

The first study, presented in chapter two, evaluated the efficacy of a three-phage

cocktail in reducing concentrations of E. coli O157:H7 in contaminated ground beef,

spinach, and cheese under various conditions. Additionally, phage resistant E. coli

O157:H7 strains were characterized in terms of phage resistance mechanisms, in vitro

stability of phage resistance, and potential fitness losses. Phage treatments significantly

reduced concentrations of E. coli O157:H7 in ground beef by 1.97 log10 CFU/mL, 0.48

log10 CFU/mL, and 0.56 log10 CFU/mL when treated samples were incubated at room

temperature for 24h, under refrigeration for 24h, or undercooked, repectively. Phage

treatments also reduced concentrations of E. coli O157:H7 in spinach by 3.28 log10

Page 12: Phage therapy applications: Efficacy and impact on the ...

x

x

CFU/mL, 2.88 log10 CFU/mL, and 2.77 log10 CFU/mL when treated samples were

incubated at room temperature for 24h, 48h and 72h, respectively. Phage treatment,

however, did not reduce concentrations of E. coli O157:H7 in contaminated cheese.

Multiple resistance mechanisms were observed. For all three phage resistant E. coli

O157:H7 strains, resistance was stably maintained in vitro throughout a four-day

subculture period in the absence of phage and no significant reductions in bacterial

growth or cell adhesion were observed in resistant strains, indicating that resistance

mitigation strategies may be necessary to optimize phage therapy.

The second study, presented in chapter three, characterized the whole genome

sequences of two E. coli O157:H7 phages used in previous studies. Phages

vB_EcoM_FFH_1 and vB_EcoM_FFH_2 had genome lengths of 108,483 bp and

139,020 bp, respectively. Based on sequence homology, FFH_1 was classified as a T5-

like phage and FFH_2 was classified as an rV5-like phage. No undesired genes, including

genes coding for antibiotic resistance, toxins, virulence, or lysogeny were observed in

either E. coli phage genomes. However, 108 out of 160 predicted genes in FFH_1 and

163 out of 220 predicted genes in FFH_2 did not match to any genes with characterized

functions in NCBI database.

The third study, presented in chapter four, evaluated the efficacy of a single,

broad-spectrum phage in reducing Salmonella serovars in ground pork and liquid egg.

Phage treatment resulted in significant reduction of Salmonella Typhimurium (ranging

from 0.29 log10 CFU/mL to 1.65 log10 CFU/mL) and Salmonella Enteritidis (ranging

from 0.25 log10 CFU/mL to 1.04 log10 CFU/mL) in ground pork when treated samples

were incubated at room temperature for 24 h and 48 h and under refrigeration for 24h and

Page 13: Phage therapy applications: Efficacy and impact on the ...

xi

xi

1 wk. Phage treatments also resulted in significant reductions of Salmonella

Typhimurium by 0.49 log10 CFU/mL to 3.14 log10 CFU/mL when treated samples were

incubated at room temperature for 24 h and 48 h and under refrigeration for 24h and 1

wk; however, significant reductions of Salmonella Enteritidis (0.93 log10 CFU/mL and

0.61 log10 CFU/mL) were only observed when treated samples were incubated at room

temperature for 24 h and 48 h. High percentages of phage resistant Salmonella (92.50%

for Salmonella Typhimurium and 50.83% for Salmonella Enteritidis) were observed after

phage treatments at room temperature for 48 h. However, the emergence of phage

resistant Salmonella was only observed in liquid egg.

The last study, presented in chapter five, demonstrated the impact of orally

administrated phages on host acute immune responses and the gut microbiome. Mice

receiving a low amount of phages (105 PFU), a high amount of phages (107 PFU), or a

placebo showed no differences in the levels of intestinal IgA and IgG at either 6 h or 24 h

post treatment. Phage inoculation did elicit some cytokine responses; however, all

cytokine concentrations were within normal ranges for mice at all sampling times across

all treatments. In additional experiments, pigs were treated with basal diet, basal diet

supplemented with antibiotic premix ASP 250, and basal diet with daily inoculation of

5×1010 PFU of phages via oral gavage for two weeks. The gut microbiome of the ileum,

cecum, and feces were characterized using 16S rRNA sequencing. While antibiotic

treatment significantly altered the ileal microbiome compared to the control, no

consistent changes were observed between the phage treated microbiome and the control

microbiome in either the overall microbiome composition or the relative taxonomic

abundance. Relative abundance differences, however, were observed with certain OTUs

Page 14: Phage therapy applications: Efficacy and impact on the ...

xii

xii

between phage and control microbiomes at three intestinal compartments and further

studies are needed to determine whether these changes are deleterious to host.

Page 15: Phage therapy applications: Efficacy and impact on the ...

1

1

CHAPTER 1. LITERATURE REVIEW

1.1 Foodborne Pathogens And Foodborne Diseases

It is estimated by the US Centers for Disease Control and Prevention (CDC) that,

each year in the US, domestically acquired foodborne pathogens result in 9.4 million

illnesses, 55,961 hospitalizations, and 1,351 deaths (Scallan et al., 2011). Norovirus is the

leading cause of foodborne illnesses, accounting for 58% of the cases, followed by non-

typhoidal Salmonella (11%), Clostridium perfrigens (10%), Campylobacter (9%),

Staphylococcus aureus (2.6%), and shiga-toxin producing E. coli (STEC, 1.9%) (Scallan

et al., 2011). Based on the incidences of top 15 foodborne pathogens, which represent

more than 95% of total foodborne illnesses, the US Department of Agriculture (USDA)

estimated an approximate $15.6 million annually, associated with foodborne illness,

namely from health care expenses and wage losses. This estimate, however, does not

include the economic losses of food industry, where a recall of contaminated food can

cost the affected company a direct millions of dollars and its reputation. Therefore, it is in

the interest of both the public and assorted food industries to reduce the contamination of

foodborne pathogens in the foods.

Great effort has been taken to study various foodborne pathogens in terms of their

pathogenesis, prevalence, and transmission. Similar effort has also focused on developing

efficient, yet practical food safety interventions. These studies are the focus of the

Page 16: Phage therapy applications: Efficacy and impact on the ...

2

2

following literature review. As the goal of the accompanying research is to develop

phage therapy as an antibacterial intervention for food safety, the following review will

focus mainly on interventions that target bacterial foodborne pathogens.

1.1.1 Pathogenesis of Major Foodborne Pathogens

Depending on several factors (e.g, etiological agent, pathogenesis, immunity of

the host, etc.), foodborne diseases can be roughly divided into three types: infection,

toxicoinfection, and intoxication. Both infection and toxicoinfection diseases are caused

by ingestion of live pathogens, with the difference that pathogens causing infectious

diseases, such as Norovirus, Salmonella, Campylobacter and Listeria, are invasive to host

tissues, while pathogens causing toxicoinfectious diseases, such as STEC and

Clostridium perfringens, are generally not invasive, but are capable of producing

cytotoxins upon colonization. On the other hand, intoxication refers to diseases caused by

ingestion of foods contaminated with preformed toxins of pathogens. Clostridium

botulinum produced neurotoxin (BoNT), Bacillus cereus produced emetic toxin

(cereulide), and Staphylococcus aureus produced enterotoxin are common causes of

intoxication.

The infective doses for live pathogens in healthy adults can range from a few

hundreds cells, as in the infections of STEC (Tilden et al., 1996) and Campylobacter (M.

H. Kothary, 2001), to more than 105 cells , as in the infection of Salmonella (M. H.

Kothary, 2001). The susceptibility of bacteria to stomach acid affects the level of

infective dose greatly. In STEC O157:H7, an rpos encoded stationary phase sigma factor

mediates acid tolerance response by transforming E. coli cells to stationary phase

Page 17: Phage therapy applications: Efficacy and impact on the ...

3

3

(Gorden and Small, 1993), which explains the low infective dose required by this bacteria.

In intoxication, oral ingestion of several micrograms of toxins per kilogram of host body

weight is sufficient to cause Staphylococcus aureus- and Bacillus cereus-toxins induced

emesis (Shinagawa et al., 1995; FDA, 2012) or as few as nanograms for BoNT of

Clostridium botulinum, (FDA, 2012). Most infective doses are host dependent and

infective doses much lower than those of healthy adults can pose a threat to patients that

are very young, old, pregnant, or immunocompromised (YOPI).

In most cases, patients of foodborne diseases develop relatively mild symptoms,

such as diarrhea, abdominal cramping, and vomiting, and the symptoms are usually self-

limiting, lasting only a few days. However, in some cases, especially for the YOPI

population, foodborne diseases can be complicated by much more severe systemic

symptoms, such as hemolytic uremic syndrome (HUS) in STEC infections and meningitis

in Listeria monocytogenes infections. Such complications could lead to organ failure or

death if not treated properly and promptly. Exceptionally high mortality rates are

observed in certain pathogens or particular isolates of pathogens. For example,

foodborne diseases caused by Listeria monocytogenes, Clostridium botulinum, and Vibrio

vulnificus have the highest mortality rates of 15.9%, 17.3% and 34.8%, respectively,

among other major foodborne diseases (Scallan et al., 2011). E. coli O104:H4, the strain

responsible for the massive outbreak in Europe during 2011, harbors virulent genes of

both STEC and EAEC (enteroaggregative E. coli) origins and, therefore, is more

pathogenic than both STEC and EAEC (Muniesa et al., 2012).

Page 18: Phage therapy applications: Efficacy and impact on the ...

4

4

1.1.2 The Transmission and Prevalence of Major Foodborne Pathogens

The term ‘from farm to fork’ identifies farm as the first critical control point of

food safety intervention. Indeed, many foodborne pathogens are carried in farm animals.

Cattle are the primary reservoir of STEC. The prevalence of STEC O157:H7, the most

notable serotype, ranged from 0.3% to 27.3% in beef cattle and from 0.2% to 48.8% in

dairy cattle, and the prevalence of non-O157 STEC ranged from 4.6% to 55.9% in beef

cattle and from 0.4% to 74% in dairy cattle (Hussein and Sakuma, 2005; Hussein, 2007).

Similarly, Salmonella is a prevalent foodborne pathogen in swine and poultry. According

to a previous review, the prevalence of Salmonella ranged from 3.4% to 33% in pigs and

from 5% to 100% in chickens (Foley et al., 2008). In addition to Salmonella,

Campylobacter is also a persistent foodborne pathogen in poultry with its prevalence

ranging from 3% to 90% in broiler chicken flocks before slaughter (Guerin et al., 2010).

Large variation, however, is regularly seen when comparing prevalence studies. This can

due to factors such as farm size, husbandry method, animal age, and sampling time.

Most foodborne pathogens are carried in the gastrointestinal tract of the animals.

Under natural exposure, carriage is generally asymptomatic and it is therefore not

practical to separate infected animals from uninfected ones. Pathogens are shed via feces.

At the pre-harvest stage, the contaminated feces can lead to transmission of pathogens

from infected animals to uninfected ones, contamination of foods harvested on farm (e.g.

raw milk and eggs), contamination of water, and contamination of fresh produce irrigated

with contaminated water. At the post-harvest stage, animal carcasses can be contaminated

via contact with hides/feathers containing fecal matters or via contact with intestinal

content during evisceration.

Page 19: Phage therapy applications: Efficacy and impact on the ...

5

5

In addition to contamination from infected animals, the post-harvest environment

also poses several other high risks to food safety. Pathogens brought by raw materials

(e.g., grains, milk, etc.), insects, rodents, and workers can persist in the environment of

processing plants. A high prevalence of pathogens is frequently associated with conveyer

belts, floor junctions, drains, and sinks, which are difficult to clean and have the high

moisture that is ideal for the formation of biofilms. Listeria is one of the top foodborne

pathogens of concern at the post-harvest stage. The bacteria are widespread, capable of

forming biofilms on the surface of various materials (Chmielewski and Frank, 2003), and

can multiply even under refrigeration. Previous studies have isolated Listeria from both

the environment and equipment of dairy, meat, and vegetable processing facilities

(Chmielewski and Frank, 2003). Studies also observed a high prevalence of Listeria in

post-processing stages, such as deli shops, with a previous study showing that 6.8% of

samples (N=314) collected prior to daily operation from 15 deli shops and 9.5% of

samples (N=4503) collected during operation from 30 deli shops were positive for

Listeria Monocytogenes (Simmons et al., 2014).

The USDA and FDA maintain policies regarding allowed levels of major

pathogens in their regulated food (USDA regulates meat, poultry and processed egg

products and FDA regulates all other foods). In 1994, the USDA Food Safety and

Inspection Service (FSIS) listed STEC O157:H7 as an adulterant in foods, which

authorizes agents to recall food if it is confirmed with O157:H7 contamination. In 2011,

six additional predominant non-O157 STEC groups, including O26, O103, O111, O121,

O45, and O145, were also added to the adulterant list. FSIS has less restrictive rules to

Salmonella and Campylobacter due to their high prevalence and relatively lower

Page 20: Phage therapy applications: Efficacy and impact on the ...

6

6

pathogenicity. According to the updated standards, FSIS established the maximum

acceptable percentages of samples positive of Salmonella and Campylobacter at 9.8%

and 15.7% in broiler carcass, 25% and 1.9% in ground chicken, and 15.4% and 7.7% in

chicken parts, respectively (FSIS, 2015). Currently, no Salmonella standards are available

for pork. On the other hand, Salmonella is considered adulterant in FDA regulated foods,

such as shell eggs. Both FSIS and FDA adopted “zero-tolerance” rule to Listeria in

cooked and ready-to-eat foods.

1.1.3 Challenges of Antibiotic Resistance

Antibiotics have been widely used in livestock and poultry production since

1950’s (Castanon, 2007). They are mainly used at sub-therapeutic levels for growth

promotion and disease prevention and at therapeutic levels for disease treatment. The use

of antibiotics in food animals could potentially reduce the prevalence of foodborne

pathogens in foods (Singer et al., 2007); however, such benefit is greatly compromised by

the emergence of antibiotic resistant bacteria. A handful of studies have highlighted the

connection between the use of antibiotics and the increase of antibiotic resistant bacteria,

both pathogenic and commensal, in agriculture animals (Bager et al., 1997; Aarestrup et

al., 2000; Price et al., 2005). Those bacteria can later be transmitted to humans via food

or animal-human contact and are considered a great threat to human health. Antibiotic

resistant pathogens could lead to human infections that are less responsive to antibiotic

treatment; and they could also spread antibiotic resistance genes to antibiotic susceptible

bacteria via horizontal gene transfer. In 2013, CDC estimated that infections associated

with antibiotic resistant bacteria and fungi lead to more than 2 million illnesses and

Page 21: Phage therapy applications: Efficacy and impact on the ...

7

7

23,000 deaths annually in the US (CDC, 2013). Because agriculture use of antibiotics

contributes to the majority of antibiotic sales in US, proposals for greater restriction and

regulation are common. In 2013, the FDA implemented a Guideline for Industry to phase

out the use of certain medically important antibiotics for growth promoters while

requiring veterinary supervision for all agriculture use of antibiotics (FDA, 2013). Based

on the previous experience of antibiotic bans in Europe, several groups predict a

consequent increase of disease susceptibility may be observed in particularly young

animals, which is economically concerning and could potentially be a food safety issue

due to the fact that animals at their less optimized health status can be more susceptible to

the infection of foodborne pathogens (Singer et al., 2007). Therefore, effective

alternatives of antibiotic treatment are greatly needed.

1.1.4 Food Safety Interventions

A quantitative risk assessment predicted that a 2-log reduction of Campylobacter

in chicken carcass could lead to a 30 fold reduction of Campylobacteriosis associated

with the consumption of chicken (Rosenquist et al., 2003). This study clearly

demonstrated the relationship between successful on-farm control of foodborne

pathogens and an effective reduction of foodborne illnesses in humans. On-farm control

of foodborne pathogens could be addressed in three ways: 1) limiting environmental

exposure to insects, rodents, vehicles, and visitors that potentially carry foodborne

pathogens; 2) increasing host resistance to pathogen infections; and 3) using

antimicrobial agents to reduce the levels of pathogens in infected animals (Lin, 2009).

Based on such consideration, several common food safety interventions include

Page 22: Phage therapy applications: Efficacy and impact on the ...

8

8

vaccination, supplementation with prebiotics and probiotics, and phage therapy. The

applications of phage therapy will be discussed in detail in the second part of the

literature review.

Vaccination is an effective approach to enhance the resistance of animal hosts to

pathogen infection. Although many foodborne pathogens are commensal bacteria to the

host, the presence of these pathogens often elicits both host systemic and mucosal

immune responses, which can, in turn, reduce the levels of the pathogens in animal

intestine and, therefore, provide the fundament for vaccine development (Lin, 2009). A

successful vaccination for the use in livestock and poultry production is expected to be

not only effective, but also cross protective, easy to apply as massive immunization, and

cost-efficient. Currently, most animal vaccines are attenuated vaccines, inactivated

vaccines, or subunit vaccines. There are several commercially available Salmonella

vaccines for poultry and swine and two STEC O157:H7 vaccines for cattle. Studies have

shown the efficacy of these vaccines in preventing or reducing pathogen colonization to

various extents, with some reduction only observed in certain organs or under certain

experimental setting (Desin et al., 2011; Wileman et al., 2011). More throughout

characterization of immunogenic components of the pathogens as well as the

development of novel adjuvants and vaccination technologies, such as DNA vaccines and

vector vaccines, may lead to more effective and consistent vaccination regimes.

Additionally, for broilers particularly, vaccination can be challenging due to their short

life span of approximately 5-7 weeks and the high cost associated with individual bird

vaccination. Such challenges might be addressed using in ovo injection techniques, which

enables delivery of vaccine into eggs at a speed of 70,000 eggs/hour (Desin et al., 2013).

Page 23: Phage therapy applications: Efficacy and impact on the ...

9

9

While in ovo techniques are widely used in vaccination against viral diseases, their

application to bacterial vaccines remains to be developed.

Restriction in growth promoting antibiotics (and possibly also prophylactic

antibiotics in the near future) opens the market for probiotics and prebiotics. Probiotics

are microorganisms that can provide beneficial effects to animals and humans when

consumed in sufficient amounts. Most of the probiotic strains belong to Lactobacillus and

Bifidobacterium. While the mechanisms of probiotics in promoting animal health are not

fully understood, their ability to reduce foodborne pathogens is likely due to their

antimicrobial effect, competitive exclusion, and stimulation of host immune system

(Callaway et al., 2008). A Lactobacillus acidophilus-based probiotic was previously

reported by several independent studies to reduce the prevalence of STEC O157:H7 in

feces and hides of cattle (Brashears et al., 2003; Younts-Dahl et al., 2004; Stephens et al.,

2007). This probiotic was also observed to increase growth efficiency and is therefore

widely used in US and Canadian cattle feedlots. Similarly, various probiotics were also

tested in chicken and swine and shown to be effective in reducing Salmonella (Pascual et

al., 1999) and Campylobacter (Ghareeb et al., 2012). Prebiotics, on the other hand, are

organic compounds that are generally indigestible to animal hosts, however, they can be

digested and utilized by specific beneficial bacteria populations in hind gut, and therefore

provide competitive advantages to these bacteria. For example, previous studies (Courtin

et al., 2008; Jung et al., 2008) have shown that the inclusion of several oligosaccharides,

such as xylooligosaccharides, arabinoxylooligosachrides, and galactooligosaccharides, in

chicken feed lead to increased levels of Bifidobacterium, which is an indigenous

commensal bacteria known to act antagonistically against pathogens. Probiotics and

Page 24: Phage therapy applications: Efficacy and impact on the ...

10

10

prebiotics are sometime used together for synergistic effect (Tanner et al., 2014).

However, inconsistencies in the efficacy of probiotics and prebiotics are commonly

observed. The failed treatments could be partly due to poor adaptation or colonization of

probiotic strains or prebiotic-selected strains in the designated animal gut environments.

Coupling with advanced molecular biotechnologies in profiling the gut microbiome may

produce more consistent and effective probiotics and prebiotics.

Besides pre-harvest interventions, post-harvest food safety interventions are

equally important. It is critical for each processing plant to conduct and maintain Good

Manufacturing Practices (GMP) as well as HACCP programs. Additionally, the use of

phage therapy (Guenther et al., 2012; Hong et al., 2014), organic acids (Castillo et al.,

2001; Park et al., 2011), and irradiation (O’Bryan et al., 2008; Kudra et al., 2011) have

shown promising results in reducing the level of foodborne pathogens at post-harvest

stages. It should also be noted that no single antimicrobial intervention could perfect the

task of eliminating foodborne pathogens. Integration of multiple interventions often

results in collective or even synergistic reductions and further studies should be

conducted to optimize such effects.

Page 25: Phage therapy applications: Efficacy and impact on the ...

11

11

1.2 Phage Therapy

Bacteriophages, or phage, are natural predators of bacteria that present

ubiquitously in the nature. The discovery of phages dates to the 1910’s, where two

microbiologists, Frederick Twort and Felix d’Herelle, independently reported an anti-

bacterial phenomenon caused by virus-like organisms. These organisms were later named

‘bacteriophage’ (‘bacteria-eater’ in Greek). Shortly after their discovery, the therapeutic

potential of phages was explored worldwide. Phage therapy showed promising results in

prevention and treatment of dysentery, cholera, typhoid fever, and skin infections (Kutter

and Sulakvelidze, 2004). However, in western countries, phage therapy was soon

overshadowed and replaced by the discovery and widespread use of antibiotics, which

have better consistency and broader spectrum. In contrast, phage therapy was continued

in Eastern Europe. The Eliava Institute in Georgia (part of former Soviet Union) and

Polish Academy of Science are the two most renowned institutions in the area of clinical

phage applications.

In recent years, there has been renewed interest in phage therapy worldwide,

especially as phages are viewed as promising weapons against antibiotic resistant bacteria.

The efficacy of phage therapy has been widely evaluated for application in human

medicine, veterinary medicine, food safety, and environmental sanitation. Compared to

other antimicrobials, phages offer unique advantages of targeting bacteria with high

specificity, while being natural and self-limiting.

Based on life cycles, phages can be classified into lytic phages and temperate

phages. Lytic phages lyse bacterial cells immediately after phage proliferation, while

temperate phages integrate into the bacterial genome upon infection and replicate as a

Page 26: Phage therapy applications: Efficacy and impact on the ...

12

12

part of bacterial genome until excision is induced. It is not uncommon that a part of

bacterial genome is packed into a new phage during the excision of temperate phages. To

avoid horizontal gene transfer associated with such inaccurate excision, obligate lytic

phages are usually preferred in phage therapy.

The mechanism of phage therapy starts with the adsorption of phages to bacterial

host cells. The host ranges of phages are commonly limited to certain bacterial species or

sub-species (Loc-Carrillo and Abedon, 2011). Such specificity is facilitated by interaction

between phage tail fiber proteins and corresponding bacterial receptors. Phage nucleic

acid is then injected into bacterial cells. Early genes in the phage genome are expressed

immediately after injection to hijack host machinery for the expression of middle and late

genes, which are involved in nucleic acid replication and protein synthesis. Following the

assembly of new functional phages, phage proteins, such as endoysin and holing in gram-

bacteria and lysine in gram+ bacteria, lyse the bacterial cell and enable the release of new

phages for further infection of new hosts.

While the mechanism is straightforward, the practical application can be

complicated. Issues such as the incomplete annotation of phage genomes, development of

phage resistance, and understudied impact of phage therapy on host immune responses

and gut microbiome remain to be addressed.

1.2.1 Pre-harvest Phage Applications

Phage has been applied at both pre-harvest and post-harvest stages to prevent or

reduce various foodborne pathogens. At the pre-harvest stage, the efficacy of phage-

based applications has been extensively studied against predominant foodborne

Page 27: Phage therapy applications: Efficacy and impact on the ...

13

13

pathogens in swine, chicken, cattle, and others. The animal gastrointestinal (GI) track, as

the essential colonization site of many major foodborne pathogens, is the main target site

in aforementioned studies. However, due to the complex physiological and

microbiological environment in GI track, the in vivo efficacy of phage applications is less

predictable compared to that of in vitro studies.

The presence of stomach acid (Dabrowska et al., 2005) and certain proteolytic

enzymes (Northrop, 1964) is the primary concern when phages are administered orally.

Phage viability is greatly impaired (Koo et al., 2001; Zhang et al., 2015) under extreme

acidic condition due to denaturation of phage structure proteins that play essential roles in

the protection of phage genetic material and recognition of bacteria host. To address such

problems, protection manners, such as microencapsulation and buffering, are commonly

applied. Wall et al. (Wall et al., 2010) demonstrated the efficacy of a microencapsulated

phage cocktail to reduce experimentally infected Salmonella Typhimurium in small pigs.

A phage cocktail protected in alginate-oil microspheres was able to reduce Salmonella in

tonsils, ileum, and cecum by 2 to 3 log after phage treatment every 2 h for 6 h. In terms

of buffering, in one study, pig stomach acid was neutralized with sodium bicarbonate

prior to the treatment of E. coli phages. Compared to control pigs where no phage was

detected in feces, pigs that were pre-treated with sodium bicarbonate consistently yielded

phages at levels of 1.5 x 106 to 7 x 103 PFU per gram feces during 3 days post phage

treatment, indicating pre-neutralization of stomach acid increased numbers of viable

phages that could reach the intestine (Jamalludeen et al., 2009). For application in

chickens, due to smaller volume of chicken stomach, phage solution containing antiacid

could provide sufficient protection. As reported previously, Salmonella phages that were

Page 28: Phage therapy applications: Efficacy and impact on the ...

14

14

suspended in phosphate buffered saline containing 30% CaCO3 showed effective

reduction of both Salmonella Enteritidis and Salmonella Typhimurium in broiler chickens

(Atterbury et al., 2007).

Alternatively, to avoid the contact with stomach acid, other delivery routes have

been evaluated. Previously, the reduction of E. coli O157:H7 in feces was compared

among steers receiving phages via oral, rectal, oral + rectal administrations and control.

Rectal or oral + rectal administration did not elicit any superior bacterial reduction effect,

probably due to a short retention time of phages in the rectum (Rozema et al., 2009).

Additionally, phages have been delivered by aerosol-spray to target bacteria on livestock

hides, chicken feathers, and farming environment. A previous study demonstrated that

phage treatment via aerosol-spray in young chicks effectively prevented the incidence of

Salmonella Enteritidis infection in that population by nearly 30% (Borie et al., 2008). A

phage-based aerosol spray to reduce STEC O157:H7 in cattle hide before slaughter is

also commercially available (Goodridge and Bisha, 2011).

