ArticlePDF Available

A review of phage mediated antibacterial applications A review of phage mediated antibacterial applications

Authors:

Abstract and Figures

Background: For over a decade, resistance to newly synthesized antibiotics has been observed worldwide. The challenge of antibiotic resistance has led to several pharmaceutical companies to abandon the synthesis of new drugs in fear of bacteria developing resistance in a short period hence limiting initial investment return. To this effect, alternative approaches such as the use of bacteriophages to treat bacterial infections are being explored. This review explores the recent advances in phage-mediated antibacterial applications and their limitations. Methods: We conducted a comprehensive literature search of PubMed, Lib Hub and Google Scholar databases from January 2019 to November 2019. The search key words used were the application of bacteriophages to inhibit bacterial growth and human phage therapy to extract full-text research articles and proceedings from International Conferences published only in English. Results: The search generated 709 articles of which 95 full-text research articles fulfilled the inclusion guidelines. Transmission Electron Microscopy morphological characterization conducted in 23 studies registered Myoviruses, Siphoviruses, Podoviruses, and Cytoviruses phage families while molecular characterization revealed that some phages were not safe to use as they harbored undesirable genes. All in vivo phage therapy studies in humans and model animals against multidrug-resistant (MDR) bacterial infection provided 100% protection. Ex vivo and in vitro phage therapy experiments exhibited overwhelming results as they registered high efficacies of up to 100% against MDR clinical isolates. Phage-mediated bio-preservation of foods and beverages and bio-sanitization of surfaces were highly successful with bacterial growth suppression of up to 100%. Phage endolysins revealed efficacies statistically comparable to those of phages and restored normal ethanol production by completely eradicating lactic acid bacteria in ethanol fermenters. Furthermore, the average multiplicity of infection was highest in ex vivo phage therapy (557,291.8) followed by in vivo (155,612.4) and in vitro (434.5).
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=tajm20
Alexandria Journal of Medicine
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/tajm20
A review of phage mediated antibacterial
applications
Kenneth Ssekatawa , Denis K. Byarugaba , Charles D. Kato , Eddie M.
Wampande , Francis Ejobi , Robert Tweyongyere & Jesca L. Nakavuma
To cite this article: Kenneth Ssekatawa , Denis K. Byarugaba , Charles D. Kato , Eddie M.
Wampande , Francis Ejobi , Robert Tweyongyere & Jesca L. Nakavuma (2021) A review of
phage mediated antibacterial applications, Alexandria Journal of Medicine, 57:1, 1-20, DOI:
10.1080/20905068.2020.1851441
To link to this article: https://doi.org/10.1080/20905068.2020.1851441
© 2020 The Author(s). Published by Informa
UK Limited, trading as Taylor & Francis
Group.
View supplementary material
Published online: 31 Dec 2020.
Submit your article to this journal
View related articles
View Crossmark data
REVIEW ARTICLE
A review of phage mediated antibacterial applications
Kenneth Ssekatawa
a,b,c
, Denis K. Byarugaba
a
, Charles D. Kato
a
, Eddie M. Wampande
a
, Francis Ejobi
a
,
Robert Tweyongyere
a
and Jesca L. Nakavuma
a
a
College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda;
b
Department of Biochemistry,
Faculty of Biomedical Sciences, Kampala International University-Western Campus, Bushenyi.;
c
African Center of Excellence in Materials
Product Development and Nanotechnology (MAPRONANO ACE), College of Engineering Design Art and Technology, Makerere University,
Kampala, Uganda
ABSTRACT
Background: For over a decade, resistance to newly synthesized antibiotics has been observed
worldwide. The challenge of antibiotic resistance has led to several pharmaceutical companies
to abandon the synthesis of new drugs in fear of bacteria developing resistance in a short
period hence limiting initial investment return. To this eect, alternative approaches such as
the use of bacteriophages to treat bacterial infections are being explored. This review explores
the recent advances in phage-mediated antibacterial applications and their limitations.
Methods: We conducted a comprehensive literature search of PubMed, Lib Hub and Google
Scholar databases from January 2019 to November 2019. The search key words used were the
application of bacteriophages to inhibit bacterial growth and human phage therapy to extract
full-text research articles and proceedings from International Conferences published only in
English.
Results: The search generated 709 articles of which 95 full-text research articles fullled the
inclusion guidelines. Transmission Electron Microscopy morphological characterization con-
ducted in 23 studies registered Myoviruses, Siphoviruses, Podoviruses, and Cytoviruses phage
families while molecular characterization revealed that some phages were not safe to use as
they harbored undesirable genes. All in vivo phage therapy studies in humans and model
animals against multidrug-resistant (MDR) bacterial infection provided 100% protection. Ex vivo
and in vitro phage therapy experiments exhibited overwhelming results as they registered high
ecacies of up to 100% against MDR clinical isolates. Phage-mediated bio-preservation of
foods and beverages and bio-sanitization of surfaces were highly successful with bacterial
growth suppression of up to 100%. Phage endolysins revealed ecacies statistically compar-
able to those of phages and restored normal ethanol production by completely eradicating
lactic acid bacteria in ethanol fermenters. Furthermore, the average multiplicity of infection
was highest in ex vivo phage therapy (557,291.8) followed by in vivo (155,612.4) and in vitro
(434.5).
ARTICLE HISTORY
Received 19 August 2020
Revised 4 November 2020
Accepted 10 November 2020
KEYWORDS
Antibiotics resistance;
bacteriophages; phage
therapy; phage mediated
biocontrol; phage efficacy
1. Background
Currently, the world populace is deemed to be at
a great risk as a result of the ever-escalating prevalence
of antibiotic resistance bringing about an epoch where
many familiar bacterial infections are becoming
increasingly hard to treat [1]. Similar to many other
developing countries, Sub-Saharan Africa is experien-
cing an elevated burden of bacterial infectious diseases
which calls for the overuse of antibiotics and conse-
quently emergence of resistant microorganisms [1,2].
The development of antibiotic resistance is also con-
tributed by self-medication with uncontrolled over-
the-counter access to drugs without any guidance
from qualified medical practitioners. In addition,
there is excessive application of antibiotics in poultry,
aquaculture, and livestock production. The unrest-
ricted access and use of antibiotics for animal disease
treatment and prophylaxis as well as growth promo-
tion have been implicated as one of the major drivers
for antibiotic resistance that may spillover to humans
[3–5]. Infectious food and water-borne illnesses are
acquired through the consumption of contaminated
food and water; and are the major cause of mortality
and morbidity worldwide owing to their extensive and
spontaneous transmission [6,7]. It was estimated that
water, sanitation, and hygiene (WSH) associated
infectious diseases are accountable for 4.0% of the
worldwide deaths and 5.7% of the universal disease
burden [7,8]. Furthermore, WHO reported that
600 million or 1 in 10 people fall ill worldwide as
a result of foodborne infections and more than
91 million people affected are in Africa [6].
The rate at which drug resistance emerges has
resulted in big pharmaceutical companies backing
away from developing new antibiotics since the latter
CONTACT Dr. Jesca L. Nakavuma jesca.nakavuma@gmail.com College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere
University, Kampala, Uganda
Supplemental data for this article can be accessed here.
ALEXANDRIA JOURNAL OF MEDICINE
2021, VOL. 57, NO. 1, 1–20
https://doi.org/10.1080/20905068.2020.1851441
© 2020 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits
unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
are rendered non-effective within a short period, mak-
ing the venture not cost-effective [9]. Therefore,
affordable alternative approaches such as the use of
probiotics, phytomedicines, and bacteriophages to
manage bacterial infections and control the emergence
of antibiotic resistance are highly commendable.
Bacteriophages (phages) are natural enemies of bac-
teria which are the most abundant replicating entities
on earth. Phages are viruses that specifically attack and
multiply in bacterial cells and have no effect on other
cell types. They are self-replicating and self-limiting as
long as the specific bacterial host cells exist. Similar to
other viruses, their genomes may either be double-
stranded or single-stranded DNA or RNA [10].
Phages have either a lytic or lysogenic type of replica-
tion cycle. The lytic cycle, also referred to as the virulent
cycle, results in the production of progeny viruses that
are released through cell lysis. The lysogenic or tempe-
rate cycle results in the incorporation of the phage
genome into the host chromosome without the produc-
tion of new virus particles. Depending on some circum-
stances, some phages can exhibit both replication cycles
[10]. Lytic phages are applied as bacterial growth inhi-
bitors, which can be categorized as phage therapy or
phage-mediated decontaminants. For therapy, phages
are mainly used like antibiotics, whereas for deconta-
mination, they are applied as disinfectants. Literally,
phage therapy is the application of phages as therapeu-
tic agents more especially in a clinical context to treat
bacterial infections while phage-mediated biocontrol
can be defined as the use of phages to suppress bacterial
growth on non-living surfaces. Safety and efficacy of
phage therapy or phage-mediated biocontrol relies on
isolation and use of only professional lytic phages,
which are obligately lytic or virulent but they are
neither temperate nor directly linked to temperate
phages [11]. Phage therapy is a proven eco-friendly
alternative approach to prevent and control pathogenic
bacterial infections [12,13].