Besides phage delivery methods, the selection of appropriate treatment time

points is equally important to the outcome of pre-harvest phage therapy. Several studies

reported transient reduction of foodborne pathogens in response to phage treatment in

animals (Berchieri et al., 1991; Chase et al.,2005; Loc Carrillo et al., 2005). Chase et al.

(Chase et al., 2005) reported that a treatment with a 37-phage cocktail in cattle resulted in

significant decrease of STEC O157:H7 in ileum and cecum initially. However, such

reduction was compromised by the regrowth of targeted bacteria a few hours post

treatment. No phage resistant bacteria were detected throughout the experiment; instead,

authors reported an associated decrease of phage concentration, indicating that transient

Page 29: Phage therapy applications: Efficacy and impact on the ...

15

15

reduction could be a collective result of phage self-limiting and bacteria re-infection. The

use of phage in combination with other antimicrobials, such as competitive exclusion

bacteria, might be effective to prevent such transient reduction (Goodridge and Bisha,

2011). In some studies, especially those using single phage treatment, the emergence of

phage resistant bacteria was observed (Loc Carrillo et al., 2005; Atterbury et al., 2007).

The presence of phage resistant bacteria could also contribute to transient reduction of

targeted bacteria. Furthermore, phage resistant bacteria could hamper the efficacy of

additional phage application if the administration design uses multiple treatments.

Considering the occurrence of bacterial regrowth and phage resistance, it is generally

suggested that pre-harvest phage therapy is more effective when applied before animals

are slaughtered.

1.2.2 Post-harvest Phage Applications

Phage applications at post-harvest stage are generally expected to be more

effective than those in pre-harvest stage. The post-harvest environment is less dynamic or

complex than the GI track of live animals, however, the intrinsic properties of foods (e.g.

composition, pH and water content) and conditions associated with food processing (e.g.

temperature, chlorinated water and UV) could affect the efficacy of phage applications

(Zhang et al., 2015) and should be carefully considered during the development of post-

harvest phage applications.

Phage applications have been tested in various foods, including meat, fresh

produce, dairy products, and processed foods. Guenther et al. (Guenther et al., 2009)

measured the efficacy of phages to reduce experimentally contaminated Listeria

Page 30: Phage therapy applications: Efficacy and impact on the ...

16

16

monocytogenes strains in a variety of ready-to-eat foods. Phage applied at a high dosage

rapidly reduced Listeria counts to undetectable levels in liquid foods and by up to 5 log

CFU in solid foods after treatment at 6 °C for 6 days. Similarly, the use of a Salmonella

phage cocktail resulted in significant reduction of Salmonella Enteritidis by 2.5 log to 3.5

log in fresh honeydew melon slices incubated at 5 °C, 10 °C, and 20 °C. No bacterial

reduction, however, was observed when using the same phage cocktail on apple slices.

The more acidic pH of apple slices was noted as the likely main reason reduced efficacy

(Leverentz et al., 2001). On the other hand, Hong and colleagues (Hong et al., 2014)

reported a temperature effect on the efficacy of phage application in ground beef. The

three-phage cocktail used in the study reduced the levels of STEC O157:H7 by 1.97 log

CFU/mL after treatment for 24 h at room temperature (24 °C). In contrast, a reduction of

only 0.48 log CFU/mL was observed under refrigeration (4 °C) for 24 h. This was likely

due to both bacteria and phage activity being inhibited at lower than optimal growth

temperatures.

Other studies also evaluated the potential of phages to sanitize bacterial

contamination in food processing equipment and environments. Some foodborne

pathogens, such as Listeria and Campylobacter, can be persistent in food processing

plants by forming biofilms at both food-contact (e.g. slicing blades and conveyer belts)

and non-food-contact surfaces (e.g. floor drains and floor-wall conjunctions). A previous

study showed that the use of a broad-spectrum phage significantly reduced Listeria cell

counts in biofilms formed in plates and on stainless steel coupon surface (Soni and

Nannapaneni, 2010). The same study also pointed out that older biofilms with multilayer

structures were less susceptible to phage treatment. The formation of extracellular

Page 31: Phage therapy applications: Efficacy and impact on the ...

17

17

polymeric substances (EPS) matrix is a main characteristic of biofilm. Older biofilms

generally have more complex EPS matrices, which are a large barrier for efficient

delivery of phages within biofilms. While it is difficult to isolate a natural phage that is

both bacteria-specific and contains an EPS-degrading enzyme, phages can be engineered

to express EPS-degrading enzymes and such genetic modification significant improved

the efficiency of biofilm removal as described in a previous study (Lu and Collins, 2007).

1.2.3 Phage Genome Characterization

Driven by the interest in phage therapy, there is a remarkable increase in the

number of completely sequenced phage genomes. At the time of writing, there are 1536

unique phage genomes in NCBI genome database. The majority of these phage genomes

are dsDNA with a small subset of genomes being ssDNA, dsRNA, and ssRNA. In terms

of size, predominant phage genomes generally fall into one of the three ranges, which are

less than 10kbp, 30-50kbp and 100-200kbp (Hatfull, 2008).

The annotation of phage genomes is a necessary characterization of phages for

many reasons. Firstly, it facilitates more comprehensive understanding of phage infection

at the genetic level by identifying genes involved in host recognition, phage genome

replication, structural protein expression, and bacterial lysis. Secondly, with comparative

genomics, new insights into phage evolution can be provided. Lastly, the identification of

undesired genes, such as those conferring antibiotic resistance, lysogeny, toxins, and

allergens, is required as an essential safety evaluation during phage commercialization.

Despite a rapid increase in the number of available phage genomes and the recent

advance in bioinformatic analysis, information on characterization of full phage genome

Page 32: Phage therapy applications: Efficacy and impact on the ...

18

18

sequences is still lacking. As a result, most published phage genomes contain very large

sections of hypothetical genes with no predicted functions. This is true even in the T4

phage genome, one of the most well-characterized phage genomes, where nearly half of

the T4 genes are yet to be characterized (Miller et al., 2003). Further annotation of the

large unknown sections could provide more precise characterization of phages both

biologically and in terms of safety, and protect against the spread of undesired genes.

1.2.4 Phage Resistance

During the co-existence of bacteria and phages, a sub-group of bacteria often

develop phage resistance mechanisms in order to preserve bacterial lineages. This is an

unintended, however, inevitable phenomenon of phage therapy. Phage resistance is

considered one of the major hurdles that may limit phage therapy being widely applied.

The emergence of phage resistant bacteria can be observed in hours to days in phage-

bacteria co-incubations in media or in phage treatments of experimentally contaminated

foods and animals, especially when a single phage is used.

The mechanisms of phage resistance can be summarized into several categories,

including prevention of adsorption, prevention of phage nucleic acid entry, nucleic acid

degradation, and abortive infection system (Labrie et al., 2010). Adsorption is the first

step of phage infection and is facilitated by phages binding to bacterial receptors. To

escape phage adsorption, bacteria undergo conformational modification of receptors

(Nordström and Forsgren, 1974; Riede and Eschbach, 1986; Bohannan and Lenski, 2000;

Morita et al., 2002), production of extracellular matrix (Mizoguchi et al., 2003), and

competitive binding of receptors (Destoumieux-Garzón et al., 2005). Preventing the entry

Page 33: Phage therapy applications: Efficacy and impact on the ...

19

19

of phage nucleic acid is mainly reported in superinfection exclusion. In T4 phage

infection, phages that have successfully infected bacteria produce two proteins, Imm and

Sp, which provide resistance to host bacteria to further infections by the same phage. This

is facilitated by Imm-induced conformational change of phage nucleic acid injection sites

and Sp-mediated inhibition of phage lysozyme activity (Lu and Henning, 1994).

Additionally, many bacteria possess a restriction-modification (R/M) system for

degradation of foreign nucleic acid. For example, the R/M system can recognize and

degrade foreign nucleic acid that is not properly methylated. CRISPR (clustered regularly

interspaced short palindromic repeats)-Cas is another host system that targets phage

nucleic acid (Horvath and Barrangou, 2010). The cas-encoded endonuclease inactivates

phages by cleaving phage genomes at a site that shows homology to spacer sequences

contained in CRISPR region. CRISPR-Cas system can also foster resistance to new

phages by acquiring sequences of the phages as new spacers during infection. Finally,

some infected bacteria undergo cell apoptosis via abortive infection systems to interfere

with phage propagation (Dy et al., 2014).

Based on our current knowledge, it remains difficult to evaluate the impact of

phage resistance to various phage applications. Several studies demonstrated that the

development of phage resistance could lead to fitness loss in resistant mutants, rendering

them less virulent or less growth competitive. In a phage treatment to reduce

Campylobacter jejuni in broiler chickens, Scott and colleagues isolated a group of phage

resistant mutants, which, compared to the original susceptible strain, could not effectively

colonize the intestinal epithelium of broiler chickens and showed reversion to phage

susceptible phenotype when reintroduced to chicken intestine in the absence of phages

Page 34: Phage therapy applications: Efficacy and impact on the ...

20

20

(Loc Carrillo et al., 2005; Scott et al., 2007). Consistent with this study, the loss of phage

resistant phenotype was also reported when continuously culturing phage resistant

Salmonella mutants in vitro in the absence of phages (Atterbury et al., 2007). Meanwhile,

growth competitiveness was measured in phage resistant Pseudomonas fluorescens

mutants, which showed 36% less competitive than phage susceptible strains when grown

in soil (Gómez and Buckling, 2011).

One feature that differentiates phage resistance from many other anti-bacterial

resistances is the potential of phages to circumvent phage resistance. Such phenomena

can be observed in a continuous bacteria-phage coincubation (Mizoguchi et al., 2003).

While it is theoretically possible that phage mutants can always be isolated to treat

bacteria with phage resistance, practically, the cost for characterization, the requirement

of new regulatory approval make continual replacement of phages impractical.

Using a phage cocktail of multiple distinct phages is one of the most practical and

effective strategies to avoid or delay the development of phage resistance. Compared to

the use of a single phage, a bacterium is much less likely to simultaneously develop

multiple, unrelated resistance mechanisms against all phages in the cocktail. However,

the expense of characterization, regulation approval and production increases with the

increase in number of phages in cocktail. Therefore, a fine balance between the

complexity of phage cocktail and the development expense must be obtained. In addition,

previous studies also suggested rotation of several phage cocktails in order to optimize

the efficiency of a long-term phage treatment.

Page 35: Phage therapy applications: Efficacy and impact on the ...

21

21

1.2.5 Animal Gut Microbiome

The animal GI tract harbors a diverse population of microorganisms. For example,

approximately 800 microbial species were identified in swine gut (Looft et al., 2012).

Collectively, these gut microorganisms are called the gut microbiome, which has been

long known to play an important role in both the growth and health of animals.

The gut microbiome serves as a natural defense against opportunistic pathogens

by competitive exclusion and production of antimicrobial compounds. Metabolically, the

gut microbiome helps extract energy from host-indigestible feed ingredients. This is

evidenced by the fact that conventional raised mice require 30% less calorie intake than

germ-free mice to maintain the same body weight (Wostmann et al., 1983). Meanwhile,

many microbial by-products are essential nutrients to host, such as vitamin K, short-chain

fatty acids, and growth factors. Short-chain fatty acids, for example, are the preferred

nutrients of colonic epithelial cells and, therefore, are important in colonic development

and integrity (Cook and Sellin, 1998). It is commonly observed that germ-free animals

have systemic immune defects at tissue, cellular and molecular levels, with, for example,

underdeveloped gut-associated lymphoid tissues, fewer CD4+ and CD8+ T cells, and

diminished expression of secretory intestinal IgA (Round and Mazmanian, 2009). These

defects can be corrected to certain levels by colonization with commensal gut

microorganisms (Mazmanian et al., 2005; Chinen and Rudensky, 2012), indicating the

importance of gut microbiome in shaping host immune system.

Among many factors that perturb gut microbiome, the supplementation of sub-

therapeutic antibiotics has been extensively studied. Many studies have shown that sub-

therapeutic levels of antibiotics in feed influence gut microbiome of agriculture animals

Page 36: Phage therapy applications: Efficacy and impact on the ...

22

22

(Collier et al., 2003; Dumonceaux et al., 2006; Looft et al., 2012). However, the

consequences are two-sided. On one hand, it is generally considered that feeding

antibiotics could reduce gut microbial load, which, consequently, reduces the nutrients

utilized by microbial metabolism as well as the energy expended by the immune system

to maintain microbiome homeostasis, and, therefore, leads to growth promotion. With the

help of metagenomic sequencing of the gut microbiome, it is also noticed that antibiotic-

treated gut microbiome had increased number of genes associated with energy production

and conversion (Looft et al., 2012), which further illustrates how antibiotics could impact

growth. However, the benefit of antibiotics comes at a cost. As expected, feeding

animals with sub-therapeutic antibiotics significantly increases the abundance and

diversity of antibiotic resistant genes in the gut microbiome (Looft et al., 2012).

Population shifts are also observed (Kim et al., 2012; Looft et al., 2012; Lin et al., 2013).

Previously, Lin et al. reported that the ileal microbiome of chickens fed with tylosin had

significantly lower abundance of Lactobacillus spp. than ileal microbiome of antibiotic-

free animals (Lin et al., 2013). Looft et al. showed that pigs receiving a diet with

antibiotic premix ASP 250 have increased abundance of E. coli along with members of

Succinivibrio and Ruminococcus genera and decreased abundance in members belonging

to the genera of Anaerobacter, Barnesiella, Papillibacter, Sporacetigenium, and Sarcina

in fecal microbiome comparing to control pigs (Looft et al., 2012). Whether and how the

abundance decrease or increase of specific microorganism is deleterious to host is still

largely unknown. However, studies with higher dosages of antibiotics indicated that

antibiotic-induced microbiome alteration was associated with reduced expression of

antimicrobial peptides, downregulation of genes involved in antigen presentation, and

Page 37: Phage therapy applications: Efficacy and impact on the ...

23

23

deficient T cell differentiation and activation in animals (Willing et al., 2011). Such an

environment could lead to increased susceptibility to infections as increased colonization

with Clostridium difficile and vancomicin-resistant Enterococcus has been frequently

documented in antibiotic-treated patients.

Compared to antibiotics, one advantage of phages is their high host specificity. It

is common for phages to only infect strains within a bacterial species and, much less

commonly, some closely related bacterial species (Loc-Carrillo and Abedon, 2011).

Therefore, it is commonly considered that phage treatment have little collateral effect on

untargeted commensal bacteria (Loc-Carrillo and Abedon, 2011; Ghannad and

Mohammadi, 2012). However, such a statement lacks the support of experimental

evidence, especially any evidence based on culture-independent studies. There is, to the

best of author’s knowledge, one paper demonstrating the effect of a E. coli phage cocktail

on healthy human gut microbiome via 16S rRNA sequencing and the study showed no

consistent changes or differences between fecal microbiome of individuals that received

oral-administered phages and a placebo (Sarker et al., 2012). No studies have

demonstrated the lack of effect on untargeted gut microbiome when phages are used in

agriculture animals. There are several factors that may differentiate the effect of phage

treatments on gut microbiome of agriculture animals from that of human, such as phage

products designed for agriculture usage may have broader target spectrum for the ease of

uses as presumptive treatments and many phage-targeted bacteria in agriculture animals

are commensal bacteria to the host. Therefore, it is also meaningful to evaluate the effect

or lack of effect of phage treatments on gut microbiome of various agriculture animals.

Page 38: Phage therapy applications: Efficacy and impact on the ...

24

24

1.2.6 Phage Commercialization and Its Hurdles

Currently in US, the availability of commercialized phage products is limited for

food safety. In pre-harvest stage, phage products are available as hide sprays to reduce

STEC O157:H7 in cattle or feather washes to reduce Salmonella in chickens prior to

slaughter (Goodridge and Bisha, 2011). More phage products are available at the post-

harvest stage for the decontamination of foods and food processing facilities that have

high risk of contamination with STEC O157:H7, Salmonella, and Listeria monocytogenes.

Several of the post-harvest phage products have GRAS (Generally Recognized As Safe)-

status and can be used in ready-to-eat foods; and several are OMRI (Organic Materials

Review Institute)-listed and can be used in organic products.

Despite the value of phage therapy to combat antibiotic resistant bacteria and the

successful examples of using phage to treat bacterial infections in human and animals in

Eastern Europe, the therapeutic phage applications remain limited in western countries.

Several research hurdles for therapeutic phage applications include, but are not limited to:

1) a lack of understanding of in vivo phage distribution and phage receptor expression; 2)

the difficulty in measuring phage pharmacokinetics; and 3) insufficient understanding of

the impact of phage resistance and the safety of in vivo phage treatments (Verheust et al.,

2010; Tsonos et al., 2014). Additionally, the regulatory framework of western countries

poses a significant hurdle for therapeutic phage applications. Phage therapy is currently

under the regulation of FDA using a draft protocol adapted from the traditional antibiotic

regulatory protocols. The current protocol requires each phage of the phage cocktail to

undergo individual clinical trials and requires re-approval of products if the composition

of phage cocktail is altered (Thiel, 2004). However, this protocol fails to address the

Page 39: Phage therapy applications: Efficacy and impact on the ...

25

25

unique nature of phage therapy and more appropriate approaches for phage therapy

regulation should be identified.

Page 40: Phage therapy applications: Efficacy and impact on the ...

26

26

1.3 Goal of The Projects

The first goal of this study was to determine whether phage-based approaches

could effectively reduce STEC O157:H7 and Salmonella in various experimentally

contaminated food matrices. In addition, the second goal was to characterize the factors

that could affect the efficacy and safety of phage-based methods, namely the

development of phage resistance and the potential impact of phage application on host

acute immune responses and gut microbiome. By achieving those goals, we expected to

comprehensively evaluate the potential of phage therapy as an antibacterial approach for

applications in animals and foods.

Page 41: Phage therapy applications: Efficacy and impact on the ...

27

27

List of References

Aarestrup, F. M., H. Kruse, E. Tast, A. M. Hammerum, and L. B. Jensen. 2000.

Associations between the use of antimicrobial agents for growth promotion and

the occurrence of resistance among Enterococcus faecium from broilers and pigs

in Denmark, Finland, and Norway. Microb. Drug Resist. 6:63–70.

Atterbury, R. J., M. A. P. Van Bergen, F. Ortiz, M. A. Lovell, J. A. Harris, A. De Boer, J.

A. Wagenaar, V. M. Allen, and P. A. Barrow. 2007. Bacteriophage therapy to

reduce Salmonella colonization of broiler chickens. Appl. Environ. Microbiol.

73:4543–4549.

Bager, F., M. Madsen, J. Christensen, and F. M. Aarestrup. 1997. Avoparcin used as a

growth promoter is associated with the occurrence of vancomycin-resistant

Enterococcus faecium on Danish poultry and pig farms. Prev. Vet. Med. 31:95–

112.

Berchieri, A., M. A. Lovell, and P. A. Barrow. 1991. The activity in the chicken

alimentary tract of bacteriophages lytic for Salmonella typhimurium. Res.

Microbiol. 142:541–549.

Bohannan, B. J. M., and R. E. Lenski. 2000. Linking genetic change to community

evolution: Insights from studies of bacteria and bacteriophage. Ecol. Lett. 3:362–

377.

Borie, C., I. Albala, P. Sánchez, M. L. Sánchez, S. Ramírez, C. Navarro, M. A. Morales,

A. J. Retamales, and J. Robeson. 2008. Bacteriophage treatment reduces

Salmonella colonization of infected chickens. Avian Dis. 52:64–67.

Page 42: Phage therapy applications: Efficacy and impact on the ...

28

28

Brashears, M. M., M. L. Galyean, G. H. Loneragan, J. E. Mann, and K. Killinger-Mann.

2003. Prevalence of Escherichia coli O157:H7 and performance by beef feedlot

cattle given Lactobacillus direct-fed microbials. J. Food Prot. 66:748–754.

Callaway, T. R., T. S. Edrington, R. C. Anderson, R. B. Harvey, K. J. Genovese, C. N.

Kennedy, D. W. Venn, and D. J. Nisbet. 2008. Probiotics, prebiotics and

competitive exclusion for prophylaxis against bacterial disease. Anim. Health Res.

Rev. 9:217–225.

Castanon, J. I. R. 2007. History of the Use of Antibiotic as Growth Promoters in

European Poultry Feeds. Poult Sci 86:2466–2471.

Castillo, A., L. M. Lucia, D. B. Roberson, T. H. Stevenson, I. Mercado, and G. R. Acuff.

2001. Lactic acid sprays reduce bacterial pathogens on cold beef carcass surfaces

and in subsequently produced ground beef. J. Food Prot. 64:58–62.

Cawthraw S., Ayling R., Nuijten P., Wassenaar T., and Newell DG. 1994. Isotype,

specificity, and kinetics of systemic and mucosal antibodies to Campylobacter

jejuni antigens, including flagellin, during experimental oral infections of

chickens. Avian Dis.38:341–349.

CDC. 2013. Antibiotic resistance threats in the United States, 2013.

Chase J, Kalchayanand N, and Goodridge LD. Use of bacteriophage therapy to reduce

Escherichia coli O157:H7 concentrations in an anaerobic digestor that simulates

the bovine gastrointestinal tract; IFTAnnual Meeting and Food Expo; 2005; New

Orleans, Louisiana. Abstract 108-6

Chinen, T., and A. Y. Rudensky. 2012. The effects of commensal microbiota on immune

cell subsets and inflammatory responses. Immunol. Rev. 245:45–55.

Page 43: Phage therapy applications: Efficacy and impact on the ...

29

29

Chmielewski, R. A. N., and J. F. Frank. 2003. Biofilm Formation and Control in Food

Processing Facilities. Compr. Rev. Food Sci. Food Saf. 2:22–32.

Collier, C. T., D. M. Albin, J. E. Wubben, V. M. Gabert, D. Bane, D. B. Anderson, H. R.

Gaskins, and B. Deplancke. 2003. Molecular ecological analysis of porcine ileal

microbiota responses to antimicrobial growth promoters. J Anim Sci.

81(12):3035–3045.

Cook, S. I., and J. H. Sellin. 1998. Review article: short chain fatty acids in health and

disease. Aliment. Pharmacol. Ther. 12:499–507.

Courtin, C. M., K. Swennen, W. F. Broekaert, Q. Swennen, J. Buyse, E. Decuypere, C.

W. Michiels, B. De Ketelaere, and J. A. Delcour. 2008. Effects of dietary

inclusion of xylooligosaccharides, arabinoxylooligosaccharides and soluble

arabinoxylan on the microbial composition of caecal contents of chickens. J. Sci.

Food Agric. 88:2517–2522.

Dabrowska, K., K. Switała-Jelen, A. Opolski, B. Weber-Dabrowska, and A. Gorski. 2005.

A review: Bacteriophage penetration in vertebrates. J. Appl. Microbiol. 98:7–13.

Desin, T. S., W. Köster, and A. A. Potter. 2013. Salmonella vaccines in poultry: past,

present and future. Expert Rev. Vaccines 12:87–96.

Desin, T. S., A. L. S. Wisner, P. K. S. Lam, E. Berberov, C. S. Mickael, A. A. Potter, and

W. Köster. 2011. Evaluation of Salmonella enterica serovar Enteritidis

pathogenicity island-1 proteins as vaccine candidates against S. Enteritidis

challenge in chickens. Vet. Microbiol. 148:298–307.

Page 44: Phage therapy applications: Efficacy and impact on the ...

30

30

Destoumieux-Garzón, D., S. Duquesne, J. Peduzzi, C. Goulard, M. Desmadril, L.

Letellier, S. Rebuffat, and P. Boulanger. 2005. The iron-siderophore transporter

FhuA is the receptor for the antimicrobial peptide microcin J25: role of the

microcin Val11-Pro16 beta-hairpin region in the recognition mechanism.

Biochem. J. 389:869–876.

Dumonceaux, T. J., J. E. Hill, S. M. Hemmingsen, and A. G. Van Kessel. 2006.

Characterization of intestinal microbiota and response to dietary virginiamycin

supplementation in the broiler chicken. Appl. Environ. Microbiol. 72:2815–2823.

Dy, R. L., R. Przybilski, K. Semeijn, G. P. C. Salmond, and P. C. Fineran. 2014. A

widespread bacteriophage abortive infection system functions through a Type IV

toxin-antitoxin mechanism. Nucleic Acids Res. 42:4590–4605.

[FDA] Food and Drug Administration. 2012. Bad bug book, Foodborne Pathogenic

Microorganisms and Natural Toxins Handbook. Available at:

www.fda.gov/downloads/Food/FoodborneIllnessContaminants/UCM297627.pdf.

[FDA] Food and Drug Administration. 2013. Guidance for industry #213. Available at:

www.fda.gov/downloads/AnimalVeterinary/GuidanceComplianceEnforcement/G

uidanceforIndustry/UCM299624.pdf.

Foley, S. L., A. M. Lynne, and R. Nayak. 2008. Salmonella challenges: prevalence in

swine and poultry and potential pathogenicity of such isolates. J. Anim. Sci. 86.

Page 45: Phage therapy applications: Efficacy and impact on the ...

31

31

[FSIS] USDA Food Safety and Inspection Service. 2015. Changes to the Salmonella and

Campylobacter varification testing program: proposed performace standards for

Salmonella and Campylobacter in Not-Ready-to-Eat comminuted chicken and

turkey products and raw chickenparts and related agency verification procedures

and other changes to agency sampling. Avalable at:

http://www.fsis.usda.gov/wps/wcm/connect/55a6586e-d2d6-406a-b2b9-

e5d83c110511/2014-0023.pdf?MOD=AJPERES.

Ghannad, M. S., and A. Mohammadi. 2012. Bacteriophage: Time to re-evaluate the

potential of phage therapy as a promising agent to control multidrug-resistant

bacteria. Iran. J. Basic Med. Sci. 15:693–701.

Ghareeb, K., W. A. Awad, M. Mohnl, R. Porta, M. Biarnes, J. Bohm, and G. Schatzmayr.

2012. Evaluating the efficacy of an avian-specific probiotic to reduce the

colonization of Campylobacter jejuni in broiler chickens. Poult. Sci. 91:1825–

1832.

Gómez, P., and A. Buckling. 2011. Bacteria-phage antagonistic coevolution in soil.

Science. 332:106–109.

Goodridge, L. D., and B. Bisha. 2011. Phage-based biocontrol strategies to reduce

foodborne pathogens in foods. Bacteriophage 1:130–137.