Phages were used to treat bacterial infections in
Europe during the pre-antibiotic era. However, with
the discovery of antibiotics and the substandard med-
ical trials conducted in the western world without
putting into consideration that phages were specific,
phage therapy was shortly after deemed impotent in
the treatment of bacterial infections. Nevertheless,
phage therapy continued to be used for the treatment
of bacterial infections in the Soviet Union since 1940
[14]. The advantages of phage applications, such as
disruption of bacterial biofilms and nondependency
on the drug resistance status of the organisms, have
rekindled their use as antibacterial agents [15,16].
Furthermore, renewed attention to phage therapy has
been registered due to an overall decline in the total
reserves of effective antibiotics. Hence, phage therapy
clinical trials and experiments in poultry, aquaculture,
crop husbandry, model animals, in vitro model
systems, and humans have been widely carried out
[17,18]. Currently, the notable human phage therapy
under application is the compassionate use of phages
as individualized therapeutic options to manage MDR
bacterial infections unresponsive to all classes of con-
ventional antibiotics [19]. Furthermore, phage pre-
parations have been used and experimented with as
diagnostic tools for bacterial infections to supplement
the available methods [12].
For use as decontaminants, several studies have been
conducted to evaluate the efficacy of phages as bio-
control agents against food and beverage borne patho-
gens [20]. Phages have been experimented with in bio-
sanitization of equipment surfaces to eradicate biofilms
in food industries [21]; and bio-preservation of perish-
able processed foods to increase shelf-life. Some phage-
specific enzymes; such as lysins which degrade the cell
wall of gram-positive bacteria, have been applied to
processed foods to enhance their safety for human
consumption [18,22–24]. The use of bacteriophages in
food products in the US, Europe, and Australia has been
reported [25]. Indeed, some phage preparations have
been approved in the USA and are commercially avail-
able; such as LISTEX P100; LMP-102
TM
, Listshield
TM
,
ECP-100
TM
(Ecoshield
TM
), SALMONELEX
TM
,
AgriPhage
TM
, and Biophage-PA [26].
This review expounds on the current level, limita-
tions, and prospects of phage applications such as
enhancing food safety and fermentation of biofuels;
phage therapy clinical trials and experiments in
humans and model animals; animal and plant disease
control and environmental bioremediation.
2. Methods
2.1. Literature search strategy
A comprehensive literature search of PubMed, Lib
Hub, and Google Scholar databases was conducted
from January 2018 to November 2018. The search
key words used were “application of bacteriophages
to inhibit bacterial growth” and “human phage ther-
apy,” Figure 1.
3. Study selection criteria
The search targeted articles published in English with-
out restriction on year of publication in an attempt to
capture all available literature about the application of
phages as antibacterial agents worldwide, Figure 1. In
addition, only full-text research articles and proceed-
ings from the International Conference on Prevention
& Infection Control were selected, Table 1–5, S1.
Review articles were excluded from this search. To
avoid bias, all the seven coauthors were involved in
the selection process. Articles were assigned to the
different coauthors blindly, review reports on the
2K. SSEKATAWA ET AL.
merits and demerits of the studies as per inclusion
criteria were submitted to the lead researcher (JLN)
and the entire selection process was conducted based
on the review reports by all the seven coauthors. In
case of any disagreement, powers were entrusted to
the most experienced researchers in bacteriophages
(JLN, DKB, and FE) to make the final decision.
4. Data extraction
A database was created in which the field of phage
application, type of phage or phage part used, source
of phages, level of phage application, type of bacteria
and strain or serovar challenged, level of phage effi-
cacy, physiochemical properties of phages, the multi-
plicity of infection (MOI) of phages and methods
used in the characterization of phages were included.
Studies where MOIs were not reported but the num-
ber of plaque-forming units/mL (PFU/mL) and the
number of colony-forming units/mL (CFU/mL)
given, MOIs were computed by dividing the PFU/
mL by CFU/mL units (O’Flynn et al., 2004). To
compare the MOI of different investigations, all stu-
dies were grouped into three categories namely;
in vivo phage therapy, ex vivo phage therapy, and
in vitro phage therapy.
Figure 1. Selection process of research articles for inclusion in this review.
Table 1. In vivo human phage therapy trials.
Phage therapy in humans Phage type Source of phages Pathogens targeted
Serovar/
pathotype efficacy Ref
Treatment of diabetic toe
ulcers
Staphylococcal phage Sb-1 Eliava Institute S. aureus (MRSA and
MSSA)
- 100% [93]
Treatment of GIT MRSA
infection
polyvalent S. aureus bacteriophages L. Hirszfeld Institute
collection
S. aureus (MRSA) - 100% [94]
Treatment of burn infections - J. Soothill P. aeruginosa - 100% [95]
Treatment of infected
venous stasis ulcers and
other poorly healing
wounds
Pyophage in PhagoBioDerm films Eliava Institute P. aeruginosa, E. coli,
S. aureus, Proteus,
and Streptococcus
- 76% [49]
Treatment of corneal
abscess and interstitial
keratitis
S. aureus bacteriophage SATA-8505 ATCC VRSA - 100% [96]
Treatment of
burn wound infection
Cocktail of P. aeruginosa phages 14/1
(Myoviridae) and PNM (Podoviridae)
and S. aureus phage ISP (Myoviridae)
Merabishvili et al 2009 S. aureus and
P. aeruginosa
- 0% [69]
Treatment of chronic otitis
antibiotic-resistant
P. aeruginosa
Infection
Biophage-PA NCIMB MDR P. aeruginosa - 80% [97]
Treatment of P. aeruginosa
UTI
PA Phage cocktail (Pyophage #051007) Eliava Institute MDR P. aeruginosa - 100% [98]
Treatment of acute bacterial
diarrhea
T4-like coliphages cocktail Microgen-Russia E. coli - 0% [70]
Treatment chronic bacterial
prostatitis
IIET bacteriophage collection IIET bacteriophage
collection sewage,
environmental, or
drinking water
Enterococcus faecalis - 100% [99]
Phage safety analysis Phage cocktail Coli Proteus Microgen Russia E. coli and proteus - - [30]
ALEXANDRIA JOURNAL OF MEDICINE 3
Table 2. Phage-mediated biocontrol of bacterial growth in ethanol fermentation, foods, and beverages.
Field of application Phage/phage part used Source of phages/part
Level of
application Bacteria type controlled Bacteria serotype/Strain Efficacy Ref.
Ethanol fermentation Streptococcal phage LambdaSa2 (λSa2) endolysin EMD Biosciences, San
Diego,CA)
Laboratory
experiment
Lactobacillus, staphylococci, and
streptococci
77.3% [100]
LysA, LysA2, LysgaY and λSa2 endolysin proteins Subcloned into the pET21a
E. coli expression vector
Ethanol fermentation Lytic enzymes LysA and LysA2 endolysins genes expressed
in Saccharomyces
cerevisiae
Laboratory
experiment
Lactobacillus fermentum,
Lactobacillus brevis, and
Lactobacillus mucosae.
~ 90% [101]
Ethanol fermentation EcoSau and EcoInf Wastewater influent Laboratory
experiment
L. fermentum 100% [37]
Ethanol fermentation ATCC® 8014-B1™ (phage B1) and ATCC® 8014-B2™ (phage
B2)
ATCC Laboratory
experiment
Lactobacillus plantarum ATCC®
8014™
ATCC® 8014™ 99% [92]
Dairy (Cheese) Phage P100 Dairy plant sewage effluent Laboratory
experiment
L. monocytogenes WSLC 1001 100% [39]
Dairy (Milk) vB_SauS-phiIPLA35 Dairy environment Laboratory
experiment
Staphylococcus aureus 100% for
cocktail [102]
Dairy (Milk) and control of E. coli
biofilms
BECP2 and BECP6 phages Sewage Laboratory
experiment
E. coli O157:H7 90% [103]
Dairy (Milk fermentation) Coliphages DT1 and DT6 Feces Laboratory
experiment
E. coli O157:H7 STEC 100% [104]
Dairy (Cheese) phage A511 - Laboratory
experiment
Listeria monocytogenes, 90% [105]
Fruits (Cucumber, Apple, and
Tomatoes)
T7 bacteriophages Laboratory
experiment
Escherichia coli BL21 99.9% [106]
Fresh-cut fruits and vegetables LM-103 and LMP-102, Intralytix, Inc. (Baltimore,
Md.).