Gorden, J., and P. L. Small. 1993. Acid resistance in enteric bacteria. Infect. Immun.

61:364–367.

Guenther, S., O. Herzig, L. Fieseler, J. Klumpp, and M. J. Loessner. 2012. Biocontrol of

Salmonella Typhimurium in RTE foods with the virulent bacteriophage FO1-E2.

Int. J. Food Microbiol. 154:66–72.

Page 46: Phage therapy applications: Efficacy and impact on the ...

32

32

Guenther, S., D. Huwyler, S. Richard, and M. J. Loessner. 2009. Virulent bacteriophage

for efficient biocontrol of Listeria monocytogenes in ready-to-eat foods. Appl.

Environ. Microbiol. 75:93–100.

Guerin, M. T., C. Sir, J. M. Sargeant, L. Waddell, A. M. O’Connor, R. W. Wills, R. H.

Bailey, and J. A. Byrd. 2010. The change in prevalence of Campylobacter on

chicken carcasses during processing: a systematic review. Poult. Sci. 89:1070–

1084.

Hatfull, G. F. 2008. Bacteriophage genomics. Curr. Opin. Microbiol. 11:447–453.

Hong, Y., Y. Pan, and P. D. Ebner. 2014. Meat science and muscle biology symposium:

Development of Bacteriophage treatments to reduce Escherichia coli O157:H7

contamination of beef products and produce. J. Anim. Sci. 92:1366–1377.

Horvath, P., and R. Barrangou. 2010. CRISPR/Cas, the immune system of bacteria and

archaea. Science 327:167–170.

Hussein, H. S., and T. Sakuma. 2005. Prevalence of shiga toxin-producing Escherichia

coli in dairy cattle and their products. J. Dairy Sci. 88:450–465.

Hussein, H. S. 2007. Prevalence and pathogenicity of Shiga toxin-producing Escherichia

coli in beef cattle and their products. J. Anim. Sci. 85.

Jamalludeen, N., R. P. Johnson, P. E. Shewen, and C. L. Gyles. 2009. Evaluation of

bacteriophages for prevention and treatment of diarrhea due to experimental

enterotoxigenic Escherichia coli O149 infection of pigs. Vet. Microbiol. 136:135–

141.

Page 47: Phage therapy applications: Efficacy and impact on the ...

33

33

Jung, S. J., R. Houde, B. Baurhoo, X. Zhao, and B. H. Lee. 2008. Effects of galacto-

oligosaccharides and a Bifidobacteria lactis-based probiotic strain on the growth

performance and fecal microflora of broiler chickens. Poult. Sci. 87:1694–1699.

Kim, H. B., K. Borewicz, B. A. White, R. S. Singer, S. Sreevatsan, Z. J. Tu, and R. E.

Isaacson. 2012. Microbial shifts in the swine distal gut in response to the

treatment with antimicrobial growth promoter, tylosin. Proc. Natl. Acad. Sci.

109:15485–15490.

Koo, J., D. L. Marshall, and A. DePaola. 2001. Antacid Increases Survival of Vibrio

vulnificus and Vibrio vulnificus Phage in a Gastrointestinal Model. Appl. Environ.

Microbiol. 67:2895–2902.

Kudra, L. L., J. G. Sebranek, J. S. Dickson, a F. Mendonca, Q. Zhang, a Jackson-Davis,

and K. J. Prusa. 2011. Control of Salmonella enterica Typhimurium in chicken

breast meat by irradiation combined with modified atmosphere packaging. J. Food

Prot. 74:1833–9.

Kutter, E., and A. Sulakvelidze. 2004. Bacteriophages: biology and applications.

Labrie, S. J., J. E. Samson, and S. Moineau. 2010. Bacteriophage resistance mechanisms.

Nat. Rev. Microbiol. 8:317–327.

Leverentz, B., W. S. Conway, Z. Alavidze, W. J. Janisiewicz, Y. Fuchs, M. J. Camp, E.

Chighladze, and A. Sulakvelidze. 2001. Examination of bacteriophage as a

biocontrol method for Salmonella on fresh-cut fruit: a model study. J. Food Prot.

64:1116–1121.

Page 48: Phage therapy applications: Efficacy and impact on the ...

34

34

Lin, J., A. a Hunkapiller, A. C. Layton, Y.-J. Chang, and K. R. Robbins. 2013. Response

of intestinal microbiota to antibiotic growth promoters in chickens. Foodborne

Pathog. Dis. 10:331–7.

Lin, J. 2009. Novel approaches for Campylobacter control in poultry. Foodborne Pathog.

Dis. 6:755–765.

Loc Carrillo, C., R. J. Atterbury, A. El-Shibiny, P. L. Connerton, E. Dillon, A. Scott, and

I. F. Connerton. 2005. Bacteriophage therapy to reduce Campylobacter jejuni

colonization of broiler chickens. Appl. Environ. Microbiol. 71:6554–6563.

Loc-Carrillo, C., and S. T. Abedon. 2011. Pros and cons of phage therapy. Bacteriophage

1:111–114.

Looft, T., T. A. Johnson, H. K. Allen, D. O. Bayles, D. P. Alt, R. D. Stedtfeld, W. J. Sul,

T. M. Stedtfeld, B. Chai, J. R. Cole, S. A. Hashsham, J. M. Tiedje, and T. B.

Stanton. 2012. In-feed antibiotic effects on the swine intestinal microbiome. Proc.

Natl. Acad. Sci. 109:1691–6.

Lu, M. J., and U. Henning. 1994. Superinfection exclusion by T-even-type coliphages.

Trends Microbiol. 2:137–139.

Lu, T. K., and J. J. Collins. 2007. Dispersing biofilms with engineered enzymatic

bacteriophage. Proc. Natl. Acad. Sci. U. S. A. 104:11197–11202.

M. H. Kothary, U. S. B. 2001. Infective dose of foodborne pathogens in volenteer: a

review. J. Food Saf. 21:49–68.

Mazmanian, S. K., H. L. Cui, A. O. Tzianabos, and D. L. Kasper. 2005. An

immunomodulatory molecule of symbiotic bacteria directs maturation of the host

immune system. Cell 122:107–118.

Page 49: Phage therapy applications: Efficacy and impact on the ...

35

35

Miller, E. S., E. Kutter, G. Mosig, F. Arisaka, T. Kunisawa, and W. Rüger. 2003.

Bacteriophage T4 genome. Microbiol. Mol. Biol. Rev. 67:86–156.

Mizoguchi, K., M. Morita, C. R. Fischer, M. Yoichi, Y. Tanji, and H. Unno. 2003.

Coevolution of bacteriophage PP01 and Escherichia coli O157#: H7 in continuous

culture. Appl. Environ. Microbiol. 69:170–176.

Morita, M., Y. Tanji, K. Mizoguchi, T. Akitsu, N. Kijima, and H. Unno. 2002.

Characterization of a virulent bacteriophage specific for Escherichia coli O157:H7

and analysis of its cellular receptor and two tail fiber genes. FEMS Microbiol. Lett.

211:77–83.

Muniesa, M., J. A. Hammerl, S. Hertwig, B. Appel, and H. Brüssow. 2012. Shiga toxin-

producing Escherichia coli O104:H4: A new challenge for microbiology. Appl.

Environ. Microbiol. 78:4065–4073.

Myszewski M. A. and N. J. Stern. 1990. Influence of Campylobacter jejuni cecal

colonization on immunoglobulin response in chickens. Avian Dis. 34:588–594.

Nordström, K., and A. Forsgren. 1974. Effect of protein A on adsorption of

bacteriophages to Staphylococcus aureus. J. Virol. 14:198–202.

Northrop, J. H. 1964. The effect of proteolytic enzymes on E. coli phages and on native

proteins. J. Gen. Physiol. 48:73–78.

O’Bryan, C. a, P. G. Crandall, S. C. Ricke, and D. G. Olson. 2008. Impact of irradiation

on the safety and quality of poultry and meat products: a review. Crit. Rev. Food

Sci. Nutr. 48:442–457.

Page 50: Phage therapy applications: Efficacy and impact on the ...

36

36

Park, S.-H., M.-R. Choi, J.-W. Park, K.-H. Park, M.-S. Chung, S. Ryu, and D.-H. Kang.

2011. Use of organic acids to inactivate Escherichia coli O157:H7, Salmonella

Typhimurium, and Listeria monocytogenes on organic fresh apples and lettuce. J.

Food Sci. 76:M293–M298.

Pascual, M., M. Hugas, J. I. Badiola, J. M. Monfort, and M. Garriga. 1999. Lactobacillus

salivarius CTC2197 prevents Salmonella enteritidis colonization in chickens. Appl.

Environ. Microbiol. 65:4981–4986.

Price, L. B., E. Johnson, R. Vailes, and E. Silbergeld. 2005. Fluoroquinolone-resistant

Campylobacter isolates from conventional and antibiotic-free chicken products.

Environ. Health Perspect. 113:557–560.

Riede, I., and M. L. Eschbach. 1986. Evidence that TraT interacts with OmpA of

Escherichia coli. FEBS Lett. 205:241–245.

Rosenquist, H., N. L. Nielsen, H. M. Sommer, B. Nørrung, and B. B. Christensen. 2003.

Quantitative risk assessment of human campylobacteriosis associated with

thermophilic Campylobacter species in chickens. Int. J. Food Microbiol. 83:87–

103.

Round, J. L., and S. K. Mazmanian. 2009. The gut microbiota shapes intestinal immune

responses during health and disease. Nat Rev Immunol 9:313–323.

Rozema, E. A., T. P. Stephens, S. J. Bach, E. K. Okine, R. P. Johnson, K. Stanford, and T.

A. McAllister. 2009. Oral and rectal administration of bacteriophages for control

of Escherichia coli O157:H7 in feedlot cattle. J. Food Prot. 72:241–250.

Page 51: Phage therapy applications: Efficacy and impact on the ...

37

37

Sarker, S. A., S. McCallin, C. Barretto, B. Berger, A. C. Pittet, S. Sultana, L. Krause, S.

Huq, R. Bibiloni, A. Bruttin, G. Reuteler, and H. Brüssow. 2012. Oral T4-like

phage cocktail application to healthy adult volunteers from Bangladesh. Virology

434:222–232.

Sauders, B. D., M. D. Sanchez, D. H. Rice, J. Corby, S. Stich, E. D. Fortes, S. E. Roof,

and M. Wiedmann. 2009. Prevalence and molecular diversity of Listeria

monocytogenes in retail establishments. J. Food Prot. 72:2337–2349.

Scallan, E., R. M. Hoekstra, F. J. Angulo, R. V Tauxe, M.-A. Widdowson, S. L. Roy, J. L.

Jones, and P. M. Griffin. 2011. Foodborne illness acquired in the United States--

major pathogens. Emerg. Infect. Dis. 17:7–15.

Scott, A. E., A. R. Timms, P. L. Connerton, C. L. Carrillo, K. A. Radzum, and I. F.

Connerton. 2007. Genome dynamics of Campylobacter jejuni in response to

bacteriophage predation. PLoS Pathog. 3:1142–1151.

Shinagawa, K., H. Konuma, H. Sekita, and S. Sugii. 1995. Emesis of rhesus monkeys

induced by intragastric administration with the HEp-2 vacuolation factor

(cereulide) produced by Bacillus cereus. FEMS Microbiol. Lett. 130:87–90.

Simmons, C., M. J. Stasiewicz, E. Wright, S. Warchocki, S. Roof, J. R. Kause, N. Bauer,

S. Ibrahim, M. Wiedmann, and H. F. Oliver. 2014. Listeria monocytogenes and

Listeria spp. Contamination Patterns in Retail Delicatessen Establishments in

Three U.S. States. J. Food Prot. 77:1929–39.

Singer, R. S., L. A. Cox, J. S. Dickson, H. S. Hurd, I. Phillips, and G. Y. Miller. 2007.

Modeling the relationship between food animal health and human foodborne

illness. Prev. Vet. Med. 79:186–203.

Page 52: Phage therapy applications: Efficacy and impact on the ...

38

38

Soni, K. A., and R. Nannapaneni. 2010. Removal of Listeria monocytogenes biofilms

with bacteriophage P100. J. Food Prot. 73:1519–1524.

Stephens, T. P., G. H. Loneragan, E. Karunasena, and M. M. Brashears. 2007. Reduction

of Escherichia coli O157 and Salmonella in feces and on hides of feedlot cattle

using various doses of a direct-fed microbial. J. Food Prot. 70:2386–2391.

Tanner, S. A., C. Chassard, A. Zihler Berner, and C. Lacroix. 2014. Synergistic effects of

Bifidobacterium thermophilum RBL67 and selected prebiotics on inhibition of

Salmonella colonization in the swine proximal colon PolyFermS model. Gut

Pathog. 6:44.

Thiel, K. 2004. Old dogma, new tricks--21st Century phage therapy. Nat. Biotechnol.

22:31–36.

Tilden, J., W. Young, A. M. McNamara, C. Custer, B. Boesel, M. A. Lambert-Fair, J.

Majkowski, D. Vugia, S. B. Werner, J. Hollingsworth, and J. G. Morris. 1996. A

new route of transmission for Escherichia coli: Infection from dry fermented

salami. Am. J. Public Health 86:1142–1145.

Tsonos, J., D. Vandenheuvel, Y. Briers, H. De Greve, J. P. Hernalsteens, and R. Lavigne.

2014. Hurdles in bacteriophage therapy: Deconstructing the parameters. Vet.

Microbiol. 171:460–469.

Verheust, C., K. Pauwels, J. Mahillon, D. R. Helinski, and P. Herman. 2010. Contained

Use of Bacteriophages#: Risk Assessment and Biosafety Recommendations. Appl.

Biosaf. 15:32–44.

Page 53: Phage therapy applications: Efficacy and impact on the ...

39

39

Wall, S. K., J. Zhang, M. H. Rostagno, and P. D. Ebner. 2010. Phage therapy to reduce

preprocessing Salmonella infections in market-weight swine. Appl. Environ.

Microbiol. 76:48–53.

Wileman, B. W., D. U. Thomson, K. C. Olson, J. R. Jaeger, L. A. Pacheco, J. Bolte, D. T.

Burkhardt, D. A. Emery, and D. Straub. 2011. Escherichia coli O157:H7

shedding in vaccinated beef calves born to cows vaccinated prepartum with

Escherichia coli O157:H7 SRP vaccine. J. Food Prot. 74:1599–1604.

Willing, B. P., S. L. Russell, and B. B. Finlay. 2011. Shifting the balance: antibiotic

effects on host-microbiota mutualism. Nat. Rev. Microbiol. 9:233–243.

Wostmann, B. S., C. Larkin, A. Moriarty, and E. Bruckner-Kardoss. 1983. Dietary intake,

energy metabolism, and excretory losses of adult male germfree Wistar rats. Lab.

Anim. Sci. 33:46–50.

Younts-Dahl, S. M., M. L. Galyean, G. H. Loneragan, N. A. Elam, and M. M. Brashears.

2004. Dietary supplementation with Lactobacillus- and Propionibacterium-based

direct-fed microbials and prevalence of Escherichia coli O157 in beef feedlot

cattle and on hides at harvest. J. Food Prot. 67:889–893.

Zhang J., Y. Hong, M. Fealey, A. Singh, K. Walton, C. Martin, N. J . Harman, J. Mahlie,

and P. D. Ebner. 2015. Physiological and molecular characterization of

Salmonella bacteriophages previously used in phage therapy. J. Food Prot. (in

press).

Page 54: Phage therapy applications: Efficacy and impact on the ...

40

40

CHAPTER 2. DEVELOPMENT OF BACTERIOPHAGE TREATMENTS TO REDUCE ESCHERICHIA COLI O157:H7 CONTAMINATION OF BEEF

PRODUCTS AND PRODUCE

2.1 Abstract

E. coli O157:H7 remains a foodborne pathogen of concern with infections

associated with products ranging from ground beef to produce to processed foods. We

previously demonstrated that phage-based technologies could reduce foodborne pathogen

colonization in live animals. Here, we examined if a three-phage cocktail could reduce E.

coli O157:H7 in experimentally contaminated ground beef, spinach, and cheese. The

three phages were chosen from our E. coli O157:H7 phage library based on their distinct

origins of isolation, lytic ranges and rapid growth (40- to 50-min life cycle). Two phages

belonged to the Myoviridae family and the other phage belonged to the Siphoviridae

family. The phage cocktail was added to ground beef, spinach leaves and cheese slices

contaminated with E. coli O157:H7 (107 CFU) at a multiplicity of infection of 1. Phage

treatment reduced (P<0.05) the concentrations of E. coli O157:H7 by 1.97 log10 CFU/mL

in ground beef when stored at room temperature (24 °C) for 24 h, 0.48 log10 CFU/mL at

refrigeration (4 °C), and 0.56 log10 CFU/mL in undercooked condition (internal

temperature of 46 °C). Likewise, phage treatment reduced (P<0.05) E. coli O157:H7 by

3.28 log10 CFU/mL, 2.88 log10 CFU/mL, and 2.77 log10 CFU/mL in spinach when stored

at room temperature for 24 h, 48 h and 72 h, respectively. Phage treatment, however, did

Page 55: Phage therapy applications: Efficacy and impact on the ...

41

41

not reduce E. coli O157:H7 concentrations in contaminated cheese. Additionally, three

phage resistant E. coli O157:H7 strains [309-PR (phage resistant) 1, 309-PR4, and 502-

PR5] were isolated and characterized to test if phage resistance could limit long-term use

of phages as biocontrol agents. Growth kinetics and adsorption assays indicated that

phage resistance in strains 309-PR4 and 502-PR5 was mediated, at least in part, by

prevention of phage adsorption. Phage resistance in strain 309-PR1 was the result of

limited phage proliferation. Phage resistance was stably maintained in vitro throughout a

four-day subculture period in the absence of phage. No significant reductions in bacterial

growth or cell adhesion were observed in resistant strains. Taken together, our results

provide additional support for the use of phage to control E. coli O157:H7 in food

products; however, the emergence of phage resistant bacteria could limit the efficacy of

phage products. Therefore, further studies are needed to develop resistance mitigation

strategies to optimize phage-based technologies.

Page 56: Phage therapy applications: Efficacy and impact on the ...

42

42

2.2 Introduction

The foodborne pathogen, E. coli O157:H7, is a well-known Shiga-toxin

producing bacterium that caused more than 63,000 illnesses, 2,138 hospitalizations, and

20 deaths in the U.S. in 2011 (Scallan et al., 2011). Ruminants are the main reservoirs.

The bacterium predominantly colonizes the recto-anal junction of cattle and is passed to

the environment through feces (Grauke et al., 2002), which often contaminate hides,

drinking water, and pen barriers leading to rapid transmission among housed cattle

(McGee et al., 2004). The pathogen can then be transferred to carcasses during

processing resulting in contaminated meat products. Traditionally, many human

infections have been associated with undercooked contaminated meat products,

particularly ground beef; however, contaminated fresh produce and other ready- to-eat

and processed foods are increasingly associated with E. coli O157:H7 infections (Rangel

et al., 2005).

The use of bacteriophage-based technologies to control foodborne pathogen

transmission has received more attention in recent years. Phages have the advantages of

being easy to isolate and very specific for targeted bacteria (Loc-Carrillo and Abedon,

2011). Currently, only a limited number of studies have examined the efficacy of phages

to limit E. coli O157:H7 contamination of food products (Sharma et al., 2009; Viazis et

al., 2011; Hudson et al., 2013). In this study, we aimed to determine whether phage-based

treatments could effectively limit pathogen contamination in food products. We isolated

wild-type E. coli O157:H7 phages and characterized them to identify phages with

optimum host-ranges and in vitro kinetics. Three potential phages were further tested for

their ability to limit E. coli O157:H7 contamination in ground beef, spinach, and cheese.

Page 57: Phage therapy applications: Efficacy and impact on the ...

43

43

Finally, we examined the development of phage resistance to determine whether this

phenomenon could limit the long-term use of phages as biocontrol agents.

Page 58: Phage therapy applications: Efficacy and impact on the ...

44

44

2.3 Materials And Methods

2.3.1 Bacteria and Bacteriophages

Sixteen E. coli O157:H7 phages were isolated from different wastewater

treatment plants located throughout Indiana using a method described previously (Wall et

al., 2010). Each phage was tested for its lytic ability against a panel of 48 distinct (i.e.,

individually typed and collected from various laboratories, outbreaks, animals, food

sources) E. coli O157:H7 isolates by standard plaque assay, as described previously

(Wall et al., 2010). From this collection, three phages, vB_EcoS_FFH_1 (FFH1),

vB_EcoM_FFH_2 (FFH2) and vB_EcoS_FFH_3 (FFH3), from three geographically

distinct locations and each showing broad lytic ranges (i.e., ability to lyse > 95% of test

isolates) were chosen for challenge experiments described below.

2.3.2 Bacteriophage Kinetics

Single-step growth curves were generated for each phage to measure their lytic

cycles using a protocol described previously (Ellis and Delbruck, 1939) with

modifications. Briefly, individual phages were mixed with 500 µL of freshly prepared E.

coli (108 CFU/mL; strain 309) at a multiplicity of infection (MOI) of 1. The mixture was

incubated at 37 °C for 1 min to allow phage adsorption and then centrifuged at 10,000 ×

g for 30 s. The pellet containing phage- adsorbed bacteria was resuspended in 1 L Luria

Bertani (LB, Becton, Dickinson and Company, Franklin Lakes, NJ) broth containing log-

phase growth E. coli O157:H7. Samples were collected at 5 min intervals for 1.5 h. One-

half of each sample was treated with chloroform (Mallinckrodt Pharmaceuticals, St.

Louis, MO) to quantify assembled phages, whereas the other half was left untreated to

Page 59: Phage therapy applications: Efficacy and impact on the ...

45

45

quantify the phages as infective centers. Phages in each sample were quantified by

standard plaque assay in duplicate. Eclipse period was determined from the curve of

assembled phages, whereas latent period and burst size were determined from the curve

of infective centers.

2.3.3 Transmission Electron Microscopy (TEM)

Phages were concentrated using a polyethylene glycol-8000 precipitation method,

as described by Sambrook et al. (1989). Concentrated phage stocks were then

characterized by transmission electron microscopy (TEM) at the Purdue Life Science

Microscopy Facility (West Lafayette, IN) based on basic morphology including size, tail

structure and head structure. The phage sample was carried on a mesh copper grid with

carbon-coated formvar film and negative stained by 2% uranyl acetate. Images (92,000 ×

magnification) were collected by a FEI/Philips CM-100 TEM, as described previously

(Wall et al., 2010).

2.3.4 Phage Treatment of E. coli O157:H7 Contaminated Food Products

The phage cocktail used in each treatment was a mixture of three wild-type E. coli

O157:H7 phages, FFH1, FFH2, and FFH3, chosen based on in vitro host-range and

growth kinetics. The three phages were mixed in equal amounts and the final

concentration of the phage cocktail was adjusted to 108 PFU/mL. For challenge studies, E.

coli O157:H7 strain 309, isolated from a beef feedlot, was fluorescently labeled via

transformation using a green fluorescent plasmid (pGFP; Clontech, Mountain view, CA).

All food samples tested in this study were purchased from local retail outlets.

Page 60: Phage therapy applications: Efficacy and impact on the ...

46

46

Phage treatment of contaminated ground beef was tested under three conditions:

room temperature (n = 10/treatment, 20 g/sample), refrigeration (n = 16/treatment, 20

g/sample), and undercooking (n = 10/treatment, 40 g/sample). For phage-treated samples,

individual ground beef samples were simultaneously inoculated with GFP-expressing E.

coli O157:H7 at a ratio of 5 × 105 CFU/g sample and the phage cocktail at an MOI of 1.

Samples receiving E. coli O157:H7 but no phage served as controls. For room

temperature and refrigerated conditions, samples were incubated at room temperature and

4 °C, respectively, for 24 h. To study the phage treatment under undercooked conditions,

both phage-treated and control samples were equally divided into two groups, where they

were cooked to an internal temperature of 63 °C (i.e., the USDA recommendation for

ground beef) or 46 °C (i.e., undercooked). Following cooking, each individual sample

was added to 100 mL peptone water (Becton, Dickinson and Company, Franklin Lakes,

NJ) with thorough mashing. The rinsate of each sample was 10-fold serially diluted in 1×

PBS, and each dilution (100 µL) was plated on LB agar supplemented with 100 µg/mL

ampicillin and incubated at 37°C overnight. Only fluorescent green colonies visualized

under UV light were enumerated.

For each spinach sample, three spinach leaves of similar size (average surface

area ~9 cm2) were selected and placed into a sterile container. All samples (n =

13/treatment) were inoculated with 100 µL mixture of E. coli O157:H7 at a concentration

of 108 CFU/mL by drop application. The phage treated group then received phage

treatment at an MOI of 1 by drop application, whereas the control group did not receive

any phage application. Samples were incubated at room temperature. The concentrations

of E. coli O157:H7 were determined at 24 h, 48 h, and 72 h post-treatment.

Page 61: Phage therapy applications: Efficacy and impact on the ...

47

47

Individual Swiss cheese samples (average surface area ~16 cm2, n = 10/treatment)

were inoculated with 100 µL of E. coli O157:H7 at a concentration of 108 CFU/mL by

drop application. The phage treated group received phage treatment at an MOI of 1 by

drop application, while the control group did not receive any phage application. Samples

were incubated at room temperature for 24 h and E. coli O157:H7 in individual samples

were enumerated.

2.3.5 Isolation of Phage Resistant E. coli O157:H7 Mutants

To isolate phage resistant E. coli O157:H7 mutants, E. coli O157:H7 strains 309

(non- GFP labeled) and 502 were infected with the 16 E. coli O157:H7 phages by spot

inoculation onto an overlay containing one of the two bacteria. After 18 h of co-

incubation, three single colonies, which were designated as 309-PR (phage-resistant) 1,

309-PR4 and 502-PR5, were isolated from FFH1-, FFH4- (vB_EcoM_FFH4) and FFH5-

(vB_EcoS_FFH5) lysed clear zones. The three colonies were purified, confirmed by PCR

(stx1, stx2 and eae, primers are listed in Table 1) and biochemical assay (API 20E,

BioMerieux Vitch, Inc., Hazelwood, MO) as E. coli O157:H7 and confirmed as phage

resistant by standard plaque assay.

2.3.6 Phage Adsorption in Phage Resistant E. coli O157:H7 Mutants

Bacterial cultures were grown to log-phase and then infected with corresponding

phages at an MOI of 0.1. Samples were taken in duplicate at 5 and 10 min post-infection

and centrifuged immediately at 10,000 × g for 10 s to separate unattached phage particles

Page 62: Phage therapy applications: Efficacy and impact on the ...