Laboratory
experiment
Listeria monocytogenes, 99.9% [107]
Dairy, poultry, beef products, sea
food, and vegetables
A511 and P100 - Laboratory
experiment
Listeria monocytogenes WSLC 1001 (serovar 1/2 c)
and Scott A (serovar 4b)
100% [108]
Beer industry Myophage SA-C12 Fresh silage Laboratory
experiment
Lactobacillus brevis 8840 (NCIMB culture
collection),
100% [109]
Chicken cuts FSP-1 and FSP-3/PSZ1 and/PSZ2 Laboratory
Experiment
Salmonella enterica Strain S49 92% [110]
Spinach - Feedlot cattle feces Laboratory
Experiment
Escherichia coli O157:H7 100% [111]
Oysters Siphoviridae phage pVp-1, - Laboratory
experiment/trial
on oysters
Vibrio parahaemolyticus CRS 09–17 ~99.999% [112]
Fermented Soy bean paste BCP1-1 and BCP8-2 Fermented food products Laboratory
experiment
Bacillus cereus ATCC27348, ATCC21768,
ATCC13061
100% [113]
Bioactive packaging materials
(meat and alfalfa seeds and
sprouts)
LinM-AG8, LmoM-AG13, and LmoM-AG20, while the E. coli
O104:H4 EcoM-HG2, EcoM-HG7 and EcoM-HG8
(Myoviridae)
Canadian Research Institute
for Food Safety
Laboratory
experiments
Listeria monocytogenes and
Escherichia coli
E. coli O104:H4, LJH391
serotype 1/2b,
100% [114]
Infant formula milk ESP 1–3 and ESP 732–1 Sewage Laboratory
experiment
Enterobacter sakazakii ATCC 29,544, 236/04, 732/03 100% [115]
Infant formula milk leB, leE and leN Slurry Laboratory
experiment
Cronobacter sakazakii C. sakazakii ATCC BAA 894,
C. sakazakii ATCC BAA 894
LUX
100% [40]
(Continued)
4K. SSEKATAWA ET AL.
Table 2. (Continued).
Field of application Phage/phage part used Source of phages/part
Level of
application Bacteria type controlled Bacteria serotype/Strain Efficacy Ref.
Pork, milk, and kitchenware fHe-Yen3-01 (Podoviridae) fHe-Yen9-01, fHe-Yen9-02, fHe-
Yen9-03 (Myoviridae)
Sewage Laboratory
experiment
Yersinia enterocolitica O:3 strain 6471/76 and O:9
strain Ruokola/71
100% [41]
Milk, sausage, and lettuce LPST10, LPST18, and LPST23(Siphoviridae family) Waste water, sewage, farm
ditch, poultry house
Laboratory
experiment
Salmonella strains Typhimurium and
Salmonella Enteritidis
Salmonella Typhimurium
ATCC 14,028
64.1% [116]
Active food packaging system
(cellulose acetate films)
BFSE16, BFSE18, PaDTA1, PaDTA9,PaDTA10 and PaDTA11 Chickenfeces, poultry
exudates, and swine
feces
Laboratory
experiment
Salmonella enterica subsp. Enterica serovar
Typhimurium ATCC 14,028
100% [117]
Bioctive food packaging system BFSE16, BFSE18, PaDTA1, PaDTA9, PaDTA10 and PaDTA11 Poultry exudates and swine
feces
Laboratory
experiment
Salmonella enterica subsp. Enterica serovar
Typhimurium ATCC 14,028.
~ 99.99% [118]
Sea food (Cockles) phT4A, ECA2 Sewage Laboratory
experiment
Escherichia coli ATCC 13,706), 90% [119]
ALEXANDRIA JOURNAL OF MEDICINE 5
Table 3. In vitro phage therapy against clinical isolates assays.
Field of application Phage/phage part used Source of phages/part Level of application Bacterial targeted Strain/serovar/pathotype Level of efficacy Ref.
Phage activity against STEC and EHEC clinical
isolates
CA911, CA933P, MFA933P and
MFA45D
Minced meat, pork sauasage &
bovine feces.
Laboratory experiment Escherichia coli STEC and EHEC 100% [44]
Phage-Antibiotics synergism against E. coli
biofilm
T4 bacteriophage ATTC 11,303-B4 LGC Standards, Middlesex, UK) Laboratory experiment E. coli biofilms E. coli 11,303 100% [120]
Phage activity against Bacillus pumilus Phage FBa1, FBa2, and FBa3 River water Laboratory experiment Bacillus pumilus 100% [121]
Phage activity against Salmonella Typhimurium phSE-1, phSE-2, and phSE-5 (family
Siphoviridae)
Sewage Laboratory experiment Salmonella
Typhimurium
Enterica serovar Typhimurium 99% [42]
Phage activity against S. aureus clinical isolates Sb-1 - Laboratory experiment Staphylococcus aureus - 100% [122]
Phage activity against Pseudomonas fluorescens
biofilms
Phage IBB-PF7A sewage treatment plant Laboratory experiment P. fluorescens biofilms 91% [123]
Phage activity against SalmonellaTyphimurium P22-B1, P22, PBST10, PBST13, PBST32,
and PBST 35)
ATCC and Hankuk University of
Foreign Studies
Laboratory experiment SalmonellaTyphimurium KCCM 40,253, ATCC 19,585,
ATCC 19,585, and CCARM
8009.
54% [124]
Phage activity against Staphylococcus aureus
biofilm
Phage DRA88 and SAB4328-A Sewage Laboratory experiment
and ex vivo-burn
models
Staphylococcus aureus
biofilm
RN6390B, RN6911, B4328,
MSSA 3, MSSA 10, MRSA 82
Reduced
biofilm
formation
[125]
Phage activity against Pseudomonas aeruginosa
biofilm
DL 52, DL 54, DL 60, DL 62, DL 64, and
DL 68
Crude sewage Laboratory experiment P. aeruginosa biofilm PAO1, PA45311, PA45291,
PA45235
95% [35]
Phage activity against Staphylococcus aureus
biofilm
DRA88 and phage K Crude sewage Laboratory experiment Staphylococcus aureus
biofilm
1598, MRSA 252, & H325 100% [36]
Phage therapy against Staphylococcus aureus phage K and phage 92 ATCC Laboratory experiment MRSA/MSSA MRSA (N315, COL, Mu50)
ATCC 6538
100% [126]
Ex vivo Phage activity against catheter MRSA
biofilm
phage K ATCC Laboratory experiment
(Catheter)
MRSA biofilms Staphylococcus aureus 46,106 99% [127]
Ex vivo Phage activity against catheter Proteus
mirabilis and Escherichia coli biofilms
Escherichia coli T4 phage ATCC
11,303-B4
Bacteriofag coli-proteic, Microgen
Pharma, Russia
Laboratory experiment
(Catheter)
Proteus mirabilis and
E. coli biofilms
E. coli ATCC 11,303 and
P. mirabilis 13 HER1094
99.99% [128]
Phage activity against Pseudomonas aeruginosa
biofilm
- Hospital environmental dirt, sewage
disposal, and cattle waste
effluents
Laboratory experiment P. aeruginosa biofilms - 50% [129]
Phage therapy against Staphylococcus aureus P128 proteins Inducible T7 expression system in
E. coli ER2566 strain
Laboratory experiment Staphylococcus aureus BK#13,725, BK#9894,
BK#13,993
99.99% [130]
Phage activity against K. pneumoniae biofilms KPO1K2 and NDP, depolymerase, and
nondepolymerase producing
phages
- Laboratory experiment K. pneumoniae biofilms B5055 (O1:K2) Significant
eradication
[131]
phage activity against MRSA & MSSA . Hospital environmental dirt, sewage
disposal, and cattle waste
Laboratory experiment MRSA and MSSA 100% for MSSA
and 78%
MRSA
[132]
Phage activity against MDR Acinetobacter
baumanni
Phage AB2 Sewage Laboratory experiment MDR Acinetobacter
baumanni
A. baumannii M3237 99.90% [56]
Phage activity against MDR P. aeruginosa Sewage Laboratory Experiment MDR P. Aeruginosa - 100% [133]
Phage therapy assay against Pseudomonas
aeruginosa and Staphylococcus spp clinical
isolates
Intesti and Pyobacteriophag Eliava BioPreparations, Tbilisi,
Georgia
Laboratory experiment P. aeruginosa and
Staphylococcus spp.
- 100% [134]
Phage bacterial lytic activity against resistant
S. aureus
SA11 (Siphoviridae family Hankuk University of Foreign studies Laboratory experiment Resistant S. aureus ATCC 13,301 and CCARM 3080 99.99& [135]
Phage bacterial lytic activity against S. aureus
biofilms isolated from orthopedic Implant
StaPhage (Myoviridae) AusPhage Pty Ltd and sewage water Laboratory experiment Staphylococcus aureus
biofilms
ORI16C02N and ORI16025 98% [136]
Phage activity against P. aeruginosa biofilms P. aeruginosa phage M4 Health Protection Agency, Colindale,
United Kingdom
Laboratory experiment
(In vitro model
system)
Pseudomonas
aeruginosa biofilms
M4 99.9% [137]
6K. SSEKATAWA ET AL.
Table 4. In vivo and ex vivo phage therapy experiments in animal models/tissues, fish, plants, poultry, piggery, and bees.