48

48

from bacteria. Titers of unattached phage in the supernatant were determined using

standard plaque assay. The experiments were independently conducted in triplicate.

2.3.7 In Vitro Stability of Phage Resistance

Phage resistant E. coli O157:H7 strains were subcultured in vitro in LB broth

every 12 h for 8 passages. The efficiency of plating (EOP) was determined for each

subculture by dividing the phage titer of the subculture by the phage titer of the phage

susceptible E. coli O157:H7 culture.

2.3.8 Fitness of Phage Resistant E. coli O157:H7 Mutants

To determine any potential fitness costs associated with phage resistance

development, growth kinetics and ability to adhere to mammalian intestinal cells were

measured in the phage resistant E. coli O157:H7 strains 309-PR1, 309-PR4, and 502-PR5.

To measure bacterial growth kinetics, log-phase cultures of phage susceptible and phage

resistant E. coli O157:H7 strains with identical OD600 values were inoculated at a ratio

of 1:100 to either fresh LB broth medium or autoclaved 10% bovine fecal suspension in

1× PBS. Cultures were sampled in triplicate at the starting time point and every hour for

9 h, and bacteria were enumerated on sorbitol MacConkey agar. Bacterial generation

times during log-phase and plateau concentrations during stationary phase were

determined from growth curves of best fit.

Adhesion experiments were conducted as described previously (Carlson et al.,

2009) with some modifications. Briefly, confluent Caco-2 monolayers were formed in 6-

well tissue culture plates (Corning Inc., Corning, NY) using Dulbecco's modified Eagle's

Page 63: Phage therapy applications: Efficacy and impact on the ...

49

49

medium (Invitrogen, Grand Island, NY) containing 20% heat-inactivated fetal bovine

serum (Atlanta Biologicals, Norcross, GA). Confluent Caco-2 monolayers were

inoculated with log-phase phage resistant and phage susceptible bacterial cultures at an

MOI of 50 and incubated at 37°C for 3 h. After infection, unattached bacteria were

removed by washing the monolayer three times with 1× PBS. The monolayer was treated

with 0.25% trypsin and attached bacteria were enumerated on sorbitol MacConkey agar.

The adhesion assay of each isolate was conducted independently in duplicate wells four

times. Adhesion percentage was calculated as the concentration of recovered E. coli

O157:H7 (CFU) compared with that of the inoculum.

2.3.9 Statistical Analysis

Statistical analysis was performed with GraphPad Prism (version 5.0) software

(GraphPad Software, San Diego, CA). Paired t-tests were used to determine if phage

treatment significantly reduced the population of viable E. coli O157:H7 on contaminated

ground beef and cheese at room temperature and refrigerated temperature. One-way

ANOVA and Tukey- Kramer multiple comparisons tests were used for undercooked meat

samples and Caco-2 cell adhesion data. Two-way ANOVA was used for spinach samples

and to compare the percentages of unabsorbed phages in adsorption assays. Treatments

were considered statistically different at P < 0.05. A lack-of-fit test was used to compare

growth curves between phage susceptible and phage resistant strains. Linear regression

was used to evaluate in vitro stability of phage resistance by examining any trends (i.e.,

decrease or increase in EOP) over 8 passages.

Page 64: Phage therapy applications: Efficacy and impact on the ...

50

50

2.4 Results

2.4.1 Phage Characterization

The three phages had similar lytic cycles, which were completed within 60 min.

With each phage, attachment was observed within 1 min and cycle started immediately.

The eclipse periods for FFH1, FFH2, and FFH3 were approximately 10 min, 25 min and

20 min and the latent periods were 33 min, 35 min, and 28 min, respectively. The burst

sizes of FFH1, FFH2, and FFH3 were 30.9, 18.6 and 524.8 PFU/cell (Figure 1).

Transmission electron microscopy revealed the morphology of the 3 coliphages

and showed that they belonged to 2 distinct families. The FFH1, FFH2, and FFH3 phages

were all tailed phages with icosahedral heads. Phage FFH2 was a member of the

Myoviridae family with a larger head and a contractile tail consisting of a baseplate and

spikes. Phages FFH1 and FFH3, as members of the Siphoviridae family, had longer but

noncontractile and flexible tails (Figure 2).

2.4.2 Phage Treatment of E. coli O157:H7 Contaminated Food Products

Concentrations of E. coli O157:H7 in phage treated ground beef were 1.97 ± 0.24

log10 CFU/mL less than those in untreated ground beef when samples were stored at

room temperature for 24 h and 0.48 ± 0.18 log10 CFU/mL less when stored under

refrigeration (Figure 3A and 3B; P < 0.05). When contaminated beef samples were

undercooked (i.e. internal temperature of 46°C), concentrations of E. coli O157:H7 in

phage treated ground beef were 0.56 ± 0.38 log10 CFU/mL less than those found in

untreated, undercooked ground beef (P < 0.05, Figure 3C). There was no significant

Page 65: Phage therapy applications: Efficacy and impact on the ...

51

51

difference in E. coli O157:H7 concentrations between phage treated and non-treated

groups when contaminated beef was cooked to an internal temperature of 63°C (Figure

3C).

Application of the phage cocktail reduced (P < 0.001) the concentration of viable

E. coli O157:H7 on spinach surfaces. Concentrations of E. coli O157:H7 on phage-

treated spinach were 3.28 ± 0.74 log10 CFU/mL, 2.88 ± 0.74 log10 CFU/mL and 2.77 ±

0.74 log10 CFU/mL less than E. coli O157:H7 concentrations on untreated spinach after

storage at room temperature for 24 h, 48 h and 72 h, respectively (Figure 4). The effect of

phage treatment on E. coli O157:H7 reduction was consistent over time.

There were no significant differences between the concentrations of recovered E.

coli O157:H7 in phage-treated cheese versus untreated cheese. After 24 h at room

temperature, phage-treated cheese had concentrations of 4.15 log10 CFU/mL rinsate

compared with 4.23 log10 CFU/mL rinsate in untreated cheese.

2.4.3 Kinetics of Phage Infection of Phage Resistant Mutants

Compared with FFH1 infection of phage-susceptible E. coli O157:H7 strain 309

(Figure 1A), which had an eclipse period of 10 min, a latent period of 33 min and a burst

size of 30.9 PFU/cell, no phage proliferation was observed during the infection of FFH1

to phage-resistant strain 309-PR1 (Figure 5A) and, therefore, kinetic indices could not be

calculated. Phage FFH1 titer was observed to slightly decrease over the 1.5 h of sampling.

Phage FFH4 infection of the phage-susceptible E. coli O157:H7 strain 309

(Figure 5B) produced an eclipse period of approximately 30 min, a latent period of 43

min and a burst size of 95.9 PFU/cell. Infection of the phage-resistant strain 309-PR4

Page 66: Phage therapy applications: Efficacy and impact on the ...

52

52

(Figure 5C) produced similar eclipse and latent periods of 30 min and 43 min,

respectively, and a burst size of 43.65 CFU/mL, indicating the 309-PR4 was not

completely resistant to FFH4.

Phage FFH5 infection of the phage-susceptible E. coli O157:H7 strain 502

(Figure 5D) produced an eclipse period of 30 min, a latent period of 50 min and a burst

size of 57.54 PFU/cell. However, similar to the infection of FFH1 to 309-PR1, no phage

proliferation was observed during FFH5 infection of phage-resistant strain 502-PR5

(Figure 5E).

Each phage-resistant E. coli O157:H7 strain was also tested for cross-resistance to

other E. coli O157:H7 phages in our library. In isolate 309-PR1, resistance to FFH1

conferred resistance to two other phages (FFH6 and FFH7). In isolate 309-PR4,

resistance to FFH4 conferred resistance to four other phages (FFH2, FFH3, FFH7 and

FFH8). Similarly, resistance to FFH5 in isolate 502-PR5 conferred resistance to three

other phages (FFH9, FFH10 and FFH11).

2.4.4 Adsorption of Phage to Phage Resistant E. coli O157:H7 Mutants

The titer of adsorbed phages was represented as the initial phage inoculum minus

the recovered phage titer in supernatant. Adsorption of FFH1 to phage resistant E. coli

O157:H7 strain 309-PR1 did not differ compared with FFH1 adsorption to the susceptible

parent strain 309 (P = 0.25, Figure 6A). At 5 min post-infection, 74.11% of the initial

FFH1 inoculum adsorbed in 309-PR1 culture versus 79.33% in 309 culture. At 10 min

post-infection, 84.01% of the initial FFH1 inoculum adsorbed in 309-PR1 culture versus

91.00% in 309 culture.

Page 67: Phage therapy applications: Efficacy and impact on the ...

53

53

Adsorption of FFH4 to phage-resistant E. coli O157:H7 strain 309-PR4 was

significantly less compared with FFH4 adsorption to the susceptible parent strain 309 (P

< 0.01, Figure 6B). At 5 min post-infection, 31.86% of the initial FFH4 inoculum

adsorbed in 309-PR4 culture versus 84.38% in 309 culture. At 10 min post-infection,

41.10% of the initial FFH4 inoculum adsorbed in 309-PR4 culture versus 84.20% in 309

culture.

Similarly, adsorption of FFH5 to phage resistant E. coli O157:H7 strain 502-PR5

was significantly lower than FFH5 adsorption to the phage susceptible parent strain 502

(P < 0.01, Figure 6C). At 5 min post-infection, 8.35% of the initial FFH5 inoculum

adsorbed in 502-PR5 culture vs. 65.41% in 502 culture. After 10 min post-infection,

35.32% of the initial FFH5 inoculum adsorbed in 502-PR5 culture versus 89.65% in 502

culture.

2.4.5 In Vitro Stability of Phage Resistance

The EOP remained constant throughout the sub-culture period in phage resistant

E. coli O157:H7 strains 309-PR1, 309-PR4 and 502-PR5, indicating that phage resistance

was stable in vitro even though phage was withdrawn. While the EOP in the sub-cultures

of 309-PR1 and 502- PR5 remained < 1.27 × 10-8 and < 1.54 × 10-8, respectively, the

EOP in the sub-cultures of 309-PR4 remained as 0.10 ± 0.04.

2.4.6 Fitness of Phage Resistant E. coli O157:H7 Isolates

LB broth was used as a common laboratory growth medium for E. coli and 10%

bovine fecal suspension in 1× PBS was used to mimic a nutrient-limited environment. No

Page 68: Phage therapy applications: Efficacy and impact on the ...

54

54

growth differences were observed among phage susceptible E. coli O157:H7 strain 309

and its two phage-resistant strains (E. coli O157:H7 strains 309-PR1 and 309-PR4) in

doubling time or plateau concentrations during incubation in either LB broth (P = 0.5261,

Figure 7A) or 10% bovine fecal suspension (P = 0.1007, Figure 7B). Similarly, no

growth differences were observed between phage-susceptible E. coli O157:H7 strain 502

and its phage-resistant E. coli O157:H7 strain 502-PR5 in either LB broth (P = 0.2065,

Figure 7C) or 10% bovine fecal suspension (P = 0.1243, Figure 7D).

In terms of bacterial adhesion ability to Caco-2 human intestinal epithelial cells,

all of the tested E. coli O157:H7 strains had low recovered adhesion percentages. The E.

coli O157:H7 strains 309, 309-PR1 and 309-PR4 had recovered adhesion percentages of

2.20 ± 0.49%, 2.06 ± 0.42% and 2.13 ± 0.63%, respectively, and no statistical differences

were observed (P = 0.9833, Figure 8A). The E. coli O157:H7 strains 502 and 502-PR5

had recovered adhesion percentages of 0.92 ± 0.41% and 1.12 ± 0.67%, and no statistical

differences were observed (P = 0.8096, Figure 8B).

Page 69: Phage therapy applications: Efficacy and impact on the ...

55

55

2.5 Discussion

Interest in using bacteriophages as antibacterial interventions has grown due to

the emergence of antibiotic resistant bacteria. The potential of phages in limiting bacterial

infections or contamination has been shown previously (Sharma et al., 2009; Wall et al.,

2010; Loc-Carrillo and Abedon, 2011; Viazis et al., 2011; Hudson et al., 2013); however,

phages are comparatively understudied, which makes it difficult to predict where and

when phage therapy can be most effective and appropriate. In this study, we isolated 16 E.

coli O157:H7 phages from wastewater treatment facilities. Three phages (i.e., FFH1,

FFH2, and FFH3) were chosen from this collection based on their ability to lyse the

greatest number of individual E. coli O157:H7 strains. Here we determined the

antibacterial efficacy of selected phages in various food products, as well as characterized

phage resistance in E. coli O157:H7 strains in order to both gain a better understanding of

some of the basic biological characteristics of wild-type E. coli O157:H7 phages and to

identify phages that could more effectively limit contamination in different food matrices.

Many foods can be easily contaminated during processing, storage and

transportation. Whereas E. coli O157:H7 infections traditionally have been associated

with ground beef, outbreaks in recent years have also been linked to contaminated

produce and other ready-to-eat products (Nyachuba 2010). Our study, as well as those of

others (Leverentz et al., 2003; O’Flynn et al., 2004), has demonstrated the effectiveness

of phage therapy in post-harvest control of foodborne bacterial pathogens in meat and

fresh produce. The results of our study, perhaps for the first time, indicated that applying

phage treatment to contaminated ground beef can effectively reduce E. coli O157:H7

populations in improperly-cooked (i.e., undercooked) meat balls. This evidence provides

Page 70: Phage therapy applications: Efficacy and impact on the ...

56

56

a possible approach to limit outbreaks associated with improperly prepared beef products.

Likewise, the phage cocktail used in our study showed promising results in reducing E.

coli O157:H7 concentration in fresh spinach at room temperature, which was consistent

with another recent report (Abuladze et al., 2008). These observations may contribute to

practical biocontrols during post-harvest processing and transport of fresh spinach,

especially in the cases where normal refrigeration temperatures inadvertently increase, as

in the case with ‘hot trucks’. Our phages, however, were not effective in reducing E. coli

O157:H7 contamination in cheese. Leverentz et al. (2003) reported that levels of L.

monocytogenes-specific phages declined rapidly in fresh-cut apple slices (pH = 4.2), but

not in honeydew melon slices (pH = 5.8). However, the pH of the Swiss cheese used in

our study (i.e., pH = 5.68 to 6.62) was significantly less acidic than that of apples and

was similar to the pH of spinach (i.e., pH = 5.5 to 6.8). Therefore, a low pH seems not to

be the major reason preventing phage function in Swiss cheese. The cheese industry takes

several measures to control phage infection from interfering with the fermentation

process during manufacturing (Emond and Moineau, 2007) and the bacteriophage

inhibitory media left on the cheese may be a threat to phages (Samson and Moineau,

2013). Further studies are required to find the critical points in cheese processing where

phage treatment may be more effective and appropriate.

Phages are obligate parasites that depend on host machinery for replication. As

expected, our phage cocktail consistently performed better at higher temperature (i.e.,

20°C) rather than lower temperature (i.e., 4°C) because 20°C is closer to the optimum

temperature for E. coli O157:H7 (i.e., 37°C). It should be noted, however, that a previous

study using the ECP-100 phage cocktail to treat E. coli O157:H7-contaminated

Page 71: Phage therapy applications: Efficacy and impact on the ...

57

57

cantaloupes indicated that ECP-100 was more effective at low temperature (i.e., 4 °C)

than high temperature (i.e., 20°C) (Sharma et al., 2009). These results may be due to the

different physiological types of coliphages. Therefore, the three phages in our cocktail

may also belong to the high temperature (HT) or mid-temperature (MT) class of phages

which lyse bacteria more effectively at or above 15°C (Seeley et al., 1980).

Previous research showed that the efficacy of phages in reduction of E. coli

appeared to be correlated with the phage concentration used: a greater phage-to-bacteria

ratio resulted in a larger reduction of viable E. coli counts in all foods (Abuladze et al.,

2008; Goode et al., 2003). The bacterial concentration with which we challenged the

foods in our study was much greater than those commonly reported in outbreak

associated foods. The greater inoculation rate was used to avoid situations where phage

treatment reduced bacterial concentrations to levels under the limits of detection (102 to

103 CFU/mL depending on the sample), which obscures results. Therefore, under more

realistic conditions, phage treatment may be more effective than shown here. This may be

true, however, only within a certain range because phage association with host bacteria is

governed by second order kinetics. Therefore, if contamination rates are extremely low,

very high concentrations of phages may be needed to eliminate the target bacteria.

The development of resistance in bacteria is an unintended, however, unavoidable

phenomenon in many anti-bacterial applications. Phage therapy is no exception and the

emergence of phage-resistant bacteria is considered one of the obstacles that may limit

phage therapy from being widely applied (Sulakvelidze et al., 2001; Tanji et al., 2004).

According to previous studies, phage-resistant mutants were frequently detected within

hours to days in bacteria-phage coincubations in liquid media, especially when a single

Page 72: Phage therapy applications: Efficacy and impact on the ...

58

58

phage was used (Bohannan and Lenski, 2000; Mizoguchi et al., 2003; Tanji et al., 2004).

Our study showed similar results with all three phage-resistant E. coli O157:H7 mutants

being isolated after 18 h of coincubation on LB agar. The mechanisms of phage

resistance can be grouped into four major categories: adsorption prevention, prevention

of phage DNA entry, degradation of phage nucleic acid, and abortive infection systems

(Labrie et al., 2010). Among all, adsorption prevention is most commonly reported in

phage resistance E. coli in previous studies. Resistance to T2, T4, T7 and several E. coli

O157:H7-specific phages in E. coli has been associated with alteration or deletion of

phage adsorption receptors in bacterial cell surface structures, such as lipopolysaccharide

(LPS), OmpA, OmpC and OmpF (Bohannan et al., 2000; Morita et al., 2002; Tanji et al.,

2004). In addition, adsorption prevention in E. coli O157:H7 was also observed through

secretion of extracellular matrix (Mizoguchi et al., 2003). All three phage resistant E. coli

O157:H7 strains examined in this study had similar colony morphology compared with

their phage-susceptible parent strains. Based on phage adsorption assays, we determined

that the development of phage resistance in E. coli O157:H7 strains 309-PR4 and 502-

PR5 were, at least in part, due to phage adsorption prevention. Further analysis of LPS

and outer membrane protein profiles in both phage susceptible and phage resistant strains

may help identify phage receptors and determine how the alteration or deletion of phage

receptors contributed to phage resistance. The adsorption of phage to strain 309-PR1, on

the other hand, was not altered. However, no phage amplification was observed in single-

step growth curves, indicating phage resistance in 309-PR1 was due to mechanisms other

than adsorption prevention. More studies are needed to further determine the exact

mechanism of phage resistance in 309- PR1.

Page 73: Phage therapy applications: Efficacy and impact on the ...

59

59

A previous study (Scott et al., 2007) reported a reversion from phage resistance to

phage susceptibility in Campylobacter jejuni when the bacteria were inoculated into

chickens in the absence of phages. Similar reversions to phage susceptibility were also

observed in phage resistant E. coli O157:H7 mutants in vitro when they were passaged

through 50 generations (O’Flynn et al., 2004). However, we did not observe any

reversion to phage susceptibility in any of our phage resistant E. coli O157:H7 strains

during 8 sub-culture passages (equal to more than 250 generations). Other studies

(Quiberoni et al., 1999; Binetti et al., 2007) showed that the stability of phage resistance

could vary greatly among mutants with some mutants remaining phage resistant

throughout the test period and other mutants reverting within the first few sub-cultures.

The particular mechanisms of phage resistance and the genetic background of bacteria are

probably key factors that determine the stability of phage resistance.

Fitness cost was another phenomenon frequently associated with the development

of phage resistance. Bacterial cell surface structures that serve as receptors of phage

adsorption are often involved in other various aspects of bacterial physiology. The outer

membrane proteins, such as OmpA, OmpC and OmpF, function both as structural

proteins to maintain the outer membrane for the survival of bacteria in harsh

environments (Sonntag et al., 1978) and as porins for passive diffusion of nutrients,

toxins, and waste (Koebnik et al., 2000). In pathogenic bacteria, some cell surface

structures function as virulence factors that facilitate bacterial colonization and confer

serum resistance (Weiser and Gotschlich, 1991; Cornick et al., 2002). Therefore, phage

resistant mutants could be out-competed during the absence of phage due to a fitness cost.

However, unlike other reports showing that phage resistant mutants had decreased growth

Page 74: Phage therapy applications: Efficacy and impact on the ...

60

60

or attenuated colonization and invasion ability (Scott et al., 2007; Gómez and Buckling,

2011), no fitness cost in terms of growth kinetics or ability to adhere to mammalian

intestinal cells was observed in our phage resistant E. coli O157:H7 strains. Previous

studies (Bohannan and Lenski, 2000; Lopez-Pascua and Buckling, 2008) indicated that

environment could affect the cost of phage resistance. Phage resistance in bacteria

growing in environments with lower productivity tends to have a more apparent fitness

cost. It is possible that the cost of resistance in our mutants was not enough to cause any

growth or adhesion difference under laboratory settings. However, the environments

where phage treatments might be applied, such as animal intestinal tract, animal carcass

and food surface, are much more limited in nutrients while containing very complex

microbial communities. In this situation, a relatively low fitness cost may be enough to

cause the phage-resistant mutants to be out-competed when no phage selection pressure is

presented.

The ability of phage to evolve and potentially overcome phage resistance in

bacteria is commonly considered a major advantage that distinguishes phage therapy

from the use of antibiotics. Corresponding to the alteration of phage receptors in bacteria,

some phages, such as T7, evolved with host-range mutations that lead to infections of

both phage-resistant and phage- susceptible bacteria (Lenski and Levin, 1985). A similar

phenomenon was observed during the coincubation of FFH5 with phage resistant strain

502-PR5 in this study (data not shown), where a new phage was detected after 5 h of co-

incubation and confirmed to be lytic to both phage resistant strain 502-PR5 and phage

susceptible parent strain 502. Further genome analysis will be used to determine if the

new phage is a descendent of FFH5 and, if so, to determine the cause of the phenotype.

Page 75: Phage therapy applications: Efficacy and impact on the ...

61

61

One strategy that has been shown to be effective in mitigating phage resistance is

the use of a cocktail of multiple distinct phages. Compared with the use of a single phage,

the use of a phage cocktail could delay the development of phage resistance (Tanji et al.,

2004) due to low probability of a bacterium simultaneously developing multiple resistant

mechanisms or alterations in multiple phage receptors. However, in our situation,

resistance to the original test phage (i.e., FFH1, FFH4 or FFH5) conferred cross-

resistance to at least two other phages in our library. It should be noted, however, that

there is little clonal variation among E. coli O157:H7 strains and cross-resistance may be

much less in other bacterial strains. Nevertheless, it would be prudent to include several

phages with distinct genetic backgrounds to optimize phage cocktails in reducing the

emergence of phage resistance within a treatment period.

Page 76: Phage therapy applications: Efficacy and impact on the ...

62

62

2.6 Summary and Conclusions

Phage-based technologies have great potential as biocontrol agents in limiting both

disease transmission and contamination in food animal production. Here we

demonstrated that a rationally designed cocktail of phages chosen based on in vitro

kinetics, killing efficiencies and host range, significantly reduced E. coli O157:H7

contamination in different food matrices. By examining the efficacy of the treatment at

different temperatures, we hoped to better predict when and where phage-based

technologies can be applied most appropriately and effectively. Our results indicate,

however, that the development of resistance could interfere with the long-term efficacy of

phage-based therapeutics in manners similar to other antibacterial treatments such as

antibiotics. Therefore, prior to the widespread use of phages, it would be prudent to focus

research on better understanding how this resistance develops in order to develop

strategies that overcome or mitigate its effects.

Page 77: Phage therapy applications: Efficacy and impact on the ...

63

63

2.7 Note

Yanying Pan contributed to the study of evaluating the efficacy of phage treatments

to reduce E. coli O157:H7 contamination in foods and the results were also included in

her Master’s thesis.

Page 78: Phage therapy applications: Efficacy and impact on the ...

64

64

0 10 20 30 40 50 60100

101

102

103

104

105

106

Infective centerAssembled phage

L

E

B

Time, min

Phag

e tite

r, PF

U/m

L

0 10 20 30 40 50 60100

101

102

103

104

105

106

Infective centerAssembled phage

E

L

B

Time, min

Phag

e tite

r, PF

U/m

L

0 10 20 30 40 50 60100

101

102

103

104

105

106

Infective centerAssembled phage

EL

B

Time, min

Phag

e tite

r, PF

U/m

L

Figure 1. Growth kinetics of E. coli O157:H7 phages. Single-step growth curves of

FFH1 (A), FFH2 (B), and FFH3 (C). PFU = plaque forming units, E = eclipse period, L =

latent period and B = burst size.

C

A

B

C

A

Page 79: Phage therapy applications: Efficacy and impact on the ...

65

65

Figure 2. Transmission electron microscopy images of E. coli O157:H7 phages. FFH1

(A) and FFH3 (C), members of the Siphoviridae family, have small heads and long, non-

contractile tails. FFH2 (B), a member of the Myoviridae family, has a large head and a

short, contractile tail. Magnification = 92,000×. Bars = 0.2 µm.

C B A

Page 80: Phage therapy applications: Efficacy and impact on the ...

66

66

Control Phage treated4

5

6

7

8

9ControlPhage treated

*E.

col

i, lo

g 10 C

FU/m

L

Control Phage treated0

2

4

6ControlPhage treated

*

E. c

oli,

log 10

CFU

/mL

46°C

+ E. c

oli

46°C

+ E. c

oli + phag

e

63°C

+ E. c

oli

63°C

+ E. c

oli + phag

e0

1

2

3

4

46°C + E. coli46°C + E. coli + phage63°C + E. coli63°C + E. coli + phage

*

E. c

oli,

log 10

CFU

/mL

Figure 3. Reduction of E. coli O157:H7 contamination in phage treated ground beef at

room temperature (A), under refrigeration (B), and when undercooked (C). Bars indicate

means within treatments. * = statistical differences (P <0.05) in mean bacterial

concentrations between treatment groups and within temperatures.

A

B

C

Page 81: Phage therapy applications: Efficacy and impact on the ...

67

67

0 24 48 720

2

4

6

8ControlBacteriaPhage treated

* * *

Time, hours

E. c

oli,

log 10

CFU

/mL

Figure 4. Reduction of E. coli O157:H7 contamination in phage treated spinach. * =

statistical differences (P <0.05) in mean bacterial concentrations between treatment

groups and within time points.