Field of application Phages Source of phages Level of application Target bacteria
Target bacteria strain/
pathotype
Level of
efficacy Ref.
Phage therapy in model organisms
Treatment of P. aeruginosa infection in
insect
PA5oct and KT28 Natural wastewater treatment plant In vivo-insect model P. aeruginosa PA PAO1 and 0038 93.60% [138]
Treatment of Burkholderia pseudomallei in
mice
Phage C34 Sea water In vivo-mouse model Burkholderia
pseudomallei
- 33.30% [139]
Treatment of S. aureus infection in BALB/C
mice
MR-10 - In vivo-mouse model S. aureus ATCC 43,300 (MRSA)
and ATCC 29,213
(MSSA)
100% [140]
Treatment of S. aureus osteomyelitis in
Rabbits
SA-BHU1, SA-BHU2, SA-BHU8, SA-BHU15
and SA-BHU21, SA-BHU37, SA-BHU47)
River, pond, and sewage In vivo-Rabbit model MRSA - 100% [58]
Treatment of GIT pathogenic E. coli
infection in white rats
EHEC-specific coliphage http://www.sumobrain.com/patents/wipo/
Methodsbacteriophage-design/WO201
0064044A1.pdf.
In vivo-mouse model E. coli. EHEC and non-EHEC
E. coli
99.9% [141]
Treatment of A. baumannii infection in
Mouse Model
BC62 of Myoviridae family Sewage water In vivo-mouse model Carbapenem resistant
A. baumannii
100% [34]
Treatment of A. baumannii pneumonia in
BALB/c mice
vB_AbaM-IME-AB2 (IME-AB2), Sewage In vivo-mouse model A. baumannii clinical
isolates (MDR and
sensitive)
- 100% [59]
Treatment of P. aeruginosa keratitis in
mice
фR18 and ФS12-1 Sewage In vivo-mouse model P. aeruginosa - 99.78% [142]
Prevention of V. cholerae infections in
mouse and rabbits
Vibrio phages ICP1, ICP2, and ICP3 Human feces In vivo-mouse and
rabbit models
V. cholerae AC 53, AC2846, and
AC4653
100% [143]
Treatment of S. Enteritidis infection in
Caenorhabditis elegans worms
ΦSP-1 and ΦSP-3 Chicken feces In vivo-worm model Salmonella enteritidis S49 94.8% [144]
Treatment of PDR A. baumannii infections
in Mice and human cells
Abp1 Sewage In vivo-mice model and
Ex vivo-human HeLa
cells
PDR A. baumanni - 100% [60]
Treatment of Pseudomonas aeruginosa
skin infections
Phage PA709 characterized Sewage water Ex vivo-human skin MDR P. aeruginosa MDR P. aeruginosa
709
99.99% [145]
Treatment of K. pneumoniae wound
infections in BALB/c mice
phage Kpn5 Sewage In vivo experiment Klebsiella pneumoniae B5055 100% [146]
Crop protection
Biocontrol of potato bacterial wilt P-PSG-3, P-PSG-4, P-PSG-1, P-PSG-8 to
P-PSG-12
water Field trial Ralstonia solanacearum PS-X4-1, PS-X10-2,
and PS-X13-1
80% in vivo
and 98%
in vitro
[147]
Biocontrol of alfalfa seeds spoilage
Salmonella enterica
Phages SSP5 and SSP6 sewage In vitro and in vivo
laboratory
experiments
Salmonella enterica S. oranienburg 0% [148]
Biocontrol of tomato bacterial wilt RsPod1EGY Soil In vitro and in vivo
laboratory
experiments
Ralstonia solanacearum K3, K9, K10,
K11, K12, K16, K17,
and K19
100% [149]
Phage biocontrol of antibiotics resistant
Dickeya dadantii which causes potato
tuber rot
Myoviridae family Caspian Sea water Laboratory experiment/
field Trial
Dickeya dadantii 100% in vitro
88.9% for
trial
[150]
Acquaculture
Aquaculture H20-Siphovirus and KVP40-Myovirus Sea water Laboratory experiment Vibrio anguillarum (BA35 and PF430-3) Reduced
biofilms
[151]
(Continued)
ALEXANDRIA JOURNAL OF MEDICINE 7
Table 4. (Continued).
Field of application Phages Source of phages Level of application Target bacteria
Target bacteria strain/
pathotype
Level of
efficacy Ref.
Phage therapy in model organisms
Aquaculture (treatment of ulcerative
lesions in catfish)
PA phages Waste water Field trial Pseudomonas
aeruginosa (MDR)
- 100% [152]
aquaculture VP-2 and VA-1 phage Sewage water Laboratory experiment
and trial
Vibrio. anguillarum 100% [13]
Aquaculture FpV-4, FpV-9, and FpV-21 pond water Laboratory Experiment Flavobacterium
psychrophilum
Reduced
bacterial
growth
[153]
Treatment of Vibrio parahaemolyticus
shrimp infections
V. parahaemolyticus phages (Myoviridae
family)
Shrimp pond water suspended sediment Laboratory experiment/
trial
Vibrio
parahaemolyticus
N1A and N7A 90% [154]
Phage therapy in poultry
In vivo phage therapy against
Campylobacter spp. infections in broiler
chicken
typeIII phages NCTC12672, 12,673, 12,674,
and 12,678 of the British phage typing
scheme
Lohmann Animal Health, GmbH. Field trial Campylobacter spp. 99% [155]
In vitro phage therapy against APEC Phage ØEC1 Chicken feces Laboratory experiment E. coli APEC O78:K80 99.99% [156]
Phage therapy in piggery
In vivo phage therapy of Staphylococcus
aureus nasal infection in pigs
phage K*710 and P68 Novolytics Ltd Trial using animal
model
Staphylococcus aureus
(MRSA)
ST398, spa type t011,
SCCmec type V
0% [71]
Phage therapy in apiculture
In vivo phage therapy of American
foulbrood caused by Paenibacillus
larvae
Siphoviridae (HB10c2) Glue-like liquid of a beehive Laboratory experiment
and field Trial
Paenibacillus larvae ERIC I DSM 7030 and
ERIC II DSM 25,430
2% [43]
8K. SSEKATAWA ET AL.
Table 5. Phage application in biosanitization.
Field of application Phage Phage source
Level of
application Target bacteria Bacteria strain/pathotype
Level of
efficacy Ref
Water and sewage treatment
Water purification vB_AspP-UFV1
(Podoviridae)
Sludge of wastewater Laboratory
experiment
A. soli, Pseudomonas sp., and Brevundimonas sp. AO1-02, AO2-07, AO1-30, and
AO1-33
Significant
biofilm
control
[157]
Coliform phage biocontrol in sewage Coliphage (Myovirus
and Podovirus)
River water Laboratory
experiment
E. coli E. coli SBSWF27 95.40% [158,159]
Biosanitization
Hospital anitizer Staphylococcal
phage and
Pyophage; GA,
Eliava Institute Laboratory
experiment
S. aureus, E. coli, and P. aeruginosa S. aureus (SA2-R73), E. coli (EC-
R60), and P. aeruginosa
(PAV6).
90% [86]
Hospital sanitizer AB1, AB2, AB6,
AB7 phages
Sewage or river water Laboratory
experiment
and trial
CR A. baumannii - 47.50% [87]
Hospital sanitizer Pyobacteriophag
polyvalent
Research and Production
Association “Microgen”
(Russia))
Trial staphylococci, streptococci, enterococci, proteus, klebsiella
(pneumoniae, and oxytoca), P. aeruginosa and E. coli
- 100% [88]
Phage cream and sanitizers Polyvalent Anti-
Staphylococcus
Phage K
ATCC 19,685-B1 Laboratory
experiment
and trial
MDR Staphylococcus aureus (MRSA and VRSA) - 100% [89]
Use of phages in semi solid creams for
control of Propionibacterium acnes
growth
PAC1 to PAC10 P. acnes strains isolated from
facial skin swabs
Laboratory
experiment
Propionibacterium acnes A1, A2, or E8 100% [90]
ALEXANDRIA JOURNAL OF MEDICINE 9
5. Data analysis
Data analysis was performed using Tukey’s multiple
comparisons test in STATA version 2018.1 to establish
whether; (a) the number of studies that reported
in vivo human phage therapy efficacy of 100% was
more pronounced than the number of studies that
recorded efficacy lower than 100%, (b) phages are
more efficient inhibitors of bacterial growth in ethanol
fermenters than phage endolysins, (c) there is
a considerable difference in in vitro phage therapy
outcomes against different species of clinical bacterial
isolates, (d) the outcomes of phage-mediated biocon-
trol in different fields are momentously dissimilar, (e)
MOIs used for ex vivo phage therapy/phage-mediated
biocontrol experiments, in vivo phage therapy and
in vitro phage therapy are soundly similar. A P value
of 0.05 indicated a significant statistical difference.