Page 82: Phage therapy applications: Efficacy and impact on the ...

68

68

0 10 20 30 40 50 60 70 80 90100

101

102

103

104

105

106Infective centerAssembled phage

Time, min

Phag

e tite

r, PF

U/m

L

0 10 20 30 40 50 60 70 80 90100

101

102

103

104

105

106Infective centerAssembled phage

EL

B

Time, min

Phag

e tite

r, PF

U/m

L

0 10 20 30 40 50 60 70 80 90100

101

102

103

104

105

106

EL

B

Assembled phageInfective center

Time, min

Phag

e tite

r, PF

U/m

L

0 10 20 30 40 50 60 70 80 90100

101

102

103

104

105

106

107Infective centerAssembled phage

EL

B

Time, min

Phag

e tite

r, PF

U/m

L

0 10 20 30 40 50 60 70 80 90100

101

102

103

104

105

106

107Infective centerAssembled phage

Time, min

Phag

e tite

r, PF

U/m

L

Figure 5. Phage growth kinetics in phage susceptible and phage resistant E. coli O157:H7.

Single-step growth curves of FFH1 infection in the phage resistant E. coli O157:H7 strain

309-PR1 (A), FFH4 infection in phage susceptible parent E. coli O157:H7 strain 309 (B)

and its phage resistant strain 309-PR4 (C), FFH5 infection in the phage susceptible parent

E. coli O157:H7 strain 502 (D) and its resistant strain 502-PR5 (E). PFU = plaque

forming units, E = eclipse period, L = latent period and B = burst size.

B

D E

A

C

Page 83: Phage therapy applications: Efficacy and impact on the ...

69

69

5 100

20

40

60

80

100309309-PR1

Time, min

Ads

orbe

d ph

ages

, %

5 100

20

40

60

80

100309309-PR4

* *

Time, min

Ads

orbe

d ph

ages

, %

5 100

20

40

60

80

100502502-PR5

**

Time, min

Ads

orbe

d ph

ages

, %

Figure 6. Phage adhesion in phage susceptible and phage resistant E. coli O157:H7. The

percentages of adsorbed phage in (A) FFH1 infection of phage susceptible (309) and

phage resistant (309-PR1) E. coli O157:H7, (B) FFH4 infection of phage susceptible

(309) and phage resistant (309-PR4) E. coli O157:H7 and (C) FFH5 infection of phage

susceptible (502) and phage resistant (502-PR5) E. coli O157:H7. * = statistical

differences (P <0.05) in adsorption % of different bacterial strains within time points.

C

A

B

Page 84: Phage therapy applications: Efficacy and impact on the ...

70

70

0 1 2 3 4 5 6 7 8 9105

106

107

108

109309309-PR1309-PR4

Time, hours

E. c

oli,

CFU

/mL

0 1 2 3 4 5 6 7 8 9105

106

107

108

109309309-PR1309-PR4

Time, hours

E. c

oli,

CFU

/mL

0 1 2 3 4 5 6 7 8 9105

106

107

108

109502502-PR5

E. c

oli,

CFU

/mL

Time, hours

0 1 2 3 4 5 6 7 8 9105

106

107

108

109502502-PR5

Time, hoursE.

col

i, C

FU/m

L

Figure 7. Growth characteristics of phage susceptible versus phage resistant E. coli

O157:H7. Bacterial growth curves of phage susceptible E. coli O157:H7 strain 309 and

phage resistant strains 309-PR1 and 309-PR4 in (A) Luria Bertani (LB) broth and (B)

10% bovine fecal suspension. Bacterial growth curves of phage susceptible E. coli

O157:H7 strain 502 and phage resistant strain 502-PR5 in (C) LB broth and (D) 10%

bovine fecal suspension

D

A B

C

Page 85: Phage therapy applications: Efficacy and impact on the ...

71

71

309 309-PR1 309-PR40

1

2

3

4

Bacterial strains

Rec

over

ed a

dhes

ion

perc

enta

ge, %

502 502-PR50

1

2

3

4

Bacterial strains

Rec

over

ed a

dhes

ion

perc

enta

ge, %

Figure 8. Adhesion of phage susceptible and phage resistant E. coli O157:H7. Recovered

adhesion percentages of phage susceptible (309) and phage resistant (309-PR1 and 309-

PR4) E. coli O157:H7 309 strains (A) and phage susceptible (502) and phage resistant

(502-PR5) E. coli O157:H7 502 strains (B) to Caco-2 human intestinal epithelial cells

after 3 h of infection at an MOI of 50. Bars indicate means within treatments. There were

no differences between treatments.

A

B

Page 86: Phage therapy applications: Efficacy and impact on the ...

72

72

Table 1. Primers used in this study

Primer DNA Sequence (5’ to 3’) Product size (bp)

Stx1-F TAG TGG AAC CTC ACT GAC GC 386

Stx1-R TAT TGT GCG TAA TCC CAC GG

Stx2-F GAC TAT CTT CAT TCA CGG CGC 536

Stx2-R GCC GGG TTC GTT AAT ACG GC

Eae-F GCA TTT GGT CAG GTC GGA GC 270

Eae-R ATC GAA GCC ATT TGC TGG GC

Page 87: Phage therapy applications: Efficacy and impact on the ...

73

73

List of References

Abuladze, T., M. Li, M. Y. Menetrez, T. Dean, A. Senecal, and A. Sulakvelidze. 2008.

Bacteriophages reduce experimental contamination of hard surfaces, tomato,

spinach, broccoli and ground beef by Escherichia coli. Appl. Environ. Microbiol.

74:6230-6238.

Binetti, A. G., N. B. Bailo, and J. A. Reinheimer. 2007. Spontaneous phage-resistant

mutants of Streptococcus thermophilus: isolation and technological characteristics.

Int. Dairy J. 17:343-349.

Bohannan, B. J. M., and R. E. Lenski. 2000. Linking genetic change to community

evolution: insights from studies of bacteria and bacteriophage. Ecol. Lett. 3:362-

377.

Carlson, B. A., K. K. Nightingale, G. L. Mason, J. R. Ruby, W. T. Choat, G. H.

Loneragan, G. C. Smith, J. N. Sofos, and K. E. Belk. 2009. Escherichia coli

O157:H7 strains that persist in feedlot cattle are genetically related and

demonstrate an enhanced ability to adhere to intestinal epithelial cells. Appl.

Environ. Microbiol. 75:5927-5937.

Cornick, N. A., S. L. Booher, and H. W. Moon. 2002. Intimin facilitates colonizaton by

Escherichia coli O157:H7 in adult ruminants. Infect. Immun. 70:2704-2707.

Emond, E., and S. Moineau. 2007. Bacteriophages and food fermentations. In: McGrath,

S., and D. Sinderen, editors. Bacteriophage: genetics and molecular biology.

Caister Academic Press, Norwich, United Kindom. p. 93-124.

Ellis, E. L., and M. Delbruck. 1939. The growth of bacteriophage. J. Gen. Physiol.

22:365-384.

Page 88: Phage therapy applications: Efficacy and impact on the ...

74

74

Gómez, P., and A. Buckling. 2011. Bacteria-phage antagonistic coevolution in soil.

Science.332: 106-109.

Goode, D., V. M. Allen, and P. A. Barrow. 2003. Reduction of experimental Salmonella

and Campylobacter contamination of chicken skin by application of lytic

bacteriophages. App. Env. Micro. 69:5032-5036.

Grauke, L. J., I. T. Kudva, J. W. Yoon, C. W. Hunt, C. J. Williams, and C. J. Hovde.

2002. Gastrointestinal tract location of Escherichia coli O157:H7 in ruminants.

Appl. Environ. Microbiol. 68:2269-2277.

Hudson, J. A., C. Billington, A. J. Cornelius, T. Wilson, S. L. W. On, and A. Premaratne.

2013. Use of a bacteriophage to inactivate Escherichia coli O157:H7 on beef.

Food. Microbiol. 36:14-21.

Koebnik, R., K. P. Locher, and P. Van Gelder. 2000. Structure and function of bacterial

outer membrane proteins: barrels in a nutshell. Mol. Microbiol. 37:239-253.

Labrie, S. J., J. E. Samson, and S. Moineau. 2010. Bacteriophage resistance mechanisms.

Nat. Rev. Microbiol. 8:317-327.

Lenski, R. E., and B. R. Levin. 1985. Constraints on the coevolution of bacteria and

virulent phage: a model, some experiments, and predictions for natural

communities. Am. Nat. 125:585-602.

Leverentz, B., W. S. Conway, M. J. Camp, W. J. Janisiewicz, T. Abuladze, M. Yang, R.

Saftner, and A. Sulakvelidze. 2003. Biochontrol of Listeria monocytogenes on

fresh-cut produce by treatment with lytic bacteriophages and a bacteriocin. Appl.

Environ. Microbiol. 69:4519-4526.

Page 89: Phage therapy applications: Efficacy and impact on the ...

75

75

Loc-Carrillo, C., and S. T. Abedon. 2011. Pros and cons of phage therapy. Bacteriophage

1:111- 114.

Lopez-Pascua, L. D. C., and A. Buckling. 2008. Increasing producitivity accelerates host-

parasite coevolution. J. Evol. Biol. 21: 853-860.

McGee, P., L. Scott, J. J. Sheridan, B. Earley, and N. Leonard. 2004. Horizontal

transmission of Escherichia coli O157:H7 during cattle housing. J. Food. Prot.

67:2651-2656.

Mizoguchi, K., M. Morita, C. R. Fischer, M. Yoichi, Y. Tanji, and H. Unno. 2003.

Coevolution of Bacteriophge PP01 and Escherichia coli O157:H7 in continuous

culture. Appl. Environ. Microbiol. 69: 170-176.

Morita, M., Y. Tanji, K. Mizoguchi, T. Akitsu, N. Kijima, and H. Unno. 2002.

Chracterization of a virulent bacteriophage specific for Escherichia coli O157:H7

and analysis of its cellular receptor and two tail fiber genes. FEMS Microbiol.

Lett. 211:77-83.

Nyachuba, D. G. 2010. Foodborne illness: is it on the rise? Nutr. Rev. 68:257:269.

O'Flynn, G., R. P. Ross, G. F. Fitzgerald, and A. Coffey. 2004. Evaluation of a cocktail

of three bacteriophages for biocontrol of Escherichia coli O157:H7. Appl.

Environ. Microbiol. 70:3417-3424.

Quiberoni, A., J. A. Reinheimer, and V. B. Suarez. 1999. Performance of Lactobacillus

helveticus spontaneous phage-resistant mutants in hard cheese production. Int.

Dairy J. 8:941-949.

Page 90: Phage therapy applications: Efficacy and impact on the ...

76

76

Rangel, J. M., P. H. Sparling, C. Crowe, P. M. Griffin, and D. L. Swerdlow. 2005.

Epidemiology of Escherichia coli O157:H7 outbreaks, United States, 1982-2002.

Emerg. Infect. Dis. 11:603-609.

Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Molecular Cloning: A Laboratory

Manual, Second Edition. Cold Spring Harbor Laboratory Press, Cold Spring

Harbor, NY.

Samson, J. E., and S. Moineau. 2013. Bacteriophages in food fermentations:new frontiers

in a continuous arms race. Annu. Rev. Food Sci. Tech. 4:347-368.

Scallan, E., R. M. Hoekstra, F. J. Angulo, R. V. Tauxe, M. A. Widdowson, S. L. Roy, J.

L. Jones, and P. M. Griffin. 2011. Foodborne Illness Acquired in the United

States-major pathogens. Emerg. Infect. Dis. 17:1339-1340.

Scott, A. E., A. R. Timms, P. L. Connerton, C. L. Carrillo, K. A. Radzum, and I. F.

Connerton. 2007. Genome dynamics of Campylobacter jejuni in response to

bacteriophage predation. PLoS Pathog. 3:1142-1151.

Seeley, N. D., and S. B. Primrose. 1980. The Effect of Temperature on the Ecology of

Aquatic Bacteriophages. J. Gen. Virol. 46:87-95.

Sharma, M., J. R. Patel, W. S. Conway, S. Ferguson, and A. Sulakvelidze. 2009.

Effectiveness of bacteriophages in reducing Escherichia coli O157:H7 on fresh-

cut cantaloupes and lettucet. J. Food Prot. 72:1481-1485. Sonntag, I., H. Schwarz,

Y. Hirota, and U. Henning. 1978. Cell envelope and shape of Escherichia coli:

multiple mutants missing the outer membrane lipoprotein and other major outer

membrane proteins. J. Bacteriol. 136:280-285

Page 91: Phage therapy applications: Efficacy and impact on the ...

77

77

Sulakvelidze, A., Z. Alavidze, and J. G. Jr. Morris. 2001. Bacteriophage therapy.

Antimicrob. Agents Ch. 45:649-659.

Tanji, Y., T. Shimada, M. Yoichi, K. Miyanaga, K. Hori, and H. Unno. 2004. Toward

rational control of Escherichia coli O157:H7 by a phage cocktail. Appl Microbiol.

Biot. 64:270- 274.

Viazis, S., M. Akhtar, J. Feirtag, A. D. Brabban, and F. Diez-Gonzalez. 2011. Isolation

and characterization of lytic bacteriophages against enterohaemorrhagic

Escherichia coli. J. Appl. Microbiol. 110:1323-1331.

Wall, S. K., J. Zhang, M. H. Rostagno, and P. D. Ebner. 2010. Phage therapy to reduce

preprocessing Salmonella infections in market-weight swine. Appl. Environ.

Microbiol. 76:48-53.

Weiser, J. N., and E. C. Gotschlich. 1991. Outer membrane protein A (OmpA)

contributes to serum resistance and pathogenicity of Escherichia coli K-1. Infect.

Immun. 59:2252- 2258.

Page 92: Phage therapy applications: Efficacy and impact on the ...

78

78

CHAPTER 3. COMPLETE GENOME SEQUENCES OF TWO ESCHERICHIA COLI O157:H7 PHAGES EFFECTIVE IN LIMITING CONTAMINATION OF FOOD PRODUCTS

3.1 Abstract

We previously demonstrated that application of bacteriophages significantly

reduced Escherichia coli O157:H7 contamination in spinach and ground beef. Here, we

present the genomic sequences of two bacteriophages, vB_EcoS_FFH_1, a T5-like phage,

and vB_EcoM_FFH_2, an rV5-like phage, used in those treatments.

3.2 Genome Annoucement

Escherichia coli O157:H7 is a shiga toxin-producing food-borne pathogen that

results in over 60,000 illnesses each year in the United States alone (Scallan et al., 2011).

We have employed bacteriophages to limit Salmonella transmission in swine (Wall et al.,

2010) and recently demonstrated that application of lytic bacteriophages to ground beef

and spinach significantly reduced E. coli O157:H7 contamination (Hong et al., 2014). We

selected two E. coli phages (vB_EcoS_FFH_1 [siphovirus] and vB_EcoM_FFH_2

[myovirus]) for genomic sequencing based on their broad spectrum and lytic capacity.

Phage DNA was purified from polyethylene glycol (PEG)-precipitated lysates and

sequenced via pyrosequencing (454; Eurofins MWG Operon, Huntsville, AL) and

sequences were assembled de novo using Newbler (version 2.6). Coding DNA sequences

Page 93: Phage therapy applications: Efficacy and impact on the ...

79

79

(CDSs) were predicted using Glimmer 3.0 (Delcher et al., 1999) and annotation was

performed using BLASTp for homology searching in the non-redundant protein

sequences database in GenBank (Benson et al., 2012). tRNA genes were predicted using

both tRNAscan-SE 1.21 (Schattner et al., 2005) and ARAGORN (Laslett and Canback,

2004). Terminal redundant ends (vB_EcoM_FFH_1) were identified using Tandem

Repeat Finder (Benson, 1999).

The genome of vB_EcoS_FFH_1 has a length of 108,483 bp and a G+C content

of 39.24%. Whole genome alignment revealed that vB_EcoS_FFH_1 showed 87%

homology to T5 (GenBank accession no. AY543070) and therefore was classified as a

T5-like phage. A total of 162 CDSs and 24 tRNA genes were predicted. Similar high

numbers of tRNA genes are found in T5. Of the CDSs, 54 matched proteins with known

functions, while 96 encoded previously identified hypothetical proteins. Twelve CDSs

did not match any proteins in the NCBI non-redundant protein database. We identified

putative Rz and Rz1 genes based on Summer et al. (Summer et al., 2007). Highly similar

sequences are also present in T5, but are not annotated in the three GenBank T5 complete

genomes and other available T5-like phage genomes. One section (79,918 to 84,241) of

the vB_EcoS_FFH_1 genome appeared largely absent from the three GenBank annotated

T5 genomes, but present in the bV_EcoS_AKFV33 genome (another T5-like phage).

Two putative tail fiber proteins and one hypothetical protein were identified in this

section.

The genome of vB_EcoM_FFH_2 has a length of 139,020 bp and a G+C content

of 43.61%. Whole genome alignment revealed that vB_EcoM_FFH_2 shared 93%

nucleotide homology to E. coli phage rV5 (GenBank accession no. DQ832317),

Page 94: Phage therapy applications: Efficacy and impact on the ...

80

80

indicating that vB_EcoM_FFH_2 is an rV5-like phage. A total of 220 CDSs and 6 tRNA

genes were predicted. Of the CDSs, 57 matched proteins with known functions, while

156 matched previously identified hypothetical proteins. Seven CDSs were not

homologous to any existing proteins in the NCBI non-redundant protein database.

Several complete genomes of rV5-like viruses are now available. The viruses share a

high number of proteins, but based on whole-genome comparisons, two rV5-like sub-

groups may exist, rV5 and Salmonella phage PVP-SE1 (Truncaite et al., 2012; Kropinski

et al., 2013; Kim et al., 2014). vB_EcoM_FFH_2 has significantly more sequence

similarity to rV5 (both of which were isolated using E. coli O157:H7), which would

make it a member of the rV5 sub-group.

The complete sequences of vB_EcoS_FFH_1 and vB_EcoM_FFH_2 were

deposited in GenBank under the accession numbers KJ190157 and KJ190158,

respectively.

Page 95: Phage therapy applications: Efficacy and impact on the ...

81

81

List of References

Benson G. 1999. Tandem repeats finder: a program to analyze DNA sequences. Nucleic

Acids Res. 27:573–580.

Benson D. A., I. Karsch-Mizrachi, K. Clark, D. J. Lipman, and E. W. Sayers. 2012.

GenBank. Nucleic Acids Res. 40:D48–D53.

Delcher A. L., D. Harmon, S. Kasif, O. White, and S. L. Salzberg. 1999. Improved

microbial gene identification with GLIMMER. Nucleic Acids Res. 27:4636–4641.

Hong Y., Y. Pan, and P. D. Ebner. 2014. Meat science and muscle biology symposium:

development of bacteriophage treatments to reduce Escherichia coli O157:H7

contamination of beef products and produce. J. Anim. Sci. 92:1366–1377.

Kim H., S. Heu, and S. Ryu. 2014. Complete genome sequence of enterobacteria phage

4MG, a new member of the subgroup “PVP-SE1-like phage” of the “rV5-like

viruses.” Arch. Virol. 159(11): 3137-40.

Kropinski A. M., T. Waddell, J. Meng, K. Franklin, H. W. Ackermann, R. Ahmed, A.

Mazzocco, J. Yates, E. J. Lingohr, and R. P. Johnson. 2013. The host-range,

genomics and proteomics of Escherichia coli O157:H7 bacteriophage rV5. Virol.

J. 10:6.

Laslett D. and B. Canback. 2004. ARAGORN, a program to detect tRNA genes and

tmRNA genes in nucleotide sequences. Nucleic Acids Res. 32:11–16.

Scallan E., R. M. Hoekstra, F. J. Angulo, R.V. Tauxe, M. A. Widdowson, S. L. Roy, J. L.

Jones, and P. M. Griffin. 2011. Foodborne illness acquired in the United States.

Emerg. Infect. Dis. 17:1339–1340.

Page 96: Phage therapy applications: Efficacy and impact on the ...

82

82

Schattner P., A. N. Brooks, and T. M. Lowe. 2005. The tRNAscan-SE, snoscan and

snoGPS web servers for the detection of tRNAs and snoRNAs. Nucleic Acids Res.

33:686–689.

Truncaite L., E. Simoliunas, A. Zajanckauskaite, L. Kalinene, R. Mankeviciute, J.

Staniulis, V. Klausa, and R. Meskys. 2012. Bacteriophage vB_EcoM_FV3: a new

member of “rV5-like viruses.” Arch. Virol. 157:2413–2435.

Wall S. K., J. Zhang, M. H. Rostagno, and P. D. Ebner. 2010. Phage therapy to reduce

preprocessing Salmonella infections in market-weight swine. Appl. Environ.

Microbiol. 76:48–53.

Page 97: Phage therapy applications: Efficacy and impact on the ...

83

83

CHAPTER 4. TREATMENT OF SALMONELLA CONTAMINATED EGGS AND PORK WITH A BROAD-SPECTRUM, SINGLE BACTERIOPHAGE:

ASSESSMENT OF EFFICACY AND RESISTANCE DEVELOPMENT

4.1 Abstract

Salmonella remains a leading cause of foodborne illness, with Salmonella

Typhimurium and Salmonella Enteritidis being two predominant serovars in both the US

and Europe. Numerous studies have focused on assessing the efficacy of using phage-

based methods to eliminate or control Salmonella contamination in various food products.

Due to the narrow spectrum of phages, many of these studies used mixtures of several

distinct phages to expand the treatment spectrum, but also to prevent problems that may

arise due to the development of phage resistance. However, there is little practical

evidence to substantiate the need for “cocktails” that contain numerous unrelated phages.

In this study, we tested the ability of a single, well-characterized broad-spectrum

Salmonella phage, vB_SalS_SJ_2 (SJ2), to reduce Salmonella Typhimurium and

Salmonella Enteritidis in ground pork and eggs. Compared to untreated samples, phage

treatment significantly (P<0.05) reduced Salmonella Typhimurium by 1.65 log10

CFU/mL, 1.39 log10 CFU/mL, 0.29 log10 CFU/mL and 0.43 log10 CFU/mL in ground

pork and by 3.14 log10 CFU/mL, 0.49 log10 CFU/mL, 0.61 log10 CFU/mL and 0.51 log10

CFU/mL in liquid egg after treatment at room temperature for 24 h and 48 h and at

refrigeration for 24 h and 1 wk, respectively. Similarly, phage treatment significantly (P <

Page 98: Phage therapy applications: Efficacy and impact on the ...

84

84

0.05) reduced Salmonella Enteritidis by 1.04 log10 CFU/mL and 1.30 log10 CFU/mL in

ground pork and by 0.93 log10 CFU/mL and 0.61 log10 CFU/mL in liquid egg after

treatment at room temperature for 24 h and 48 h, respectively. Under refrigeration,

significant reductions (P<0.05) of Salmonella Enteritidis were only seen in phage-treated

meat (0.25 log10 CFU/mL and 0.64 log10 CFU/mL reductions at 24 h and 1 wk,

respectively). No reductions of Salmonella Typhimurium or Salmonella Enteritidis were

observed under refrigerated conditions for 2 wk in either food product. The susceptibility

of Salmonella isolates to phage SJ2 was also tested after each phage treatments.

Significantly (P<0.05) higher percentages of phage resistant Salmonella Typhimurium

(92.50% vs. 0.56% of control) and Salmonella Enteritidis (50.83% vs. 0.56% of control)

isolates were observed in phage-treated liquid egg samples than non-treated samples after

incubation at room temperature for 48 h. Interestingly, such differences were not

observed between phage-treated and non-treated ground pork samples that were

incubated under the same conditions, indicating food matrix could influence the

emergence of phage resistance in foods. Taken together, this study demonstrated that a

single, broad-spectrum phage (SJ2) can effectively reduce Salmonella and the

development of phage resistance may be dependent on the targeted food matrix.

Page 99: Phage therapy applications: Efficacy and impact on the ...

85

85

4.2 Introduction

Salmonella is a ubiquitous foodborne pathogen that has been associated with

recalls of a wide variety of foods. Although multiple strategies have been employed to

prevent and eliminate Salmonella contamination at various points in the production and

processing chains, Salmonellosis remains a top foodborne illness in the US (Scallan et al.,

2011), indicating there is a need for more effective anti-Salmonella strategies. Among

many others, phage-based methods have shown potential to control Salmonella in various

food matrices and offer the unique advantages of targeting specific bacteria while being

natural and self-limiting. Previous studies have shown the efficacy of phages to reduce

Salmonella, mainly serovars Typhimurium and Enteritidis, in various food under

laboratory conditions, including chicken breast, shell and liquid eggs, lettuce, and juice

(Leverentz et al., 2001; Hungaro et al., 2013; Spricigo et al., 2013). The use of phages is

not limited to the control of Salmonella, as others have employed phages to target various

other pathogens including E. coli O157:H7 (Abuladze et al., 2008; Carter et al., 2012;

Hong et al., 2014), Campylobacter (Kittler et al., 2013), and Listeria (Leverentz et al.,

2003; Anany et al., 2011). Currently, there are commercially available phage-based

products for food processing in both the US and elsewhere. Some products have received

further approval as GRAS for use in ready-to-eat food or OMRI for use in organic

production.

The majority of the aforementioned studies used phage cocktails, or mixtures of

several distinct phages, largely in attempt to increase the host-spectrum as phages can be

highly specific for certain bacterial species and sub-species. Such high host specificity

can be problematic in reality where phages generally are applied before the exact bacteria

Page 100: Phage therapy applications: Efficacy and impact on the ...

86

86

in the foods or environment are known. Alternatively, using broad-spectrum phages

could also address the problem. Compared to the use of phage cocktail, however,

treatments based on single-phage can been more precisely defined and thus could more

easily receive regulatory approval (Carlton et al., 2005). A major concern, however, is the

development of resistance, which would theoretically be slower or absent if phage

treatments consist of several unrelated phages. To date, the efficacy of using a single,

broad-spectrum phage to reduce foodborne pathogens in foods has been far less studied

than the use of cocktails. In this study, we tested the potential of the well-characterized,

broad-spectrum Salmonella phage vB_SalS_SJ_2 (SJ2) to reduce Salmonella

Typhimurium and Salmonella Enteritidis in ground pork and liquid eggs. We also

measured the development of bacterial resistance to the phage to determine its impact on

efficacy.

Page 101: Phage therapy applications: Efficacy and impact on the ...

87

87

4.3 Materials And Methods

4.3.1 Bacteria and Bacteriophage

Salmonella Typhimurium γ4232 (nalidixic acid resistant) was previously isolated

from a diseased pig (Ebner and Mathew, 2000) and Salmonella Enteritidis was kindly

provided by Dr. Arun K. Bhunia (Purdue University, West Lafayette, IN). Phage SJ2 was

previously isolated from a wastewater treatment plant in Indiana using Salmonella

Typhimurium as a host (Wall et al., 2010). The phage has been well characterized both

genetically and kinetically. It is a siphorvirus with a genome size of 152,460 bp and

contains 197 predicted genes. The phage also showed the broadest spectrum among a

library of Salmonella phages, with high killing efficiency against Salmonella serovars

Typhimurium, Enteritidis, Indiana, and Kentucky (Zhang et al., 2015).

4.3.2 Phage Treatment of Salmonella Contaminated Food Products

Ground pork and shell eggs were purchased from local retail stores. All samples

were tested for Salmonella prior to any further phage testing. To test phage efficacy in

ground pork, ten samples were used for both control and phage treatment. Fifteen gram

samples were placed in a sterile stomacher bag (VWR International, Radnor,

Pennsylvania) and inoculated with 107 CFU of either Salmonella Typhimurium or

Salmonella Enteritidis in 100ul of Luria-Bertani (LB) broth, and massaged by hand

thoroughly to ensure even contamination throughout the sample. Phage treated samples

were then inoculated with 108 PFU of phage SJ2 suspended in 100 uL of phosphate

buffered saline (PBS), while untreated samples received a same amount of PBS only. All

samples were once again massaged by hand after inoculation and incubated under one of

Page 102: Phage therapy applications: Efficacy and impact on the ...

88

88

five conditions: room temperature for 24 h, room temperature for 48 h, 4 °C for 24 h,

4 °C for 1 wk, and 4 °C for 2 wk. After incubation, meat samples were resuspended in

100 mL of buffered peptone water (Becton, Dickinson and Company, Franklin Lakes,

NJ), the suspension was serially diluted and the concentration of Salmonella were

determined by plating on either XLT4 agar plates (Becton, Dickinson and Company,

Franklin Lakes, NJ) containing 50 ug/mL of nalidixic acid (Alfa Aesar, Ward Hill, MA)

for Salmonella Typhimurium or XLD agar plates (Becton, Dickinson and Company,

Franklin Lakes, NJ) for Salmonella Enteritidis, as XLT4 plates greatly inhibited the

growth of Salmonella Enteritidis strain used in this study (data not shown). Experiment

was repeated independently three times.

To test the efficacy of phage treatment in liquid egg, ten samples were used for

both control and phage treatment. Egg shells were wiped with 70% alcohol and air-dried

before breaking to avoid unintended contamination of Salmonella from outer surfaces.

Eggs were then mixed and homogenized at high speed. Liquid egg samples (15 mL)

were inoculated with 107 CFU of Salmonella Typhimurium or Salmonella Enteridis in

100 ul volume and vortexed to mix thoroughly. Phage treated samples were treated with

108 PFU of SJ2 in 100 ul of PBS and control samples received a same amount of PBS

only. Samples were once again vortexed after inoculation and incubated under one of

same five conditions previously described for pork experiments. After incubation,

Salmonella concentrations were quantified as previously described. Each experiment was

repeated independently three times.

Page 103: Phage therapy applications: Efficacy and impact on the ...

89

89

4.3.3 Phage Susceptibility of Salmonella After Phage Treatment

To test the development of phage resistance in the challenge Salmonella after

treatment with phage SJ2 under different conditions, 24 Salmonella isolates from each

phage treated sample and 12 Salmonella from each control samples were collected.

Isolates were picked from enumeration plates using sterilized toothpicks and were

resuspended in individual tubes containing 900 ul of PBS. Following a brief vortex, a 5

uL of each suspension was dropped onto an overlay containing a high concentration of

SJ2 and the plates were incubated at 37 °C overnight. Salmonella isolates that formed

complete bacterial lawns at dropped spots were considered resistant to phage SJ2.

4.3.4 Statistical Analysis

A random block design with replications was used to test the effect of phage SJ2

in reducing Salmonella contamination in both ground pork and liquid egg. Both the

concentration of Salmonella in foods and the percentage of phage resistant colonies were

analyzed using a mixed model analysis of variance (SAS 9.4) with the percentages of

phage resistant colonies being transformed into ranks.

Page 104: Phage therapy applications: Efficacy and impact on the ...

90

90

4.4 Results

4.4.1 Phage Treatment of Salmonella Contaminated Ground Pork

In Salmonella Typhimurium contaminated ground pork, samples that received

treatment of phage SJ2 had significantly (P<0.05) lower concentrations of Salmonella

Typhimurium than untreated samples by 1.65 Log10 CFU/mL (Fig 1A) and 1.39 Log10

CFU/mL (Fig 1B) after incubation at room temperature for 24 and 48 h, respectively.

While less effectively numerically, the concentrations of Salmonella Typhimurium in SJ2

treated ground pork were also significantly lower (P<0.05) than those in control samples

by 0.29 Log10 CFU/mL (Fig 1C) and 0.43 Log10 CFU/mL (Fig 1D) after incubation at

4 °C for 24 hr and 1 wk, respectively. However, the concentrations of Salmonella

Typhimurium in ground pork recovered after incubation at 4 °C for 2 wk were not

different between control and phage treated groups (Fig 1E).

Similarly, in Salmonella Enteritidis contaminated ground pork, samples that were

treated with SJ2 had significantly (P<0.05) lower concentrations of Salmonella

Enteritidis than untreated samples by 1.04 Log10 CFU/mL (Fig 2A) and 1.30 Log10

CFU/mL (Fig 2B) after incubation at room temperature for 24 or 48 h, respectively. The

reduction was again lower under refrigeration (4 °C), but still significant, with the

concentrations of Salmonella Enteritidis lower in SJ2-treated than in control samples by

0.25 Log10 CFU/mL (Fig 2C) and 0.64 Log10 CFU/mL (Fig 2D) after 24 and 48 h,

respectively. No difference was observed between control and phage treated samples in

the concentration of Salmonella Enteritidis after incubation at 4 °C for 2 wk (Fig 2E).

Page 105: Phage therapy applications: Efficacy and impact on the ...

91

91

4.4.2 Phage Treatment of Salmonella Contaminated Liquid Egg

In Salmonella Typhimurium contaminated liquid eggs, the concentration of

Salmonella Typhimurium in SJ2-treated samples was significantly (P<0.05) lower than

that in control samples by 3.14 Log10 CFU/mL (Fig 3A) and 0.49 Log10 CFU/mL (Fig 3B)

after incubation at room temperature for 24 h and 48h, respectively. Under refrigeration,

the concentrations of Salmonella Typhimurium in SJ2-treated samples were lower than

control samples by 0.61 Log10 CFU/mL (P<0.05, Fig 3C) and 0.51 Log10 CFU/mL

(P<0.05, Fig 3D) after incubation for 24 h and 1 wk, respectively. No concentration

difference was observed when samples were incubated at 4 °C for 2 wk (Fig 3E).

In Salmonella Enteritidis contaminated liquid egg, the concentrations of

Salmonella Enteritidis in SJ2-treated samples were significantly lower than that in control

samples by 0.93 Log10 CFU/mL (Fig 4A) and 0.62 Log10 CFU/mL (Fig 4B) after

incubation at room temperature for 24 h and 48 h, respectively. The concentrations of

Salmonella Enteritidis were not different between control and phage treated samples after

incubation at 4 °C for 24 h, 1 wk or 2 wk (Fig 4C-4E).

4.4.3 Development of Phage Resistant Salmonella

By using non-parametric analysis, it was observed that, at room temperature, the

percentage of phage resistant Salmonella Typhimurium in phage-treated liquid eggs was

numerically higher than that of untreated eggs after incubation for 24 h (untransformed

means of 14.44% vs. 0.56%, P=0.0794, Fig 5A) and significantly higher for 48 h

(untransformed means of 92.50% vs. 0.56%, P<0.05, Fig 5B). Similarly, at room

temperature, the percentage of phage resistant Salmonella Enteritidis in phage-treated

Page 106: Phage therapy applications: Efficacy and impact on the ...

92

92

liquid eggs was not significantly different from that of untreated eggs after incubation for

24 h (untransformed means of 3.75% vs. 1.11%, P=0.2036, Fig 5C) and significantly

higher for 48 h (untransformed means of 50.83% vs. 0.56%, P<0.05, Fig 5D). Such

differences were not observed between phage treated and non-treated ground meat

samples incubated at room temperature for 24 h or 48 h, in either Salmonella serovar (Fig

6A-D). Additionally, the percentages of phage resistant colonies were not different

between phage treated samples and controls at 4 °C for all incubation periods and

Salmonella seravars (data not shown).

Page 107: Phage therapy applications: Efficacy and impact on the ...

93

93

4.5 Discussion

Phage-based antibacterial methods have received much attention in recent years

due to their unique advantages as well as their ability to kill antibiotic resistant bacteria.

However, in practice, one of the distinct advantages of phages, the high target specificity,

may limit the use of phages for certain treatments (Loc-Carrillo and Abedon, 2011), such

as the applications in foods, where the exact species or strain is often not known. Several

groups have used phage cocktails (Leverentz et al., 2001; Leverentz et al., 2003; Hungaro

et al., 2013; Spricigo et al., 2013; Hong et al., 2014) and broad-spectrum phages (Carlton

et al., 2005; Guenther et al., 2009; Guenther et al., 2012) in attempt to increase the host-

spectrum. While using phage cocktails can easily expand the target spectrum, it is not

without its own set of challenges (Chan et al., 2013). As has been reported previously,

phages in the cocktail can function synergistically or antagonistically (Hall et al., 2012),

therefore, in order to optimize the effect of a phage cocktail, phages must be carefully

selected and highly characterized to avoid deleterious interactions within the cocktail

itself. In addition, most published phage genomes contain very large un-annotated

sections, which complicates the comparison between unrelated phages. For example,

73.6% of the predicted genes in phage SJ2 genome (Zhang et al., 2015) do not match any

record with known functions in NCBI database. Similar published phage genomes

contain comparative percentages of unknown and un-annotated sequences (Kropinski et

al., 2013; Hong et al., 2014; Zhang et al., 2014). Additionally, large unknown sections

may include undesirable elements such as genes involved in lysogeny or virulence. This

likelihood, while quantitatively unknown, would increase with the number of phages

included in the cocktail. Thus, it may be safer and more time-efficient to use a single,

Page 108: Phage therapy applications: Efficacy and impact on the ...

94

94

very well characterized phage if challenges associated with resistance development are

easily overcome. Finally, with available phage products, each phage in the cocktail has

historically required individual regulatory approval, leading to additionally higher

development and production costs and time (Thiel, 2004).

While the balance of achieving relative broad-spectrum and maintaining

simplicity in the phage product may be found in the use of single broad-spectrum phage,

the efficacy of such application in foods is currently understudied. While nearly 2,500

Salmonella serovars have been documented, Salmonella Typhimurium and Salmonella

Enteritidis are the predominant serovars both in the US and Europe (Guenther et al., 2012)

and were therefore selected in this study. Foods of animal origins, such as meat, poultry

and eggs, are major vehicles of Salmonella. Particularly, Salmonella Enteritidis is a

predominant serovar in eggs. Ground meat and liquid egg, due to the nature of the

products as a mixture of meat or eggs from multiple sources, may have higher risk of

Salmonella contamination. The efficacy of using single, broad-spectrum anti-Salmonella

phage was tested under both refrigerated temperature to resemble conditions during food

storage and transportation and room temperature to mimic temperature-violation

situations. As expected, reductions of Salmonella were less effective under refrigeration.

The reduction in efficacy was seen in both Salmonella serovars as well as both food

matrixes tested. Similar results were also observed in our previous research focused on

reducing E. coli O157:H7 in ground beef, where a reduction of 0.48 log CFU/ml of E.

coli O157:H7 was observed at 4 °C after 24 h, while incubation at room temperature

reduced the concentration by 1.97 log CFU/mL (Hong et al., 2014). Phages are obligate

parasites of their host bacteria; therefore, the optimal growth temperature of the host

Page 109: Phage therapy applications: Efficacy and impact on the ...

95

95

bacterium is likely also the optimal temperature for the corresponding phage. Under

lower than optimal temperature, the penetration of phage genetic materials into host cells

is inhibited (Jończyk et al., 2011) leading to the lower lytic activity of the phages. While

a previous study indicated that phages were more effective under refrigeration (Sharma et

al., 2009), it should be noted that a higher MOI (MOI=100) was used in that experiment,

where phages could lyse or at least adhere to bacteria during the first few minutes of

treatment. Lower MOIs (i.e. MOI of 10 was used in this study), however, do not favor

lysis from without. Therefore, samples infected with lower concentrations of phages may

require a more optimum temperature to result in appreciable cell lysis.

A high concentration of Salmonella inoculum (approximately 106 CFU/gram in

ground pork and 106 CFU/mL in liquid egg) was used to prevent bacteria concentrations

from falling under detection levels (103 CFU/gram in meat and 10 CFU/mL in liquid egg)

after refrigeration and phage treatment. In reality, pathogens are usually present in lower

concentrations (i.e. <104 CFU/mL, Bigwood et al., 2009) leading to the use of higher

MOI, which will likely result in greater pathogen reductions as shown in a previous study

(Abuladze et al., 2008). Although, for bacteria at extreme lower concentrations, the

reduction becomes independent of MOI and a sufficiently high concentration of phage

will results in similar reduction regardless of the resulted MOIs (Bigwood et al., 2009).

One purpose of using phage cocktail is to mitigate the development of phage

resistance, as it is much less probable that the bacterial host would develop resistance

simultaneously to several distinct phages (Hong et al., 2014). Here we aimed to

determine how often and how soon targeted bacteria might become resistant to a single

phage within a food matrix. Interestingly, our results showed that the emergence of phage

Page 110: Phage therapy applications: Efficacy and impact on the ...

96

96

resistant bacteria varied between the two different food matrixes. Significantly higher

levels of phage resistant Salmonella isolates were detected in liquid egg samples, but not

in ground pork samples, after phage treatment at room temperature for two days. Similar

results were observed for both Salmonella Typhimurium and Salmonella Enteritidis,

indicating that food matrix may influence the development of phage resistant Salmonella

isolates. A previous study (Guenther et al., 2009) pointed out that the limited diffusion of

phages in different food matrices is an important factor to considerate during the

development of phage applications in foods. Phage diffusion is expected to be most

difficult in foods, such as meat, with large and uneven surface area. Although in this

experiment ground meat samples were hand massaged after phage inoculation, the

complex matrix of meat, such as the presence of fat, could prevent the contact between

phages and bacteria. In contrary, the diffusion of phages in liquid egg, while not as freely

as in the chocolate milk or mozzarella cheese brine tested in the aforementioned study, is

better than that in ground meat samples. Therefore, bacteria and phage in liquid egg are

more accessible to each other. The increase in phage-bacteria interactions could, by

probability, increase the likelihood of selecting phage resistant bacteria. Additionally,

other bacteria presented in the foods may also influence the development of phage

resistance. The reduction of Salmonella by phage provides surrounding bacteria an

opportunity to grow, which in turn results in growth competition with phage resistant

Salmonella strains. Due to the nature of foods, ground pork likely harbors a more diverse

microbiome than liquid eggs, which are often functionally sterile. Therefore, phage

resistance strains that emerge in ground pork may be subject to stronger growth

competition. High levels (10 out of 10 tested colonies) of phage resistant isolates were

Page 111: Phage therapy applications: Efficacy and impact on the ...

97

97

also recovered from egg yolk and chocolate milk after treatment with a single Salmonella

phage for 6 days at 15 °C (Guenther et al., 2012). The same study also showed that under

a high MOI setting where great reductions of bacteria resulted, the emergence of phage

resistant isolates might not compromise the initial phage efficacy, at least as observed in

chocolate milk. While not statistically significant, numerically higher percentages of

phage resistant Salmonella Typhimurium isolates were also detected in phage treated

liquid egg samples after one day at room temperature, suggesting development of phage

resistance may be rapid in certain foods. Additionally, the percentages of phage resistant

isolates were not different between phage treated and non-treated samples under

refrigerated conditions at any time point. This is understandable as low temperature likely

inhibited the physical activity of both the host bacteria as well as the phage.

Taken together, this study demonstrated the potential of using a single, broad-

spectrum phage, SJ2, in controlling Salmonella Typhimurium and Salmonella Enteritidis

contamination in ground pork and liquid egg. In future studies, the efficiency of SJ2

should also be tested at lower bacterial conditions, which more closely resemble a

production-like setting. While the significantly higher levels of phage resistant

Salmonella isolates in phage-treated liquid egg samples at room temperature indicate that

liquid egg might not be an ideal food matrix for phage treatment, it would still be of

interest to see whether similar high percentages of phage resistance occur at high MOI

setting and quantify the potential impact of resistance on subsequent phage applications.

Page 112: Phage therapy applications: Efficacy and impact on the ...

98

98

Control Phage

5

6

7

8

9

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Control Phage

5

6

7

8

9

10

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Control Phage

3

4

5

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Control Phage

0

1

2

3

4

5

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Control Phage

0

1

2

3

4

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Figure 1: Reduction of Salmonella Typhimurium contamination in phage treated ground

pork at room temperature after 24 h (A) and 48 h (B), and at 4 °C after 24 h (C), 1 wk (D)

and 2 wk (E). Bars indicate means within treatments. * = statistical differences (P <0.05)

in mean bacterial concentrations between treatment groups.

* *

*

*

A B

C D

E

Page 113: Phage therapy applications: Efficacy and impact on the ...

99

99

Control Phage6.0

6.5

7.0

7.5

8.0

8.5

9.0S

alm

onel

la E

nter

itidi

s,

log1

0CFU

/mL

Control Phage

6

7

8

9

10

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Control Phage

4.0

4.5

5.0

5.5

6.0

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Control Phage

3.5

4.0

4.5

5.0

5.5

6.0

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Control Phage3.5

4.0

4.5

5.0

5.5

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Figure 2: Reduction of Salmonella Enteritids contamination in phage treated ground pork

at RT after 24 h (A) and 48 h (B), and at 4 °C after 24 h (C), 1 wk (D) and 2 wk (E). Bars

indicate means within treatments. * = statistical differences (P <0.05) in mean bacterial

concentrations between treatment groups.

A B

C D

E

* *

* *

Page 114: Phage therapy applications: Efficacy and impact on the ...

100

100

Control Phage

4

5

6

7

8

9

10S

alm

onel

la T

yphi

mur

ium

, lo

g10C

FU/m

L

Control Phage

8

9

10

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Control Phage

3

4

5

6

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Control Phage

3

4

5

6

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Control Phage

3

4

5

6

Sal

mon

ella

Typ

him

uriu

m,

log1

0CFU

/mL

Figure 3: Reduction of Salmonella Typhimurium contamination in phage treated liquid

egg at RT after 24 h (A) and 48 h (B), and at 4 °C after 24 h (C), 1 wk (D) and 2 wk (E).

Bars indicate means within treatments. * = statistical differences (P <0.05) in mean

bacterial concentrations between treatment groups.

* *

A B

C D

E

*

* *

Page 115: Phage therapy applications: Efficacy and impact on the ...

101

101

Control Phage

7

8

9

10

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Control Phage

7

8

9

10

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Control Phage

4.5

5.0

5.5

6.0

6.5

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Control Phage

3

4

5

6

7

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Control Phage

2.5

3.0

3.5

4.0

4.5

5.0

Sal

mon

ella

Ent

eriti

dis,

lo

g10C

FU/m

L

Figure 4: Reduction of Salmonella Enteritids contamination in phage treated liquid egg at

RT after 24 h (A) and 48 h (B), and at 4 °C after 24 h (C), 1 wk (D) and 2 wk (E). Bars

indicate means within treatments. * = statistical differences (P <0.05) in mean bacterial

concentrations between treatment groups.

* *

A B

C D

E

Page 116: Phage therapy applications: Efficacy and impact on the ...

102

102

Control Phage

0

20

40

60

80

100P

hage

res

ista

nt is

olat

es (%

)

Control Phage0

20

40

60

80

100

Pha

ge r

esis

tant

isol

ates

(%)

CONTROL PHAGE

0

20

40

60

80

100

Pha

ge r

esis

tant

isol

ates

(%)

Control Phage0

20

40

60

80

100P

hage

res

ista

nt is

olat

es (%

)

Figure 5: Percentage of phage resistant Salmonella Typhimurium isolates recovered from

phage-treated liquid eggs after incubation at room temperature for 24 h (A) and 48 h (B).

Percentage of phage resistant Salmonella Enteritidis isolates recovered from phage-

treated liquid eggs after incubation at room temperature for 24 H (C) and 48 h. (D)* =

statistical differences (P <0.05) in the percentage of phage resistant bacterial isolates

between treatment groups.

A

C

*

*

B

D

Page 117: Phage therapy applications: Efficacy and impact on the ...

103

103

Control Phage0

20

40

60

80

100

Pha

ge r

esis

tant

isol

ates

(%)

Control Phage0

20

40

60

80

100

Pha

ge r

esis

tant

isol

ates

(%)

Control Phage0

20

40

60

80

100

Pha

ge r

esis

tant

isol

ates

(%)

Control Phage0

20

40

60

80

100

Pha

ge r

esis

tant

isol

ates

(%)

Figure 6: Percentage of phage resistant Salmonella Typhimurium isolates recovered from

phage-treated ground pork after incubation at room temperature for 24 h (A) and 48 h (B).

Percentage of phage resistant Salmonella Enteritidis isolates recovered from phage-

treated ground pork after incubation at room temperature for 24 h (C) and 48 h (D)* =

statistical differences (P <0.05) in the percentage of phage resistant bacterial isolates

between treatment groups.

A B

D C

Page 118: Phage therapy applications: Efficacy and impact on the ...

104

104

List of References

Abuladze, T., M. Li, M. Y. Menetrez, T. Dean, A. Senecal, and A. Sulakvelidze. 2008.

Bacteriophages reduce experimental contamination of hard surfaces, tomato,

spinach, broccoli, and ground beef by Escherichia coli O157:H7. Appl. Environ.

Microbiol. 74:6230–6238.

Anany, H., W. Chen, R. Pelton, and M. W. Griffiths. 2011. Biocontrol of Listeria

monocytogenes and Escherichia coli O157:H7 in meat by using phages

immobilized on modified cellulose membranes. Appl. Environ. Microbiol.

77:6379–6387.

Bigwood, T., J. A. Hudson, and C. Billington. 2009. Influence of host and bacteriophage

concentrations on the inactivation of food-borne pathogenic bacteria by two

phages: RESEARCH LETTER. FEMS Microbiol. Lett. 291:59–64.

Carlton, R. M., W. H. Noordman, B. Biswas, E. D. De Meester, and M. J. Loessner. 2005.

Bacteriophage P100 for control of Listeria monocytogenes in foods: Genome

sequence, bioinformatic analyses, oral toxicity study, and application. Regul.

Toxicol. Pharmacol. 43:301–312.

Carter, C. D., A. Parks, T. Abuladze, M. Li, J. Woolston, J. Magnone, A. Senecal, A. M.

Kropinski, and A. Sulakvelidze. 2012. Bacteriophage cocktail significantly

reduces Escherichia coli O157: H7 contamination of lettuce and beef, but does

not protect against recontamination. Bacteriophage 2:178–185.

Chan, B. K., S. T. Abedon, and C. Loc-carrillo. 2013. Phage cocktails and the future of

phage therapy. Future Microbiol.:769–783.

Page 119: Phage therapy applications: Efficacy and impact on the ...

105

105

Ebner, P. D., and A. G. Mathew. 2000. Effects of antibiotic regimens on the fecal

shedding patterns of pigs infected with Salmonella Typhimurium. J. Food Prot.

63:709–714.

Guenther, S., O. Herzig, L. Fieseler, J. Klumpp, and M. J. Loessner. 2012. Biocontrol of

Salmonella Typhimurium in RTE foods with the virulent bacteriophage FO1-E2.

Int. J. Food Microbiol. 154:66–72.

Guenther, S., D. Huwyler, S. Richard, and M. J. Loessner. 2009. Virulent bacteriophage

for efficient biocontrol of Listeria monocytogenes in ready-to-eat foods. Appl.

Environ. Microbiol. 75:93–100.

Hall, A. R., D. De Vos, V. P. Friman, J. P. Pirnay, and A. Buckling. 2012. Effects of

sequential and simultaneous applications of bacteriophages on populations of

Pseudomonas aeruginosa in vitro and in wax moth larvae. Appl. Environ.

Microbiol. 78:5646–5652.

Hong, Y., Y. Pan, and P. D. Ebner. 2014. Meat science and muscle biology symposium:

Development of Bacteriophage treatments to reduce Escherichia coli O157:H7

contamination of beef products and produce. J. Anim. Sci. 92:1366–1377.

Hong, Y., Y. Pan, N. J. Harman, and P. D. Ebner. 2014. Complete Genome Sequences of

Two Escherichia coli O157:H7 Phages Effective in Limiting Contamination of

Food Products. Genome Announc.2(5): e00519-14.

Hungaro, H. M., R. C. S. Mendonça, D. M. Gouvêa, M. C. D. Vanetti, and C. L. de O.

Pinto. 2013. Use of bacteriophages to reduce Salmonella in chicken skin in

comparison with chemical agents. Food Res. Int. 52:75–81.

Page 120: Phage therapy applications: Efficacy and impact on the ...

106

106

Jończyk, E., M. Kłak, R. Międzybrodzki, and A. Górski. 2011. The influence of external

factors on bacteriophages--review. Folia Microbiol. (Praha). 56:191–200.

Kittler, S., S. Fischer, A. Abdulmawjood, G. Glünder, and G. Kleina. 2013. Effect of

bacteriophage application on Campylobacter jejuni loads in commercial broiler

flocks. Appl. Environ. Microbiol. 79:7525–7533.

Kropinski, A. M., T. Waddell, J. Meng, K. Franklin, H.-W. Ackermann, R. Ahmed, A.

Mazzocco, J. Yates, E. J. Lingohr, and R. P. Johnson. 2013. The host-range,

genomics and proteomics of Escherichia coli O157:H7 bacteriophage rV5. Virol.

J. 10:76.

Leverentz, B., W. S. Conway, Z. Alavidze, W. J. Janisiewicz, Y. Fuchs, M. J. Camp, E.