For comparison of phage therapy and phage-mediated
biocontrol efficacy across the different fields, only
fields that had three or more studies reporting phage
therapy efficacy in percentages were considered for
Tukey’s multiple comparisons test to prevent skewing
of data.
6. Results and discussion
6.1. Literature search
A total of 709 articles were generated through an
electronic database literature search conducted
between January and November 2018. The databases
were PubMed, Lib Hub, and Google Scholar, which
yielded 51, 416, and 242 articles, respectively.
Following the removal of duplications, 204 articles
were screened on the basis of their titles and abstracts.
Of the 204 articles; 90 did not meet the specified
inclusion criteria; and five full-text articles were not
accessible. Finally, 109 full-text articles were reviewed,
of which 95 full-text research articles fulfilled the
inclusion guidelines for this review, Figure 1. Studies
included in this review were grouped into in vivo
human phage therapy, in vivo phage therapy in
model organisms, phages as biocontrol agents in bio-
fuels fermentation, phages as biocontrol agents in
foods and beverages, in vitro phage therapy experi-
ments using clinical isolates, in vivo phage therapy in
crop protection, application of phages as biocontrol
agents in water purification, in vivo phage therapy in
aquaculture, in vivo phage therapy in apiculture,
in vivo phage therapy in a piggery in vivo and
in vitro phage therapy in poultry, application of phages
as bio-sanitizers, and in vitro use of phages as biocon-
trol agents in creams, Table 1–5, Figure 3.
Phage characterization; a prerequisite for phage-
mediated biocontrol of bacterial growth and in vivo
phage therapy
Phage-mediated biocontrol and phage therapy rely
on the ability of lytic phages to infect bacterial host
cells, hijacking the host metabolism and utilizing it to
produce their progeny. As a result, the lytic phages lyse
bacteria cells to release multiple phage virions which
spread to infect other host cells [10]. Contrary to that,
after infecting the bacterial host cells, lysogeny phages
incorporate their genetic material into the host gen-
ome resulting in their permanent existence as pro-
phages within host cells and all their offspring.
Figure 2. Comparison of mean MOIs between in vivo, in vitro, and ex vivo phage therapy. Tukey’s multiple-comparison test was
used to compute and compare MOIs P value of 0.0002 < 0.05 generated indicating significant variation between ex vivo/in vivo PT
and in vitro PT.
10 K. SSEKATAWA ET AL.
Phages neither replicate into virions nor lyse bacteria
throughout their lysogeny life time, hence called tem-
perate phages [10]. Furthermore, the integration of the
phage nucleic acids into its host bacterium protects the
temperate phage genome and has the ability to modify
the phenotype of the host bacterium cell [27].
Unfortunately, temperate phages might harbor toxin
encoding genes, virulent genes, and genetic determi-
nants of antibiotic resistance acquired from other bac-
terial hosts. Therefore, temperate phages may
transform the phenotype of the host bacteria and all
their progeny from avirulent/less virulent and antibio-
tic susceptible strains to highly virulent and antibiotic-
resistant strains [28,29]. Appropriately professionally
isolated and characterized phages must be used to
prevent horizontal gene transfer of undesirable genes
through phage-mediated biocontrol and phage ther-
apy [18,30,31]. Therefore, phages must be character-
ized morphologically by TEM and SDS PAGE protein
profiling to establish their families or if they are novel
phages followed by molecular characterization by
WGS to confirm their families and to detect any inte-
grase, toxin, and virulent genes in addition to antibio-
tic resistance genes by cross-referencing with known
phage genomes, virulent factors, toxin genes, and anti-
biotic-resistant genes libraries. A cheaper but less-
sensitive alternative to detect the presence of known
integrase gene, virulent factors (VF) and genetic deter-
minants of antibiotic resistance in phages is PCR
amplification using conventional integrase gene VF,
toxin genes, and antibiotic resistance genes primers.
However, PCR amplification has limitations as it will
not detect any possible novel VF and antibiotic resis-
tance genes harbored by phages hence making
molecular characterization of phages by WGS
a prerequisite prior to phage-mediated biocontrol of
bacterial growth and in vivo phage therapy [32,33].
However, only 12.6% (12) of the studies included in
this review conducted WGS. Bioinformatics analyses
and annotation demonstrated that myophages BɸC62
[34], DL52, DL60 and DL680 [35], DRA88 and phage
K [36], EcoInf [37], coliphagesɸAPCEc01, ɸAPCEc02
and ɸAPCEc03 [38], Phage P100 [39], leB, leE and leN
[40]:, podophages DL54, DL 62 and DL 64 [35]:, fHe-
Yen3-01 [41] and siphophages EcoSau [37], phSE-1,
phSE-2 and phSE-5 [42], fHe-Yen3-01, fHe-Yen9-01,
fHe-Yen9-02 and fHe-Yen9-03 [41] were safe to use
since they harbored no undesirable genes while
siphophage HB10c2 had a gene encoding a putative
beta-lactamase like protein [43]. Additionally, PCR
detected Stx I and II proteins encoding genes and
lysogeny module genetic determinants in phages
CB60P, MFA60N, CCO103, CBO103, and CCO113
[44], Table S1. If such phages are used in phage ther-
apy and phage-mediated biocontrol, they can facilitate
the horizontal flow of undesirable genes. This exorbi-
tantly underlines the importance of screening phages
using very sensitive tools like WGS. Nevertheless, only
36.8% (39) research articles included in this review
attempted to characterize phages; 3.2% (3) used PCR
to detect VFs and lysogeny modules while only 12.6%
(12) studies carried out WGS to fully illustrate the
phage genomes indicating that there is still a big gap
in ensuring phage therapy safety as per all the
reviewed articles that were in English, though all the
phages used for in vivo human phage therapy were
previously characterized by committed phage research
hubs. Furthermore, the morphology of phages was
Figure 3. Comparison of phage therapy (PT), phage-mediated biocontrol/diagnosis mean efficacy percentages. Tukey’s multiple-
comparison test was used to calculate and compare the mean percentage efficacies generating a P value of 0.148 > 0.05 after
exclusion of fields with less than three studies (water, piggery poultry, and apiculture).
ALEXANDRIA JOURNAL OF MEDICINE 11
determined by transmission electron microscopy
(TEM) in only 24.2% (23) studies. Basing on morphol-
ogy, the phages belonged to various families as follows:
Myoviridae; Siphoviridae; and Podoviridae in twenty,
nine and ten studies respectively. Whereas, one study
in each case reported phages as B1 morphology,
Phage-like particle, and Cytoviridae family, Table S1.
7. Phage stability
Establishing the abiotic conditions affecting phage activ-
ity and/or viability was done in 16.8% (16) studies. This is
an important criterion for selection since phage viability,
occurrence, and storage are affected by temperature, pH,
humidity, salinity, and other environmental conditions.
Deviation from the favorable physicochemical factors can
lead to the destruction of phages’ structural elements,
protein envelope, and loss of genetic material thereby
inactivating the phages [45,46]. These phages are isolated
from natural environments such as sewage, hospital, and
animal farm effluents, water bodies, foods, and beverages
and evaluated for in vitro, in vivo, and ex vivo phage
therapy and phage-mediated biocontrol where the pre-
vailing physicochemical factors are completely different,
Table 1–5. Hence, the need to establish the optimum
conditions for the highest phage efficacy. However, such
drawbacks can be mitigated by isolation of phages from
local geographical locations and similar hosts as for
in vivo phage therapy accompanied by assessing phage
stability via exposing them to different physicochemical
factors. Furthermore, during the preparation of commer-
cial phage-based remedy, physicochemical properties are
supposed to be investigated as they determine the shelf-
life of phages [47]. Despite that concern, only 9 (9.5%)
and 7 (7.4%) out of 95 research articles included in this
review evaluated the thermal and pH stability of phages,
respectively, Table S1. This partly explains why some
research articles reported very low or 0% phage efficacy
in in vivo studies.
8. Specicity of phages
Specificity restricts phage infections to only certain bac-
teria with corresponding receptors to which they can
bind; this determines the phage’s host range [48]. For
that reason, the application of phage therapy relies on an
accurate characterization of all the strains, pathotypes,
and serotypes of the target bacteria. Interestingly, if
phage therapy overcomes the current obstacles hindering
its approval universally, single phage and phage cocktail
formulations must be designed indicating the pharma-
ceutical dosage and the phage host range for a given
bacteria which calls for robust characterization of given
target host bacteria. Conversely, this review identified
gross deviation from the recommended procedure if
meaningful phage therapy outcomes are to be achieved
as only 55.8% (53) of studies reviewed attempted to use
identified bacterial host strains, serovars, and pathotypes,
Table 1–5. Worst still, no human in vivo phage therapy
trial reported characterization of the target bacteria to
their strains, pathotypes, and serotypes. Nevertheless, the
spectrum and efficacy of phages can be enhanced by the
use of phage cocktails. Phage cocktails also present
another advantage of preventing phage resistance [49,50].