Chighladze, and A. Sulakvelidze. 2001. Examination of bacteriophage as a

biocontrol method for Salmonella on fresh-cut fruit: a model study. J. Food Prot.

64:1116–1121.

Leverentz, B., W. S. Conway, M. J. Camp, W. J. Janisiewicz, T. Abuladze, M. Yang, R.

Saftner, and A. Sulakvelidze. 2003. Biocontrol of Listeria monocytogenes on

fresh-cut produce by treatment with lytic bacteriophages and a bacteriocin. Appl.

Environ. Microbiol. 69:4519–4526.

Loc-Carrillo, C., and S. T. Abedon. 2011. Pros and cons of phage therapy. Bacteriophage

1:111–114.

Scallan, E., R. M. Hoekstra, F. J. Angulo, R. V Tauxe, M.-A. Widdowson, S. L. Roy, J. L.

Jones, and P. M. Griffin. 2011. Foodborne illness acquired in the United States--

major pathogens. Emerg. Infect. Dis. 17:7–15.

Page 121: Phage therapy applications: Efficacy and impact on the ...

107

107

Sharma, M., J. R. Patel, W. S. Conway, S. Ferguson, and A. Sulakvelidze. 2009.

Effectiveness of bacteriophages in reducing Escherichia coli O157:H7 on fresh-

cut cantaloupes and lettucet. J. Food Prot. 72:1481–1485.

Spricigo, D. A., C. Bardina, P. Cortés, and M. Llagostera. 2013. Use of a bacteriophage

cocktail to control Salmonella in food and the food industry. Int. J. Food

Microbiol. 165:169–174.

Thiel, K. 2004. Old dogma, new tricks--21st Century phage therapy. Nat. Biotechnol.

22:31–36.

Wall, S. K., J. Zhang, M. H. Rostagno, and P. D. Ebner. 2010. Phage therapy to reduce

preprocessing Salmonella infections in market-weight swine. Appl. Environ.

Microbiol. 76:48–53.

Zhang, J., Y. Hong, N. J. Harman, A. Das, and P. D. Ebner. 2014. Genome sequence of a

Salmonella phage used to control salmonella transmission in Swine. Genome

Announc. 2.

Zhang J., Y. Hong, M. Fealey, A. Singh, K. Walton, C. Martin, N. J . Harman, J. Mahlie,

and P. D. Ebner. 2015. Physiological and molecular characterization of

Salmonella bacteriophages previously used in phage therapy. J. Food Prot. (in

press)

Page 122: Phage therapy applications: Efficacy and impact on the ...

108

108

CHAPTER 5. THE IMPACT OF ORAL ADMINISTERED PHAGES ON HOST: ACUTE IMMUNE RESPONSES AND GUT MICROBIOME

5.1 Abstract

Extensive studies have shown the efficacy of phage therapy in reducing the

carriage of foodborne pathogens in food animals. Far fewer studies have focused on the

safety of phage therapy, especially in terms of how high concentrations of phage

challenges may elicit animal acute response and affect the gut microbiome. Here we

administered E. coli O157:H7 phages to mice (n = 12) at either low (105 PFU) or high

(107 PFU) concentration. At 6 h and 24 h post treatment, blood and fecal samples were

collected to measure the levels of 12 pro-inflammatory cytokines (IL1A, IL1B, IL2, IL4,

IL6, IL10, IL12, IL17A, IFNγ, TNFα, G-CSF, and GM-CSF) as well as intestinal IgA

and IgG. To test the effect of phages on animal gut microbiome, three-week-old pigs

were administered a standard diet containing ASP 250 (chlortetracycline, sulfamethazine,

and penicillin; n = 5), a standard diet with daily inoculation of 5×1010 PFU of phages via

oral gavage (N=6), and a standard diet without antibiotics or phage (control; n = 6). After

two weeks, pigs were euthanized, microbial DNA of ileal, cecal, and fecal contents was

extracted, and the microbiome composition was analyzed using 16S rRNA. Our results

showed no differences in the levels of intestinal IgA and IgG among control, low phage,

and high phage groups at both 6 h and 24 h post treatment. However, phage treatment did

elicit some cytokine response, with the levels of G-CSF at 6 h post-low phage treatment,

Page 123: Phage therapy applications: Efficacy and impact on the ...

109

109

levels of G-CSF, IL1A, IL1B, IL10, and IL17A at 6 h post-high phage treatment, levels

of IL17A, IFNα, G-CSF, and GM-CSF at 24 h post-low phage treatment, and levels of

IL1A, IL2, IL10 and G-CSF at 24 h post-high phage treatment being significantly higher

(P < 0.05) than corresponding cytokine levels in control group. All cytokine

concentrations, however, were within normal ranges for mice at all sampling times across

all treatments. In pig experiments, no performance differences (ADG, WG, F:G) were

observed among control, antibiotic, and phage groups. However, antibiotic treated pigs

had numerically higher WG than pigs in the other treatments in the second week (4.5 kg

in antibiotic group compared to 3.67 kg in control and 3.6 kg phage-treated group, P=

0.0834). The differences in both phylogeny and relative taxon abundance were accounted

for microbiome composition comparison. Compared to control microbiome, the antibiotic

treatment significantly altered the microbiome composition at ileum (P<0.05), with the

Bacilli class being mostly affected (22% in antibiotic group vs. 76% in control, FDR =

0.0572). No significant differences were observed in cecal and fecal microbiome between

control and antibiotic-treated pigs. Compared to control, phage treatment did not alter the

composition of the gut microbiome at any of the tested intestinal compartments.

Significant abundance differences were observed at the OTU level, with OTUs belonging

to genera such as Lactobacillus and Streptococcus being over or underrepresented in

either antibiotic or phage treated groups compared to controls. Determining whether these

changes are deleterious to host, however, requires further studies. Taken together, our

study indicated that phage administrations, even at high concentrations, would not elicit

adverse acute immune response or greatly alter gut microbiome.

Page 124: Phage therapy applications: Efficacy and impact on the ...

110

110

5.2 Introduction

Recently, there has been regained interest in the development of phage-based

antibacterials. Extensive studies have focused on measuring the efficacy of phage therapy

in preventing or reducing undesired bacteria in food animals, food processing facilities,

medical environments, and even humans. In agriculture, phage therapy has showed

effective in reducing various foodborne pathogens, such as Salmonella in swine (Wall et

al., 2010) and poultry (Atterbury et al., 2007), Campylobacter in poultry (Loc Carrillo et

al., 2005), and E. coli O157:H7 in ruminants (Rozema et al., 2009).

Compared to many other antimicrobial strategies, especially antibiotic treatments,

phage therapy has the unique advantage of being highly specific. It is common for phages

to only affect only specific bacterial serovars or even strains within a serovar. (Loc-

Carrillo and Abedon, 2011). As such, it is generally believed that, phages, even when

applied in high concentrations via oral delivery, have limited collateral effect on

untargeted bacteria and, as a result, are less likely to cause the microbial imbalances that

are commonly observed as a side effects in antibiotic treatments. However, few studies,

especially those using culture-independent techniques, have evaluated the impact of

phages on gut microbiome.

It is well established that the gut microbiome plays essential roles in animal

growth and health. With the recent advance in 16S rRNA analysis and metagenomic

analysis, it is clear that altered gut microbiome can significantly impact different

metabolic and immunologic functions. Therefore, it is essential to more comprehensively

evaluate the impact of phage therapy on host gut microbiome. Additionally, the extent to

which phages to elicit host acute immune response remains understudied as well. While

Page 125: Phage therapy applications: Efficacy and impact on the ...

111

111

animals are constantly exposed to phages in natural environment, much higher

concentrations of phages are commonly used in phage therapy. Thus, it is equally

important to evaluate whether phage challenge at high concentrations induces any

adverse immune response to the animal host. In this study, the impact of phages on host

animals was carefully characterized in terms of both host acute immune response and gut

microbiome.

Page 126: Phage therapy applications: Efficacy and impact on the ...

112

112

5.3 Materials And Methods

5.3.1 Bacteriophages

An E. coli O157:H7 phage cocktail containing three unrelated phages was used to

measure the acute immune response to phage treatment in a mouse model. A previous

study demonstrated that the same phage cocktail was effective in reducing E. coli

O157:H7 contamination in ground beef and spinach (Hong et al., 2014). Each phage was

propagated separately and combined in equal amount. The titer of the phage mixture was

determined by plaque assay and then diluted to solutions of 108 PFU/mL (high phage

group) and 106 PFU/mL (low phage group).

A ten-Salmonella phage cocktail was used to evaluate the impact of orally

administrated phages on animal gut microbiome in a pig model. The same phage cocktail

was effective in controlling lairage-associated Salmonella infection in pigs (Wall et al.,

2010; Saez et al., 2011). Similarly, each phage was propagated separately and combined

in equal amount. The final concentration of mixed phage cocktail was 1.07 x 1010

PFU/mL.

5.3.2 Mice Immune Responses to Phage Treatment

All animal experiments were approved by the Purdue University Animal Care and

Use Committee (PACUC). Thirty-six BALB/c mice (18 males and 18 females; 5-week-

old) were treated with 2 g/L of streptomycin (Sigma-Aldrich, MO) in drinking water for

3 d. Mice were then divided into 3 groups (6 male and 6 female mice per group) and

subjected to feed withdrawal for 24 h before phage treatment. Mice in the high phage

group received 100 ul of E. coli O157:H7-phage cocktail at a concentration of 108

Page 127: Phage therapy applications: Efficacy and impact on the ...

113

113

PFU/mL; mice in the low phage group received 100 ul of same phage cocktail at a

concentration of 106 PFU/mL; and mice in control group received 100 ul of 1x PBS. All

treatments were delivered via oral gavage. At 6 h and 24 h post phage treatment, feces

and blood (submandibular) were collected from each mouse. Blood samples were

centrifuged at 1,500 x g to separate plasma from blood cells. Plasma samples were

immediately used to measure the concentrations of 12 pro-inflammatory cytokines (IL1A,

IL1B, IL2, IL4, IL6, IL10, IL12, IL17A, IFNγ, TNFα, G-CSF, and GM-CSF) using a

mouse inflammatory cytokines multi-analyte ELISArray kit (SABiosciences, Valencia,

CA). Fecal samples were used to measure the levels of intestinal IgA and IgG using a

mouse IgA, IgG ELISA Quantitation set (Bethyl Laboratories, Montgomery, TX)

according to the manufacturer’s instructions.

5.3.3 Impact of Phage Therapy on Pig Gut Microbiome

Seventeen, three-week-old pigs were obtained from a high health commercial

farm. Sows were not administered antibiotics post-farrowing and pigs were not exposed

to antibiotics in their nursery diet or creep feed before arrival. Upon arrival, pigs were

housed in the same pen for one week as an adjustment period before they were assigned

to one of the three treatments: control group (n = 6) where pigs received only basal diet,

phage group (n = 6) where pigs received basal diet plus daily inoculation of 5 ml

Salmonella-phage cocktail at a concentration of 1.07 x 1010 PFU/mL via oral gavage, and

antibiotic group (n = 5) where pigs received basal diet supplemented with an antibiotic

premix ASP 250 (100 g chlortetracycline, 100 g sulfamethazine, and 50 g penicillin per

ton of feed, Zoetis, Florham Park, NJ). Pigs were housed individually with each pen

Page 128: Phage therapy applications: Efficacy and impact on the ...

114

114

equipped with separate feed and water systems. The three treatment groups were

maintained at three separated regions of the animal house to prevent cross-contamination.

All pigs were weighted weekly and feed intake and health status were recorded daily.

After two weeks of treatment, all pigs were euthanized with captive bolt gun and ileal

contents, cecal contents and fecal samples were collected.

Microbial DNA of each intestinal sample was extracted using a FastDNATM spin

kit for soil (MP Biologicals, Santa Ana, CA) following the manufacturer’s protocol. For

samples with high liquid content, liquid was removed by centrifugation at 10,000 x g for

10 min before the suggested amount of sample was measured for DNA extraction. DNA

samples were quantified using QuantiFluor® dsDNA system (Promega, Madison, WN),

assessed for DNA integrity by gel electrophoresis, and stored at -20 °C before use.

The construction of the 16S rRNA library was carried out in two steps. First, the

V3 to V4 region of bacterial 16S rRNA gene was amplified using universal primers 338F

(5’-ACTCCTACGGGAGGCAGC) and 806R (5’-GGACTACHVGGGTWTCTAAT)

(Fadrosh et al., 2014) containing Illumina adapter sequences and a cycling program of: 1)

one cycle of 94 °C for 5 min; 2) 25 cycles of 94 °C for 30s, 60 °C for 30 s and 72 °C for

30 s; and 3) a final cycle of 72 °C for 5 min. Each PCR included 25 ul KAPA HiFi

hotstar readymix, 2X (Kapa Biosystems, Wilmington, MA), 10 uM of each primer, 1 ng

of template DNA, 0.5 mg/ml BSA (bovine serum albumin, Sigma-Aldrich, St. Louis,

MO), and sterile ddH2O to a final volume of 50 ul. The resulting amplicon was purified

using the Agencourt AMPure XP PCR purification system (Beckman Coulter, Pasadena,

CA). Illumina 8x12 TruSeq dual index sequencing primers (Illumina, San Diego, CA)

were used to barcode individual samples in second PCR using a cycling program of:

Page 129: Phage therapy applications: Efficacy and impact on the ...

115

115

1) one cycle of 94 °C for 3 min; 2) eight cycles of 94 °C for 30 s, 64 °C for 30 s and

72 °C for 30 s; and 3) a final cycle of 72 °C for 5 min. Each PCR reaction included 25 ul

KAPA HiFi hotstar readymix, 2X (Kapa Biosystems, Wilmington, MA), 10 uM of each

primer, 5 ul of purified amplicon of first PCR, and sterile ddH2O to a final volume of 50

ul. The resulting amplicon was purified by gel electrophoresis and gel extraction using

QIAquick Gel Extraction kit (Qiagen, Venlo, Netherlands). The final amplicon of each

sample was quantified using QuantiFluor® dsDNA system (Promega, Madison, WN) and

mixed in equal amounts and sequenced as a 16S rRNA library with the Illumina Miseq

platform (2 x 300 bp).

Sequence reads were sorted by barcodes, quality filtered, and adapter-trimmed.

The reads sequenced from 5’-end and 3’-end were merged based on the overlapping

sequences and a total of 37,891 to 547,061 merged reads were obtained for each samples.

Due to the difference in total number of reads among samples, relative abundance was

used. QIIME (Version 1.8.0, Caporaso et al., 2010) was used to analyze and compare the

microbiome between samples. All merged reads underwent operational taxonomic unit

(OTU) picking, taxonomic assignment and phylogenetic tree construction using Green

Genes (97, 13_5 version) as a reference database. Weighted UniFrac distance matrices,

accounting for differences in both phylogeny and relative taxon abundance, were

generated and compared pairwise between treatments within each intestinal compartment

using ANOSIM. The relative abundance difference at taxonomic and OTU levels was

also compared pairwise between treatments within each intestinal compartments using

phyloseq software package (McMurdie and Holmes, 2013) and a FDR (false discovery

rate) of 0.05 was used to determine statistical significance.

Page 130: Phage therapy applications: Efficacy and impact on the ...

116

116

5.3.4 Statistical Analysis

For mouse experiments, IgG and IgA concentrations were compared among

treatment groups using a one-way ANOVA and also compared between male and female

mice within treatment groups using a two-sample t-test of the SAS TTEST procedure.

Percentages of mice with elevated immune responses were compared among control, low

phage, and high phage groups by a Chi-square test using the SAS FREQ procedure. For

pig experiments, the growth performance data were compared among three groups using

a one-way ANOVA.

Page 131: Phage therapy applications: Efficacy and impact on the ...

117

117

5.4 Results

5.4.1 Immune Responses of Mice to Phage Treatment

All mice remained healthy throughout the experiment. Intestinal IgG and IgA

were compared among control, low phage, and high phage groups, and also compared

between male and female mice within treatment groups. Bacteriophages were detected in

fecal samples at 6 h but not 24 h post phage treatment (data not shown). At 6 h post

phage treatment, total IgA and IgG did not differ among the groups (Fig. 1A and 2A).

Similar results were observed at 24 h post phage administration (Fig. 1B and 2B).

However, effects of gender in the immune response were identified in the high phage

treated group at 6 h post phage treatment. Higher levels of IgA and IgG in female mice

were detected than in the male mice (Fig. 1C and 2C). The effect was not observed at 24

h post treatment in high phage group (Fig. 1D and 2D). Intestinal IgA was also higher in

control female mice at 24 h post treatment than in control male mice (Fig. 1D).

Among the 12 cytokines tested at 6 h and 24 h post treatment, most cytokines did

not show significant differences between phage treated mice and controls. However, at 6

h post treatment, G-CSF in low phage treated mice and G-CSF, IL1A, IL1B, IL10 and

IL17A in high phage treated mice were significantly (P < 0.05) higher than those in

control mice. At 24 h post treatment, IL17A, IFNα, G-CSF and GM-CSF in low phage

treated mice and IL1A, IL2, IL10 and G- CSF in high phage treated mice were higher

(P<0.05) than those in control mice. All cytokine levels, however, were in the normal

range for mice in all treatment groups at all sampling points.

Page 132: Phage therapy applications: Efficacy and impact on the ...

118

118

5.4.2 Pig Growth Performance

All pigs, except one in the phage group, remained healthy throughout the

experiment. The pig showed mild diarrhea before the experiment and sporadic diarrhea

throughout the experiment. Therefore, both the growth performance data and the 16S

rRNA sequences generated from that pig were not used.

Pig growth performance, in terms of average daily feed intake (ADF), weekly

weight gain (WG) and feed/gain ratio (F/G), was not different (P > 0.05) among control,

phage, and antibiotic groups (Table 1). However, pigs in the antibiotic group had a

tendency to have higher WG than pigs in the other two groups at second week of

experiment (4.5 kg in antibiotic group compared to 3.67 kg in control and 3.6 kg phage

group, P= 0.0834).

5.4.3 Gut Microbiome Analysis and Comparison

It was consistent across the animals, regardless of treatments, that Firmicutes and

Proteobacteria were the most abundant phyla in the ileal microbiome of pigs, accounting

for approximately 85% and 14% of reads, respectively. Firmicutes, Bacteriodetes, and

Proteobacteria were the top three abundant phyla in the cecal content microbiome,

accounting for 56%, 31%, and 11% of reads, respectively, and in the fecal microbiome as

well, counting for 58%, 26%, and 11% of reads, respectively (Fig. 3).

Microbial communities were compared pairwise between treatments within each

intestinal compartment. Weighted UniFrac distance matrices, which account for

differences in both phylogeny and relative taxon abundance, were used as inputs and

ANOSIM were used as the statistical method. The ileal microbiome of antibiotic treated

Page 133: Phage therapy applications: Efficacy and impact on the ...

119

119

pigs was statistically different from that of control pigs (P = 0.049); however, differences

were not observed in either the cecal (P = 0.123) or fecal (P = 0.348) microbiome

between the two treatments. No differences were detected in the ileal (P = 0.12), cecal (P

= 0.331) or fecal (P = 0.732) microbiome between phage treated pigs and controls. No

relative abundance differences were observed between treatments at taxonomic levels.

There were numerically less Bacilli class in antibiotic treated ileal microbiome compared

to the control ileal microbiome (22% vs. 76%, FDR = 0.0572), and numerically less

Actinobacteria (5.67 x 10-4% vs. 4.4 x 10-3%, FDR = 0.0582) and Bacilli (4% vs. 18%,

FDR = 0.0582) classes in the antibiotic treated cecal microbiome than in the phage

treated cecal microbiome. Significant relative abundance differences were only observed

at the OTU level, with OTUs associated with various bacteria being over or

underrepresented in either antibiotic (Fig. 5) or phage treated group (Fig. 4) compared to

control at all three intestinal compartments.

Page 134: Phage therapy applications: Efficacy and impact on the ...

120

120

5.5 Discussion

Phage therapy has shown great promise in preventing or reducing the carriage of

foodborne pathogens in agriculture animals. To better assess the safety of phage therapy

and understand the outcome of oral phage treatments at high concentrations to applied

animals, this study evaluated the impact of phages on host acute immune responses and

the gut microbiome.

In our experiments, we observed that orally administrated phages did elicit some

cytokine responses in mice, but the cytokine levels were in the normal ranges of healthy

mice (Stenina et al., 2012). Additionally, we found that there were no differences in

intestinal IgA and IgG among treatment groups after 24 h post treatment. This may be

due to the clearance of phage from the intestine, as phages were not recovered after 24 h

post treatment. It should also be noted that 24 h may not be enough time to detect an

appreciable humoral immune response. In order to more accurately describe the immune

response pattern of mice, multiple sampling points over longer sampling periods may be

needed. Interestingly, at 6 h post treatment, female mice had higher levels of intestinal

IgA and IgG than male mice. This phenomenon may be due to stronger and longer lasting

immune response in female mice (Ansar Ahmed et al., 1985). In accord with our results,

a recent phage safety test performed on 15 healthy human adults demonstrated that oral

phage administration did not elicit appreciable levels of phage-specific IgG, IgM, and

IgA (Bruttin and Brüssow, 2005). While these results provide some evidence that phages

by themselves are not likely to elicit adverse acute immune response, extra consideration

must be taken when phage-targeting bacteria are also presented, in which case, the lysis

of bacterial cells could lead to a sudden increase of endotoxins in the host and

Page 135: Phage therapy applications: Efficacy and impact on the ...

121

121

consequently lead to extensive acute immune response, or even death in animals

(Boratynski et al., 2004).

Our study also investigated the impact of phage treatment on pig gut microbiome.

One pig in control group and one in phage group failed to yield ileal samples due to lack

of ileal content during collection. Additionally, sequence reads of all three intestinal

samples collected from the aforementioned diarrhea pig in phage group and those of the

ileal sample collected from a pig in antibiotic group were not included in analysis.

Diarrhea was considered to have an effect on gut microbiome. With a rapid passage of

lumen content through the intestine, the diarrhea cecal and fecal microbiome resembled

more closely to ileal microbiome composition than the cecal or fecal microbiome

compositions of healthy pigs (data not shown). The ileal microbiome of antibiotic group

was removed due to its abnormal high percentage of Proteobacteria (98.52%).

In general, the gut microbiome, regardless of treatments, resembled the typical

swine microbiome shown previously. For example, consistent with previous fecal

microbiome studies (Lamendella et al., 2011; Looft et al., 2012; Looft et al., 2014), the

fecal microbiome of this study was dominated by bacteria belong to Firmicutes,

Batereroidetes, and Proteobacteria phyla, with Prevotella being the most abundant genus

in Bacteroidetes phylum. Meanwhile, the cecal microbiome had similar composition as

the fecal microbiome, while the ileal microbiome was almost exclusively dominated by

Firmicutes and Proteobacteria. These results are consistent with another study that

characterized the microbiome at different intestinal compartments in swine (Looft et al.,

2014).

Page 136: Phage therapy applications: Efficacy and impact on the ...

122

122

Our results showed that phage treatment did not alter the overall composition of

the gut microbiome at any tested gut compartments (ileum, cecum, colon). Similar results

were also reported in a recent human study, where the oral administration of an E. coli

phage cocktail in healthy adults at doses as high as 3 x 109 PFU did not result in

consistent changes in the fecal microbiome when compared to adults receiving a placebo

(Sarker et al., 2012). The conservation of gut microbiome under phage challenge

provided experimental evidence to the previous assumption that phages are highly host

specific and have limited affect on untargeted gut microbiome. It is notable that both

studies were conducted under conditions where animals or humans were not infected with

phage-targeting bacteria. This reflects a common phage application condition in practice,

especially in agricultural uses, where phages are mostly designed for preventative

interventions. On the other hand, a recent study evaluated the impact of phages on the

mouse gut microbiome during infection where mice were colonized with an

extraintestinal, pathogenic E. coli. Treatment with a phage cocktail effectively reduced

the amount of challenge E. coli in the intestine without significantly affecting the

surrounding microbiome at family levels using 16S rRNA anaylsis (Sordi et al., 2015,

Poster communication, 21st Evergreen International Phage Meeting). Unlike the

extraintestinal E. coli in the aforementioned study, many foodborne pathogens are

commensal bacteria in agriculture animals, possibly having an established niche in gut

microbiome. In addition, for better application flexibility, phage cocktails designed for

agriculture uses probably have broad host spectrum. Therefore, there remain some

unanswered questions regarding the impact of phages on the gut microbiome of infected

animals under agriculture settings.

Page 137: Phage therapy applications: Efficacy and impact on the ...

123

123

Compared to the control, no consistent relative abundance changes were observed

in the phage treated microbiome at any tested intestinal compartments. Significant

abundance increase or decrease in the phage treated microbiome compared to the control

microbiome was only observed at OTU level at all intestinal compartments. Deeper

sequencing and improvement in 16S rRNA annotation in reference databases could

enhance the OTU taxonomic assignment and therefore possibly provide a better idea of

whether abundance changes of certain OTUs reflect abundance changes in certain

bacterial genera or species. Likewise, further experiments are required to determine

whether such changes are deleterious to animals.

Conversely, as was seen in related studies (Collier et al., 2003; Rettedal et al.,

2009; Looft et al., 2012), feeding the antibiotic growth promoter (ASP250) significantly

altered the gut microbiome in pigs with the biggest impact in the ileum. The alteration in

the antibiotic treated ileal microbiome was mainly driven by the relative abundance

decrease in Bacilli class. Lactobacillus was one of the most affected genera in Bacilli

class, with 21 OTUs assigned to Lactobacillus showing significant abundance decrease in

the antibiotic group. This was consistent with previous findings that feeding antibiotic

growth promoters significantly affected Lactobacilli population (Lin, 2011). However,

both abundance increase and decrease have been reported in Lactorbacillus species in

responding to different antibiotic treatments (Rettedal et al., 2009)(Lin, 2011), which

partly explains the fact that significant abundance changes were only observed at the

OTU level and not the genus level. Additionally, previous studies showed significant

abundance decreases in Streptococcus (Looft et al., 2014) and Turicibacter (Rettedal et

al., 2009) of the antibiotic-treated pig ileal microbiome. Significant abundance decreases

Page 138: Phage therapy applications: Efficacy and impact on the ...