9. Multiplicity of infection (MOI)
MOI is defined as PFU/CFU ratio [51]. MOI is an
imperative factor to be considered for prospective
phage therapy application. Increasing the PFU/CFU
ratio enhances the probability of phage particles
infecting their host bacteria. Therefore, in vivo and
ex vivo phage therapies require higher MOIs than
in vitro phage therapy as it is harder for phages to
locate and infect their hosts within living tissues, sur-
face of foods, and other materials being infected by
phages. Some studies recommend an MOI of over 100
for ex vivo and in vivo phage therapy and less than 10
for in vitro phage therapy [52]. This is in agreement
with the studies incorporated in this review that
reported MOI. The average MOI was highest in ex
vivo experiments (557,291.8), followed by in vivo
phage therapy (155,612.4) and in vitro phage biocon-
trol experiments had the lowest average MOI of 434.5
significantly different from ex vivo and in vivo MOIs,
Table S1 and Figure 2. Contrary to this, other studies
disregard the term MOI as it only describes the phage
quantities administered during dosing in relation to
the population of the target bacteria but does not put
into consideration the fact that; some phages fail to
penetrate tissues/materials and get inactivated before
adsorbing to the host cells, the host cell population is
liable to change before phage application, the bacterial
population may not easily be determined in case of
infections and physicochemical factors such as tem-
perature, pH, salinity, and humidity may inactivate
phages before adsorption. As a result, MOI input
may differ from the actual effective MOI [53].
Furthermore, to increase the prospect of phages
adhering and infecting their hosts; for experimental-
induced infections a very high MOI of >10
5
is recom-
mended [54] whereas in vivo phage therapy of natural
infection a very high titer value of > 1 × 10
8
PFU is
appropriate as bacterial host cells are lysed by simply
adsorption of phages before injection of their nucleic
acids into the host cells and replication [52,54].
However, phages are immunogenic when applied at
very high doses [55]; therefore, the host immune sys-
tem may identify and inactivate them. Additionally,
the MOI against biofilm infections should be higher as
indicated by the studies reviewed which compared
optimum MOI against bacterial suspension or free-
living bacteria to that against biofilms and/or immo-
bile bacteria, Table S1 and Figure 2. In in vitro
12 K. SSEKATAWA ET AL.
experiments, MOIs of 0.1, 1, and 10; and 100, 1,000,
and 10,000 [56]; 0.1 and 10 [36] were administered
against bacterial planktons and biofilms, respectively.
It is worth mentioning that in addition to high MOI,
the most suitable phages for phage-mediated manage-
ment of biofilm infections should encode polysacchar-
ide depolymerase which degrades the biofilm
polysaccharide matrix to ease phage interaction with
the host cells in the lower layers of the matrix [57].
10. Ecacy of phage therapy against drug
resistant and sensitive bacterial infections and
isolates
In vivo human phage therapy studies reported mixed
levels of efficacy ranging from 0% to 100%. The mode
and median efficacies were 100% while Tukey’s multiple
comparison test generated a P value of 0.009 < 0.05 indi-
cating that phage therapy efficacies of 100% were more
pronounced than efficacies lower than 100% in all the
in vivo human phage therapy. Interestingly, efficacies of
100% were scored when treating MRSA diabetic foot
ulcers, GIT MRSA infection, VRSA corneal abscess and
interstitial keratitis, and MDR Pseudomonas aeruginosa
UTI with phages. Furthermore, in vivo phage treatment
of MRSA osteomyelitis in Rabbits [58], carbepenem resis-
tant Acinetobacter baumanii infection in mice [34], MDR
Acinetobacter baumanii pneumonia in mice [59] and pan
drug resistant (PDR) Acinetobacter. baumannii infec-
tions in mice [60] provided 100% protection to model
animals against the super bugs while in vivo phage ther-
apy of MDR Pseudomonas aeruginosa ulcerative lesions
in catfish species achieved 100% success. It is also worth
noting that ex vivo phage therapy against MDR
Pseudomonas aeruginosa skin infections, MRSA biofilms
induced onto porcine skin burns, and PDR Acinetobacter
baumanni human HeLa cells infections recorded over-
whelming success. In vitro phage therapy against MRSA,
MDR Acinetobacter baumanni, MDR Pseudomonas aer-
uginosa scored an inhibitory efficacy ranging from 78% to
100% with an average of 95.4%. Data from around the
globe show an overall decline in the total reserves of
antibiotics efficacy: resistance to all first-line and last-
resort antibiotics is increasing [3]. For instance, in sub-
Saharan Africa, India, Latin America, and Australia,
MRSA incidence is still intensifying [3,6162], and esti-
mated at 47% in India in 2014, and 90% in Latin
American hospitals in 2013 [61][]. MRSA causes
35–46% of wound complication in Mulago referral hos-
pital [63,64]. The increased prevalence of community
acquired E. coli isolates coding for extended-spectrum
beta lactamases competent of hydrolyzing approximately
all beta lactams antibiotic except carbapenems has been
reported globally [65]. In more than a decade, carbape-
nem resistance in Enterobacteriaceae bacteria has been
observed yet Carbapenems such as imipenem, ertape-
nem, meropenem, and doripenem are the newest
synthesized molecules with the broadest spectrum of
activity and consequently considered the first-line ther-
apy antibiotics in the treatment of multi-resistant gram-
negative bacterial infections [66,67]. The magnitude of
MDR Pseudomonas aeruginosa and Acinetobacter bau-
mannii is a great threat to the health sector worldwide
[68]. The promising outcomes of in vivo, ex vivo, and
in vitro phage therapy of MDR bacterial infections and
isolates exhibit that phage therapy if employed appropri-
ately is more effective than antibiotics and therefore can
replace or supplement antibiotics as a routine in the
management of both resistant and sensitive bacterial
infections. However, limited success was attained when
treating S. aureus and P. aeruginosa wound infection in
humans, acute human E. coli infections, MRSA nasal
infections in pigs and American foulbrood caused by
Paenibacillus larvae [43,69–71]. This is in contrary to
the in vitro experiments carried out in two of the studies
where total eradication of the bacteria was achieved
[43,71]. This can be attributed to the change in physiolo-
gical conditions: loss of phage viability due to deviation
from their optimum temperature and pH in unnatural
environments [46].
11. Endolysins versus phage particles
Phages code tail spike proteins for identification and
adhesion to receptors on the host cell surface. The tail
spikes proteins are often incorporated with peptidoglycan
hydrolases that locally hydrolyze the bacterial cell wall
peptidoglycan, thus creating an opening for injection of
phage nucleic acids which marks the initiation of the
infection process [72]. An additional type of phage-
derived enzymes; the peptidoglycan hydrolases called
endolysins degrade the peptidoglycan liberating the pro-
geny virions from the host cell at the end of the lytic phage
cycle [73]. Gram-positive bacteria do not possess
a shielding outer layer thereby making exogenous appli-
cation of endolysins achieve speedy and effective lysis.
This property makes endolysins promising possible alter-
native antimicrobial agents [23]. Several studies have
reported endolysins as potential therapeutic agents with
high efficacy and safety [74]. In addition, endolysins
possess an added advantage over conventional antibiotics
as; they exhibit great specificity exerting selective pressure
on target pathogenic bacteria populations [75,76], emer-
gence of resistance against endolysins is implausible given
that phage (endolysins) coevolve with their host bacteria,
the host receptor site where endolysins bind are highly
conserved thereby making their alteration highly detri-
mental to the host bacterium [76,77]. Furthermore, endo-
lysins degrade the cell wall externally without the burden
of entering the bacterial cell hence evading the common
antibiotic resistance mechanisms such as the active efflux
pump and decreased membrane permeability [78]. A lot
of ethical and safety concerns have been vehemently
expressed about the use of live viruses as therapeutics in
ALEXANDRIA JOURNAL OF MEDICINE 13
the treatment of bacterial infections; currently, the
immediate hope lies in the use of phage endolysins in
the near future to combat the increasing antibiotic resis-
tance. Fortunately, to meet the high demand, endolysins
can be produced using recombinant DNA technology
[79–81]. This review compared the use of phages and
endolysins to suppress bacterial growth during ethanol
fermentation. Phages demonstrated superior efficacy
than recombinant phage endolysins with mean efficacy
of 99.5% for phages and 83.6% for phage endolysins but
not significantly divergent as revealed by one-way
ANOVA (P value of 0.13 > 0.05). This clearly supports
the use of phages and endolysins hand in hand as ther-
apeutic agents.
12. Application of phages in Biosanitization
and Biopreservation
Infectious food and water-associated diseases are the
major causes of mortality and morbidity worldwide
[6,7]. Irrational use of antibiotics in livestock has resulted
in antibiotic resistance which spillover to humans
through contaminated food, water, and environment [-
3–5,67]. Fortunately, in 2006 the US Food and Drug
Administration (FDA) approved the utilization of 6 inde-
pendently purified LMP-102 phages as biopreservative
antimicrobial agents in RTE meat and poultry products
against Listeria monocytogenes [82]. In this review, the
literature search yielded 21 (22.1%) research articles
reporting foods and beverages phage-mediated bio-
preservation with average, mode, median efficacy of
96.5%, 100%, and 100%, respectively. In a water deconta-
mination study, phages eradicated 95.4% of the coliform.