124

124

in OTUs associated with each genus were observed in this study. It was noticed that one

antibiotic treated ileal microbiome, compared to the rest antibiotic treated ileal

microbiome, deviated less from the control ileal microbiome. Interestingly, growth

performance analysis indicated that the corresponding animal had significantly lower

feed intake than the rest of animals in antibiotic group, which explains less antibiotic

effect on ileal microbiome. No antibiotic-induced alteration was observed in either cecum

or feces. Besides the fact that ileum was the first site exposed antibiotics, the much less

complex composition of ileal microbiome, compared to that of cecal and fecal

microbiome, likely rendered the ileal microbiome more susceptible to alterations.

The antibiotic-induced alteration of gut microbiome is considered one of the

fundamentals that contribute to antibiotic growth promotion effect. One possible

explanation is that feeding antibiotics could reduce total bacterial load, and, consequently,

reduce microbial nutrient utilization as well as the energy expended by the immune

system in maintaining microbiome homeostasis. In our experiment, antibiotic treated pigs

had numerically higher (P = 0.0834) weight gain than control and phage groups at second

week. The lack of significant growth promotion could be due to a short experiment

period and the fact that pigs in this study were housed in a very clean and bio-controlled

environment, which has previously been shown to reduce growth promoting effects of

antibiotics (Cromwell, 2002).

Taken together, our studies evaluated the potential of oral administrated phages,

at high concentrations, to elicit host acute immune responses and affect microbiome at

different intestinal compartments. While phage treatment did elicit some cytokine

responses, no adverse acute immune response was observed in mice. Meanwhile,

Page 139: Phage therapy applications: Efficacy and impact on the ...

125

125

compared to untreated pigs, no significant composition changes or taxonomic abundance

changes were observed after phage treatment; in contrast, antibiotic treatment resulted in

significant composition changes in the ileal microbiome. While significant abundance

changes were observed at some OTUs, further studies will be needed to determine

whether such changes are deleterious to animal host. Additionally, metagenomic analysis

on the gut microbial DNA samples may further reveal whether phage treatment is

associated with changes in certain functional genes and is considered for future studies.

Page 140: Phage therapy applications: Efficacy and impact on the ...

126

126

5.6 Note

Jiayi Zhang contributed to the study of evaluating the impact of oral administered

phages on host acute immune responses and the results were also included in his Ph.D.

dissertation.

Page 141: Phage therapy applications: Efficacy and impact on the ...

127

127

Contro

l

Low Phag

e

High Phag

e1.0

1.5

2.0

2.5

Treatment Groups

log

IgA

ng/

ml

Contro

l

Low Phag

e

High Phag

e1.4

1.6

1.8

2.0

2.2

Treatment Groups

log

IgA

ng/

ml

Contro

l F

Contro

l M

low phag

e F

low phag

e M

high phag

e F

high phag

e M

1.0

1.5

2.0

2.5 *

Treatment Groups

log

IgA

ng/

ml

Contro

l F

Contro

l M

Low Phag

e F

Low Phag

e M

High Phag

e F

High Phag

e M1.4

1.6

1.8

2.0

2.2

Treatment Groups

log

IgA

ng/

ml

*

Figure 1. Intestinal IgA concentrations of all mice and mice of different sex, respectively,

at 6 h (A and C) and 24 h (B and D) after phage administration. * =significant difference

(P<0.05).

A

C

B

D

*

Page 142: Phage therapy applications: Efficacy and impact on the ...

128

128

Contro

l

Low phag

e

High phag

e0.4

0.5

0.6

0.7

0.8

0.9

Treatment Groups

log

IgG

ng/

ml

Contro

l

Low phag

e

High phag

e0.0

0.5

1.0

1.5

Treatment Groups

log

IgG

ng/

ml

Contro

l F

Contro

l M

Low phag

e F

Low phag

e M

High phag

e F

High phag

e M0.4

0.5

0.6

0.7

0.8

0.9

Treatment Groups

log

IgG

ng/

ml

Contro

l F

Contro

l M

Low phag

e F

Low phag

e M

High phag

e F

High phag

e M0.0

0.5

1.0

1.5

Treatment Groups

log

IgG

ng/

ml

Figure 2. Intestinal IgG concentrations of all mice and mice of different sex, respectively,

at 6 h (A and C) and 24 h (B and D) after phage administration. *=significant difference

(P<0.05).

*

C D

B A

Page 143: Phage therapy applications: Efficacy and impact on the ...

129

Figure 3. The composition of ileal, cecal and fecal microbiome of pigs at order level. Treatments within each intestinal

compartment were arranged as, from left to right, control, phage treatment, and antibiotic treatment.

129

Page 144: Phage therapy applications: Efficacy and impact on the ...

130

130

B

A

Page 145: Phage therapy applications: Efficacy and impact on the ...

131

131

C

Figure 4. OTUs of significant abundance changes in the phage treated ileal (A), cecal (B)

and fecal (C) microbiome compared to that in the corresponding control microbiome.

Page 146: Phage therapy applications: Efficacy and impact on the ...

132

132

A

B

Page 147: Phage therapy applications: Efficacy and impact on the ...

133

133

C

Figure 5. OTUs of significant abundance changes in the antibiotic treated ileal (A), cecal

(B) and fecal (C) microbiome compared to that in the corresponding control microbiome.

Page 148: Phage therapy applications: Efficacy and impact on the ...

134

134

Table 1: The growth performance of pigs, including average daily feed intake (ADF),

weekly weight gain (WG), and feed/gain ration (F/G) during the two week of experiment.

Control Phage Antibiotics P-value

W1 ADF 0.68 0.66 0.67 0.9901

WG 3.33 3.70 4.20 0.2784

F/G 1.44 1.27 1.13 0.2132

W2 ADF 0.77 0.84 0.98 0.4163

WG 3.67 3.60 4.50 0.0834

F/G 1.53 1.65 1.52 0.7222

Page 149: Phage therapy applications: Efficacy and impact on the ...

135

135

List of References

Ansar Ahmed, S., W. J. Penhale, and N. Talal. 1985. Sex hormones, immune responses,

and autoimmune diseases. Mechanisms of sex hormone action. Am. J. Pathol.

121:531–51.

Atterbury, R. J., M. A. P. Van Bergen, F. Ortiz, M. A. Lovell, J. A. Harris, A. De Boer, J.

A. Wagenaar, V. M. Allen, and P. A. Barrow. 2007. Bacteriophage therapy to

reduce Salmonella colonization of broiler chickens. Appl. Environ. Microbiol.

73:4543–4549.

Boratynski, J., D. Syper, B. Weber-Dabrowska, M. Lusiak-Szelachowska, G. Pozniak,

and A. Gorski. 2004. Preparation of endotoxin-free bacteriophages. Cell Mol Biol

Lett 9:253–259.

Bruttin, A., and H. Brüssow. 2005. Human volunteers receiving Escherichia coli phage

T4 orally: a safety test of phage therapy. Antimicrob. Agents Chemother.

49:2874–8.

Caporaso, J. G., J. Kuczynski, J. Stombaugh, K. Bittinger, F. D. Bushman, E. K. Costello,

N. Fierer, A. G. Peña, J. K. Goodrich, J. I. Gordon, G. A. Huttley, S. T. Kelley, D.

Knights, J. E. Koenig, R. E. Ley, C. A. Lozupone, D. McDonald, B. D. Muegge,

M. Pirrung, J. Reeder, J. R. Sevinsky, P. J. Turnbaugh, W. A. Walters, J.

Widmann, T. Yatsunenko, J. Zaneveld, and R. Knight. 2010. QIIME allows

analysis of high-throughput community sequencing data. Nat. Methods 7:335–336

Page 150: Phage therapy applications: Efficacy and impact on the ...

136

136

Collier, C. T., D. M. Albin, J. E. Wubben, V. M. Gabert, D. Bane, D. B. Anderson, H. R.

Gaskins, and B. Deplancke. 2003. Molecular ecological analysis of porcine ileal

microbiota responses to antimicrobial growth promoters. J Anim Sci. 81: 3035-

45.

Cromwell, G. L. 2002. Why and how antibiotics are used in swine production. Anim.

Biotechnol. 13:7–27.

Fadrosh, D. W., B. Ma, P. Gajer, N. Sengamalay, S. Ott, R. M. Brotman, and J. Ravel.

2014. An improved dual-indexing approach for multiplexed 16S rRNA gene

sequencing on the Illumina MiSeq platform. Microbiome 2:6.

Hong, Y., Y. Pan, and P. D. Ebner. 2014. Meat science and muscle biology symposium:

Development of Bacteriophage treatments to reduce Escherichia coli O157:H7

contamination of beef products and produce. J. Anim. Sci. 92:1366–1377.

Lamendella, R., J. W. S. Domingo, S. Ghosh, J. Martinson, and D. B. Oerther. 2011.

Comparative fecal metagenomics unveils unique functional capacity of the swine

gut. BMC Microbiol. 11:103.

Lin J. 2011. Effect of Antibiotic Growth Promoters on Intestinal Microbiota in Food

Animals: A Novel Model for Studying the Relationship between Gut Microbiota

and Human Obesity? Front Microbiol. 2: 53.

Loc Carrillo, C., R. J. Atterbury, A. El-Shibiny, P. L. Connerton, E. Dillon, A. Scott, and

I. F. Connerton. 2005. Bacteriophage therapy to reduce Campylobacter jejuni

colonization of broiler chickens. Appl. Environ. Microbiol. 71:6554–6563.

Loc-Carrillo, C., and S. T. Abedon. 2011. Pros and cons of phage therapy. Bacteriophage

1:111–114.

Page 151: Phage therapy applications: Efficacy and impact on the ...

137

137

Looft, T., H. K. Allen, T. a Casey, D. P. Alt, and T. B. Stanton. 2014. Carbadox has both

temporary and lasting effects on the swine gut microbiota. Front. Microbiol.

5:276.

Looft, T., T. A. Johnson, H. K. Allen, D. O. Bayles, D. P. Alt, R. D. Stedtfeld, W. J. Sul,

T. M. Stedtfeld, B. Chai, J. R. Cole, S. A. Hashsham, J. M. Tiedje, and T. B.

Stanton. 2012. In-feed antibiotic effects on the swine intestinal microbiome. Proc.

Natl. Acad. Sci. U. S. A. 109:1691–6.

Looft T., H. K. Allen, B. L. Cantarel, U. Y. Levine, D. O. Bayles, D. P. Alt, B. Henrissat,

and T. B. Stanton. 2014. Bacteria, phages and pigs: the effects of in-feed

antibiotics on the microbiome at different gut locations. The ISME Journal. 8:

1566–1576.

McMurdie, P. J., and S. Holmes. 2013. phyloseq: an R package for reproducible

interactive analysis and graphics of microbiome census data. PLoS One 8:e61217.

Rettedal, E., S. Vilain, S. Lindblom, K. Lehnert, C. Scofield, S. George, S. Clay, R. S.

Kaushik, A. J. M. Rosa, D. Francis, and V. S. Brözel. 2009. Alteration of the ileal

microbiota of weanling piglets by the growth-promoting antibiotic

chlortetracycline. Appl. Environ. Microbiol. 75:5489–95.

Rozema, E. A., T. P. Stephens, S. J. Bach, E. K. Okine, R. P. Johnson, K. Stanford, and

T. A. McAllister. 2009. Oral and rectal administration of bacteriophages for

control of Escherichia coli O157:H7 in feedlot cattle. J. Food Prot. 72:241–250.

Saez, A. C., J. Zhang, M. H. Rostagno, and P. D. Ebner. 2011. Direct feeding of

microencapsulated bacteriophages to reduce Salmonella colonization in pigs.

Foodborne Pathog. Dis. 8:1269–74.

Page 152: Phage therapy applications: Efficacy and impact on the ...

138

138

Sarker, S. A., S. McCallin, C. Barretto, B. Berger, A. C. Pittet, S. Sultana, L. Krause, S.

Huq, R. Bibiloni, A. Bruttin, G. Reuteler, and H. Brüssow. 2012. Oral T4-like

phage cocktail application to healthy adult volunteers from Bangladesh. Virology

434:222–232.

Stenina M. A., L. I. Krivov, D. A. Voevodin, V. I. Savchuk, L. V. Kovalchuk, and V. N.

Yarygin. 2012 Cytokine profile of the blood in mice with normal and abnormal

heart rhythm. Bull Exp Biol Med 152:692-5.

Wall, S. K., J. Zhang, M. H. Rostagno, and P. D. Ebner. 2010. Phage therapy to reduce

preprocessing Salmonella infections in market-weight swine. Appl. Environ.

Microbiol. 76:48–53.

Page 153: Phage therapy applications: Efficacy and impact on the ...

139

139

CHAPTER 6. GENERAL CONCLUSIONS

The main objective of this study is to develop phage-based food safety

interventions to reduce contamination of food by two common foodborne pathogens, E.

coli O157:H7 and Salmonella, from “farm to fork”. In this study, the antibacterial

efficacy of phage treatments was tested in different food matrices, under different

conditions, as well as in both phage cocktail and single phage forms. Additionally, this

study also examined phage genome sequences, bacterial phage resistance, and the impact

of orally administrated phages on animal acute immune response and gut microbiome in

effort to more clearly assess the safety and long-lasting efficacy of phage treatments.

Both the second and fourth chapters have demonstrated the efficacy of phage

treatment in reducing bacterial pathogens in various food matrices. In chapter two, the

capacity of a three-phage cocktail to reduce the contamination of E. coli O157:H7 in

ground beef (room temperature for 24h, refrigeration for 24h, and undercooked

condition), spinach, and cheese slices (room temperature for 24h, 48h, and 72h for both

spinach and cheese slices) was tested. In chapter four, the ability of a single-broad-

spectrum phage to reduce Salmonella serovars in ground pork and liquid eggs (room

temperature for 24h and 48h and refrigeration for 24h, 1w and 2w) was tested. While

both experiments demonstrated the potential of phage treatments as antibacterial

interventions in foods, the results also showed that: 1) food matrices could interfere

Page 154: Phage therapy applications: Efficacy and impact on the ...

140

140

greatly with phage treatments; and 2) temperature could affect the efficacy of phage

treatment. The effects of food matrix observed in this study were two-fold. In chapter two,

it was showed that phage treatment significantly reduced the levels of E. coli O157:H7 in

phage treated ground beef and spinach, but not in cheese slices. And in chapter four, it

was shown that after treatment at room temperature for two days, phage treatment

resulted in an emergence of phage resistant Salmonella in liquid eggs, but not in ground

pork. The food matrix effect, either in terms of efficacy or the development of phage

resistance, could be associated with the differences in food composition, structure, and

production processes. Meanwhile, experiments also showed that treatments with either

the E. coli O157:H7 phage cocktail or the Salmonella single, broad-spectrum phage were

more effective at room temperature, which was more close to the optimal growth

temperature of host bacteria, than at refrigeration.

Whole genome annotation is considered an essential step in phage

characterization and can provide information of phages at the molecular level and help

ensure the absence of undesired genes. The genome sequences of several Salmonella

phages have been characterized previously (Zhang et al., 2014). In chapter three, the

genome sequences of two E. coli O157:H7 phages used in chapter one were characterized.

Phages vB_EcoM_FFH_1 and vB_EcoM_FFH_2 had genome lengths of 108,483 bp and

139,020 bp, respectively. Based on sequence homology, FFH_1 was classified as a T5-

like phage and FFH_2 was classified as an rV5-like phage. No undesired genes, including

genes coding for antibiotic resistance, toxins, virulence, or lysogeny, were observed in

either E. coli phage genomes. However, 108 out of 160 predicted genes in FFH_1 and

Page 155: Phage therapy applications: Efficacy and impact on the ...

141

141

163 out of 220 predicted genes in FFH_2 did not match to any genes with characterized

functions in NCBI database.

The development of phage resistance is generally considered one of the obstacles

preventing phage application being widely used in practice (Tanji et al., 2004). In chapter

two, several phage resistant E. coli O157:H7 strains were characterized in terms of phage

resistant mechanisms, in vitro stability of phage resistance, and potential fitness losses.

Different resistance mechanisms were observed among phage resistant bacterial strains.

Unlike in some other studies (O’Flynn et al., 2004; Scott et al., 2007), where phage

resistant strains quickly reverted to susceptibility after the absence of phages, all three

resistant strains of this study remained resistant to corresponding phages after a four-day

in vitro sub-culturing. Meanwhile, no significant reductions in bacterial growth or

adhesion to mammalian intestinal cells were observed in any resistant strains. While it is

possible that, in practice, fitness loss could still result in the out-competition of phage

resistant strains due to limited nutrients and more complex microbial community in the

environments where phage treatments are commonly applied, the results of this study

indicated that the emergence of phage resistant bacteria could interfere with the efficacy

of phage treatments, and therefore, mitigation strategies should be developed. One

potential mitigation strategy is the use of phage cocktails consisting of several distinct

phages, as it is much less likely for bacteria to develop resistance to multiple, unrelated

phages simultaneously. While phage resistance was not tested after the treatment with the

three-phage cocktail against E. coli O157:H7 (Chapter two), as expected, high

percentages of phage resistant Salmonella colonies were observed after treatment with

single-broad-spectrum Salmonella phage, at least in liquid eggs (Chapter four).

Page 156: Phage therapy applications: Efficacy and impact on the ...

142

142

While extensive studies have been conducted examining phage efficacy, far fewer

studies are available demonstrating the impact of phage treatments on animal acute

immune responses and the gut microbiome. In Chapter five, acute immune responses

were measured in mice treated with both a low amount of phages (105 PFU), and a high

amount of phages (107 PFU). No differences were observed in the levels of intestinal IgA

and IgG among different treatment groups at 6 h or 24 h post treatment. Phage treatments

did elicit some cytokine response, however, all cytokine concentrations were within

normal ranges for mice at all sampling times across all treatments. The gut microbiome at

the ileum, cecum and feces were also characterized using 16S rRNA sequencing in pigs

receiving control (basal nursery diet), phage treatment (basal diet with daily inoculation

of 5 x 1010 PFU phages via oral gavage) and antibiotic treatment (basal diet supplemented

with antibiotic premix ASP 250) for two weeks. While antibiotic treatment significantly

altered the ileal microbiome compared to control, no consistent changes were observed

between the phage treated microbiome and the control microbiome, in either the overall

microbiome composition or the relative taxonomic abundance. Relative abundance

differences, however, were observed with certain OTUs between the phage treated and

control microbiomes at three intestinal compartments and further studies are needed to

determine whether these changes are deleterious to host.

Taken together, this study demonstrated the potential of phage treatments as

antibacterial interventions in various foods. Phage resistant bacteria, however, were

recovered after treatment using single-broad-spectrum phages in liquid eggs. The

characterization of phage resistant E. coli O157:H7 strains indicated that the development

of phage resistance may result in only limited fitness loss in resistant strains, and

Page 157: Phage therapy applications: Efficacy and impact on the ...

143

143

therefore, mitigation strategies, such as using phage cocktails, should be considered.

Additionally, this study also showed that phage administration would not elicit adverse

acute immune response or greatly alter the gut microbiome in animals, which provides

more knowledge for the applications of phages in live animals.

Page 158: Phage therapy applications: Efficacy and impact on the ...

144

144

List of Reference

O’Flynn, G., R. P. Ross, G. F. Fitzgerald, and A. Coffey. 2004. Evaluation of a Cocktail

of Three Bacteriophages for Biocontrol of Escherichia coli O157:H7. Appl.

Environ. Microbiol. 70:3417–3424.

Scott, A. E., A. R. Timms, P. L. Connerton, C. L. Carrillo, K. A. Radzum, and I. F.

Connerton. 2007. Genome dynamics of Campylobacter jejuni in response to

bacteriophage predation. PLoS Pathog. 3:1142–1151.

Tanji, Y., T. Shimada, M. Yoichi, K. Miyanaga, K. Hori, and H. Unno. 2004. Toward

rational control of Escherichia coli O157:H7 by a phage cocktail. Appl. Microbiol.

Biotechnol. 64:270–274.

Zhang, J., Y. Hong, N. J. Harman, A. Das, and P. D. Ebner. 2014. Genome sequence of a

Salmonella phage used to control Salmonella transmission in Swine. Genome

Announc. 2.

Page 159: Phage therapy applications: Efficacy and impact on the ...

145

VITA

Page 160: Phage therapy applications: Efficacy and impact on the ...

145

145

VITA

Yingying Hong

EDUCATION

PhD, Animal Sciences Dec 2015

Purdue University, West Lafayette, IN

Concentration: Microbiology and food safety

Dissertation: Phage therapy applications: efficacy and impact on the microbiome

Advisors: Paul Ebner and Alan Mathew

MS, Animal Sciences May 2011

University of Tennessee, Knoxville, TN

Concentration: Microbiology and food safety

Thesis: Evaluation of chromosomally integrated luxCDABE and plasmid-borne GFP

markers for the study of localization and shedding patterns of shiga toxin producing E.

coli (STEC) O91:H21 in calves

Advisor: Alan Mathew

BS, Bioresource Science Jun 2008

Zhejiang University, Hangzhou, China

Advisor: Yueming Wu

Page 161: Phage therapy applications: Efficacy and impact on the ...

146

146

RESEARCH EXPERIENCE

Graduate Research Assistant, Purdue University Jun 2011-Present

Evaluation of the impact of orally administrated phages on intestinal microbiome of pigs

• Lead and conducted animal experiments

• Designed protocols and constructed 16s rRNA library to profile the composition of

gut microbiome

• Collaborated with colleagues from Genomic Core at Purdue in data analysis

Treatment of Salmonella contaminated eggs and pork with a broad-spectrum, single

bacteriophage

• Designed protocol and evaluated the efficacy of a broad-spectrum, single phage

based treatment to control Salmonella contamination in liquid eggs and ground pork

• Developed high throughput assay to measure phage susceptibility

• Assessed treatment-associated development of phage resistance

Analysis of bacteriophage genome

• Designed protocols for genome annotation of E. coli and Salmonella phages using

various bioinformatic software and databases

• Conducted phage genome extraction, whole genome annotation, and data submission

to NCBI database

Characterization of phage resistance in Escherichia coli O157:H7

• Built a collection of phage resistant Escherichia coli O157:H7 strains

• Designed protocols and investigated the impact of phage resistance in phage

applications, including mechanisms, stability of phage resistance, and growth fitness

of phage resistant bacteria

Page 162: Phage therapy applications: Efficacy and impact on the ...

147

147

Graduate Research Assistant, University of Tennessee Aug 2008-May 2011

Characterization of E. coli O91:H21 (STEC) shedding patterns in calves using a novel

bacterial challenge model

• Constructed luminescent and green fluorescent STEC strains

• Assessed the application of two STEC models in calves

• Evaluated the effect of environmental temperature on shedding of STEC in calves

Undergraduate Research Project, Zhejiang University Sep 2007-Jun 2008

Effect of Chinese herbal additives on growth performance and diarrhea prevention of

early-weaned piglets

• Performed ethanol extraction of bioactive components from Chinese herbal medicine

• Measured animal growth performance

OTHER EXPERIENCE

Undergraduate Research Abroad Program (URAP), Purdue University

2007 and 2011-2015

• Participated as an undergraduate student during the summer of 2007 and completed a

two-month research project as part of a larger project focused on phage therapy in

livestock production

• Participated as a graduate student during the summers of 2011 to 2015 and mentored

six undergraduate students of URAP in conducting and reporting their research

projects

4-H Workshop for Youth, Purdue University Jun 2013

• Guided 210 high school students with hands-on experiments to demonstrate food

safety challenges in daily food handling and preparation

Page 163: Phage therapy applications: Efficacy and impact on the ...

148

148

Mentor for Undergraduate Research, Purdue University 2012-present

• Directly mentored over 10 undergraduates in for-credit research internships on

various food safety topics

• Coached undergraduates through data collection, analysis, and publication

Teaching Assistant, University of Tennessee-Knoxville s Spring 2010

• Arranged laboratory course for ANSC160 (Introduction to Animal Science) for

approximately 70 undergraduate students and assisted instructors with exam grading

PUBLICATIONS

• Hong Y., Y. Pan, and P. D. Ebner. 2014. Meat science and muscle biology

symposium: Development of bacteriophage treatments to reduce Escherichia coli

O157:H7 contamination of beef products and produce. J. Anim. Sci. 92:1366–1377.

• Hong Y., Y. Pan, N.J. Harman, and P. D. Ebner. 2014. Complete genome sequences

of two Escherichia coli O157:H7 phages effective in limiting contamination of food

products. Genome Announc. 2(5): e00519-14.

• Zhang J, Y. Hong, M. Fealey, A. Singh, K. Walton, C. Martin, N.J. Harman, J.

Mahlie, and Ebner PD. Physiological and molecular characterization of Salmonella

bacteriophages previously used in phage therapy (in press)

• Zhang J., Y. Hong, N.J. Harman, A. Das, and P. D. Ebner. 2014. Genome sequence

of a Salmonella phage used to control Salmonella transmission in swine. Genome

Announc. 2(5): e00521-14.

• Mathew A. G., Liamthong S., Lin J., and Hong Y. (2009) Evidence of class 1

integron transfer between Escherichia coli and Salmonella spp. on livestock farms.

Foodborne Pathog Dis 6(8): 959-964.

Page 164: Phage therapy applications: Efficacy and impact on the ...

149

149

PUBLICATIONS IN PREPARATION

• Hong Y., K. Schmidt, and P. D. Ebner. 2015. Phage therapy and the host: acute

immune response and impact on the microbiome.

• Hong Y., K. Schmidt, L. Albino, A. M. Marshall, and P. D. Ebner. 2015. Treatment

of Salmonella contaminated eggs and pork with a broad-spectrum, single

bacteriophage: assessment of efficacy and resistance development

PRESENTATIONS

• Hong Y., J. Zhang, J. Thimmapuram and P.D. Ebner (2015) “The effect of phage

treatment on animal immune response and intestinal microbiome” Evergreen

International Phage Meeting, Olympia, WA (Poster presentation)

• Hong Y. and P. D. Ebner (2015) “The use of bacteriophages to improve food safety”

Indiana Small Farm Conference (Extension Conference), Danville, IN (Poster

presentation)

• Hong Y., Y. Pan, and P. D. Ebner (2014) “Bacteriophage-based alternatives to

antibiotics in livestock and food production” Indiana Small Farm Conference

(Extension Conference), Danville, IN (Poster presentation)

• Hong Y. and P.D. Ebner (2013) “Characterization of phage-resistant Escherichia coli

O157:H7” ADSA®-ASAS Joint Annual Meeting, Indianapolis, IN (Oral

presentation)

• Hong Y. and A.G. Mathew (2011) “Characterization of a chromosomally integrated

luxCDABE marker for investigation of Shiga toxin-producing Escherichia coli

O91:H21 shedding in cattle” SPIE, Orlando, FL (Oral presentation)