This is a clear indicator of the potency of phages as bio-
preservative and bio-decontamination agents and conse-
quently their approval to preserve food and decontami-
nate water following robust characterization should be
considered to prevent transmission of antibiotic-resistant
and susceptible food and water-associated infection.
Furthermore, the hospital environment polluted by
infected patients with antibiotic-resistant bacteria is
incriminated as the main route of transmission
[83,84]. This has been a result of the emergence of
bacterial resistance to the conventional disinfectants
[83]. The possibility of a horizontal flow of mobile
genetic elements encoding antibiotic resistance from
clinical to environmental bacteria within the hospital
is high hence advancing the evolution of new antibiotic-
resistant bacterial strains [85]. On a good note, bio-
disinfection using phages as demonstrated by this
review is promising: for instance, phage-mediated bio-
sanitization eradicated 90% of Staphylococcus aureus,
Escherichia coli, and Pseudomonas aeruginosa tainted
on plastic, glass, and ceramic materials mimicking hos-
pital surfaces [86] while phage-mediated sterilization
trial of the ICU reduced the prevalence of carbapenem-
resistant Acinetobacter baumanii by 47.5% [87]. In
another phage sanitization trial, phages completely
eliminated staphylococci, streptococci, enterococci, pro-
teus, Klebsiella pneumoniae, Klebsiella oxytoca,
Pseudomonas aeruginosa, and Escherichia coli from the
hospital environment [88] while phage-based sanitiza-
tion cream completely inhibited MRSA and
Propionibacterium acnes growth [89,90]. With those
laudable bio-sanitization results, the use of phages to
complement conventional disinfection strategies could
exhibit valuable outcomes.
13. Phages and endolysin as alternative
antibacterial decontamination agents
Lactic acid bacteria (LAB) are by far the commonest
bacterial contaminants of biofuel production facilities
and are believed to hamper the ethanol fermentation
process hence limiting ethanol production. Ethanol fer-
mentation presents an environment of high ethanol con-
centration, low pH, and low oxygen concentration
thereby favoring the growth of Lactobacillus sp which
are well adapted to survive under such conditions.
Currently, there is no appropriate strategy to combat
ethanol loss due to LAB contamination as all possible
measures have limitations [91]. Contrary to that, the four
experimental studies which employed phages and endo-
lysins to control LAB growth during ethanol fermenta-
tion analyzed in this review demonstrated eye-catching
bacterial growth suppression outcomes with mean effi-
cacy of 91.6%. Most importantly, phage and endolysins
mediated ethanol fermentation facility decontamination
restored normal ethanol yield without losing their viabi-
lity [37,92]. Because of the promising results, to eliminate
the use of antibiotics for decontamination in the ethanol
fermentation business, phages and endolysins should be
considered as alternatives.
14. Limitations
Hypothetically, all bacteria can be lysed by at least one
type of bacteriophage. In the light of this, phages are
considerably more efficacious than antibiotics.
However, phage antibacterial applications have limita-
tions. Most phages have demonstrated a broad spectrum
hence can lyse both the target pathogenic strains and
potentially beneficial bacterial strains. Additionally, it is
difficult to isolate phages without any undesirable genes
such as antibiotic-resistant genes, bacterial virulent genes,
and integrase genes. Phages with such genes may con-
tribute to the development of highly pathogenic antimi-
crobial-resistant bacteria. Furthermore, phage-based
therapeutic formulation and stabilization is still
a challenge as previous studies reported that the stability
of phage formulations for clinical use is stringently influ-
enced by the phage type. Thus, each phage type requires
its unique stabilization strategy and this is extremely
complicated for phage cocktail formulations. The
14 K. SSEKATAWA ET AL.
evolution of bacterial resistance against phages mainly
mediated by loss or alteration of the bacterial phage
receptors and bacterial secretions that prevent phage
adsorption has been implicated as another limitation
affecting phage therapy. Inactivation of phages by the
immune system has also been reported as a drawback of
phage therapy.
15. Conclusion
The high prevalence of MDR infections has resulted in
familiar bacterial diseases becoming difficult to treat.
Moreover, hospital-associated infections (both sensitive
and MDR) are mainly acquired through contaminated
surfaces and medical equipment. However, phage-
mediated bio-sanitization, in vivo, ex vivo, and in vitro
phage therapy experiments and trials analyzed by this
review showed that phages can mitigate the burden
caused by MDR infections and contamination of hospital
surfaces as well as medical devices. Furthermore, water
and food-borne bacterial infections have been implicated
as the major cause of mortality and morbidity globally
and LAB as the main cause of yield loss in the biofuels
fermentation industry. Analysis of phage/endolysin
mediated bio-preservation and bio-decontamination stu-
dies by this review showed that phages and endolysins
were highly effective. Thus, phage technology presents an
opportunity for developing alternative therapeutic, bio-
preservative, bio-decontamination, and bio-sanitization
approaches. Despite the undisputable efficacy of phage
therapy and phage-mediated biocontrol, rigorous inves-
tigations using highly sensitive techniques should be car-
ried out to ensure that only appropriate professionally
lytic and safe phages are used. Thus, for low- and middle-
income countries, there is a need to develop affordable
and appropriate methods for screening of phages for
undesirable genes. Moreover, the challenge of immuno-
genicity that may be associated with in vivo application of
phages needs to be explored further.
Acknowledgements
We are thankful to MAPRONANO ACE for funding this work.
Disclosure statement
The authors declare that they have no competing interests.
Funding
The authors declare that this review article was funded by
Africa Centre of Excellence in Materials, Product
Development & Nanotechnology; MAPRONANO ACE
Makerere University;African Center of Excellence in
Materials, Product Development and Nanotechnology,
College of Engineering Design, Art and Technology
Makerere University [P151847IDA credit 5797-UG].
Notes on contributors
Kenneth Ssekatawa is a PhD candidate at Makerere
University and Lecturer Department of Biochemistry,
Kampala International University. Kenneth specializes in
molecular microbiology, antimicrobial resistance and
nanobiotechnology.
Prof. Denis K. Byarugaba He is a Professor of Microbiology,
College of Veterianary Medicine Animal Resources and
Biosecurity, Makerere University. He is currently involved
in research on influenza viruses and highly pathogenic
pathogens with potential for causing pandemic threats and
antimicrobial resistance.
Dr. Charles D. Kato is a lecturer at college of Veterinary
Medicine Animal Resources and Biosecurity. Charles'
research interests are in biomark research regarding disease
co-infection and host-parasite interactions in infectious and
zoonotic diseases. Charles is conducting research in phage
mediated therapy.
Dr. Eddie M. Wampande currently works as a senior lec-
turer College Veterinary Animal Resources and Biosecurity,
Makerere University. He specializes in Microbiology and
Parasitology.
Prof. Francis Ejobi is a Professor of Microbiology, Makerere
University specializing in infectious disease, epidemiology
and antimicrobial resistance
Prof. Robert Tweyongyere Robert is a research fellow at the
Uganda Virus Research Institute, Entebbe and a Senior
Lecturer at College of Veterinary Medicine Animal
Resources and Biosecurity Makerere University. His
research interests are in infectious diseases.
Dr. Jesca L. Nakavuma Jesca works as a senior lecturer and
microbiologist, Department of Biomolecular and
Biolaboratory Sciences, College of Veterinary Medicine
Animal Resources and Biosecurity, Makerere University.
She is the Founder Chair Phage Team Uganda and currently
working on a project aimed at using phage therapy to con-
trol and treat bacterial infections in fish.
ORCID
Kenneth Ssekatawa http://orcid.org/0000-0003-4061-
9345
Ethics and consent to participate:
Not applicable
Consent for publication:
Not applicable
Availability of Data and Materials:
Supplementary data summarized in tabular form have been
submitted with the manuscript.
Authors’ contributions
This work was carried out in collaboration between all
authors. Jesca L. Nakavuma (JLN), Dennis K Byarugaba
ALEXANDRIA JOURNAL OF MEDICINE 15
(DKB), Robert Tweyongyere (RB), and Francis Ejobi (FB)
conceptualized this project and designed the format for this
review. Kenneth Ssekatawa (KS), Edward Wampande (EW),
Charles Kato Drago (CKD) & JLN performed the literature
search and data analysis. All authors drafted the section of
the literature review. KS, JLN, and CKD wrote the first draft
of the manuscript and managed manuscript revisions. All
authors read and approved the final manuscript.
References
[1] CDC, Facility Guidance for Control of
Carbapenem-resistant Enterobacteriaceae (CRE) 2015.
[2] UNAS. Antibiotic resistance in Uganda: situation
analysis and recommendations. Uganda National
Academy of Science; 2015.
[3] Centre for Disease Dynamics Economics and Policy-
CDDEP. The state of the world’s antibiotics.
Washington DC-New Delhi; 2015.
[4] Economou V, Gousia P. Agriculture and food animals
as a source of antimicrobial-resistant bacteria. J Infec
drug resist. 2015;8: 49.
[5] Warnes SL, Highmore CJ, Keevil CW. Horizontal trans-
fer of antibiotic resistance genes on abiotic touch sur-
faces: implications for public health. 2012 3(6); J MBio.
[6] World Health Organization., World health organization
estimates of the global burden of foodborne diseases.
WHO, Geneva, Switzerland (2015). http://apps.who.int
[7] Lewin S, et al. Estimating the burden of disease attri-
butable to unsafe water and lack of sanitation and
hygiene in South Africa in 2000. S. Afr Med Journal;
2007;97(8):755–762.
[8] Fenwick AJS. Waterborne infectious diseases—could
they be consigned to history? 2006;313
(5790):1077–1081.
[9] Shlaes DM. The perfect storm, in antibiotics. The
Netherlands: Springer; 2010. p. 1–7.
[10] Guttman B, Raya R, Kutter EJBBA. Basic phage
biology. 4 (2005).
[11] Abedon S. Bacteriophage clinical use as antibacterial
“drugs”: utility and precedent. Microbiol. Spectr.
2017;5:BAD-0003-2016.
[12] Kutateladze M, Adamia R. Bacteriophages as poten-
tial new therapeutics to replace or supplement anti-
biotics. G. Eliava Institute of Bacteriophages. Trends
Biotechnol. 2010;28(12):591–595.
[13] Silva YJ, et al. Phage therapy as an approach to pre-
vent Vibrio anguillarum infections in fish larvae
production. 2014;9(12):e114197.
[14] Cortés ME, Bonilla JC, Sinisterra RD. Biofilm forma-
tion, control and novel strategies for eradication.
2011;2:896–905. %J Sci Against Microbial Pathog
Commun Curr Res Technol Adv.
[15] Abedon S. Bacteriophages and biofilms: ecology.
2011;557:J Phage Therapy Plaques.
[16] Hughes KA, Sutherland IW, Jones M. Biofilm sus-
ceptibility to bacteriophage attack: the role of
phage-borne polysaccharide depolymerase. 1998;144
(11):3039–3047. V %J Microbio.
[17] Wittebole X, De Roock S, Opal S. A historical over-
view of bacteriophage therapy as an alternative to
antibiotics for the treatment of bacterial pathogens.
M %J Virulence. 2014;5(1):226–235.
[18] Borysowski J, Międzybrodzki R, Górsk A. Phage ther-
apy: current research and applications. Caister
Academic Press; 2014.
[19] Chan BK, et al. Phage treatment of an aortic graft
infected with Pseudomonas aeruginosa. 2018;2018
(1):60–66.
[20] Kazi M, Annapure US. Bacteriophage biocontrol of
foodborne pathogens. J food sci tech. 2016;53
(3):1355–1362.
[21] Roy B, et al. Biological inactivation of adhering Listeria
monocytogenes by listeriaphages and a quaternary
ammonium compound. 1993;59(9):2914–2917.
[22] Greer G. Effects of phage concentration, bacterial
density, and temperature on phage control of beef
spoilage. J Food Sci. 1988;53(4):1226–1227.
[23] Fischetti VA. Bacteriophage endolysins: a novel
anti-infective to control Gram-positive pathogens.
Int J Med Microbiol. 2010;300(6):357–362.
[24] Le TS, et al. Bacteriophages as biological control
agents of enteric bacteria contaminating edible
oysters. 2018;75(5):611–619.
[25] Hudson J, et al. Bacteriophages as biocontrol agents
in food. 2005;68(2):426–437.
[26] Chatain-LY MH. The factors affecting effectiveness of
treatment in phages therapy. 2014;5:51. J Frontiers
microbio.
[27] Gill J, Abedon STJAF. Bacteriophage ecology and
plants. 2003;1–17.
[28] Colomer-Lluch M, Jofre J, Muniesa M. Antibiotic resis-
tance genes in the bacteriophage DNA fraction of
environmental samples. PLoS One. 2011;6(3):e17549.
[29] Faruque S, Mekalanos J. Phage-bacterial interactions
in the evolution of toxigenic Vibrio cholerae.
Virulence. 2012;3(7):556–565.
[30] McCallin S, et al. Safety analysis of a Russian
phage cocktail: from metagenomic analysis to oral
application in healthy human subjects. 2013;443
(2):187–196.
[31] Gill JJ, Hyman PJCPB. Phage choice, isolation, and
preparation for phage therapy. 2010;11(1):2–14.
[32] Hagens S, Loessner MJJCPB. Bacteriophage for bio-
control of foodborne pathogens: calculations and
considerations. 2010;11(1):58–68.
[33] Mahony J, et al. Bacteriophages as biocontrol agents
of food pathogens. 2011;22(2):157–163.
[34] Jeon J, et al. In vivo application of bacteriophage as
a potential therapeutic agent to control OXA-66-like
carbapenemase-producing Acinetobacter baumannii
strains belonging to sequence type 357. 2016;82
(14):4200–4208.
[35] Alves DR, et al. A novel bacteriophage cocktail
reduces and disperses P seudomonas aeruginosa bio-
films under static and flow conditions. J Microbial
biotech. 2016;9(1):61–74.
[36] Alves D, et al. Combined use of bacteriophage K and
a novel bacteriophage to reduce Staphylococcus aur-
eus biofilm formation. J App Environ Microbio.
2014;80(21):6694–6703.
[37] Liu M, et al. Bacteriophage application restores etha-
nol fermentation characteristics disrupted by
Lactobacillusfermentum. 2015;8(1):132.
[38] Dalmasso M, Strain R, Neve H, et al. Three new
Escherichia coli phages from the human gut show
promising potential for phage therapy. PLoS One.
2016;11(6):e0156773. .
[39] Carlton RM, Noordman WH, Biswas B, et al.
Bacteriophage P100 for control of Listeria monocyto-
genes in foods: genome sequence, bioinformatic ana-
lyses, oral toxicity study, and application. Regul
Toxicol Pharmacol. 2005;43(3):301–312. .
16 K. SSEKATAWA ET AL.
[40] Endersen L, Buttimer C, Nevin E, et al. Investigating
the biocontrol and anti-biofilm potential of a three
phage cocktail against Cronobacter sakazakii in dif-
ferent brands of infant formula. Int J Food Microbiol.
2017;253:1–11.
[41] Jun JW, et al. Bacteriophages reduce Yersinia enter-
ocolitica contamination of food and kitchenware.
2018;271:33–47.
[42] Pereira C, et al. Bacteriophages with potential to
inactivate Salmonella Typhimurium: use of single
phage suspensions and phage cocktails.
2016;220:179–192.
[43] Beims H, et al. Paenibacillus larvae-directed bacter-
iophage HB10c2 and its application in American
foulbrood-affected honey bee larvae. 2015;81
(16):5411–5419.
[44] Dini C, De Urraza PJ. Isolation and selection of coli-
phages as potential biocontrol agents of enterohemor-
rhagic and Shiga toxin-producing E. coli (EHEC and
STEC) in cattle. J Appl Microbiol. 2010;109
(3):873–887.
[45] Ackermann H-W, Tremblay D, Moineau S. Long-
term bacteriophage preservation. 2004;38:35e40.
WFCC Newsletters.
[46] Jończyk E, et al. The influence of external factors on
bacteriophages. 2011;56(3):191–200.
[47] Vandenheuvel D, Lavigne R, Brüssow H.
Bacteriophage therapy: advances in formulation stra-
tegies and human clinical trials. Annu Rev Virol.
2015;2(1):599–618.
[48] Hyman P, Abedon ST. Bacteriophage host range and
bacterial resistance, in advances in applied microbiol-
ogy. Elsevier; 2010. p. 217–248.
[49] Markoishvili K, et al. A novel sustained-release matrix
based on biodegradable poly (ester amide) s and
impregnated with bacteriophages and an antibiotic
shows promise in management of infected venous
stasis ulcers and other poorly healing wounds.
2002;41(7):453–458.
[50] Jikia D, et al. The use of a novel biodegradable pre-
paration capable of the sustained release of bacterio-
phages and ciprofloxacin, in the complex treatment of
multidrug-resistant Staphylococcus aureus-infected
local radiation injuries caused by exposure to Sr90.
2005;30(1):23–26.
[51] O’flynn G, et al. Evaluation of a cocktail of three
bacteriophages for biocontrol of Escherichia coli
O157: H7. J Applied Environ Microbio. 2004;70
(6):3417–3424.
[52] Johnson R, et al. Bacteriophages for prophylaxis and
therapy in cattle, poultry and pigs. 2008;9(2):201.
[53] Abedon S. Phage therapy dosing: the problem(s) with
multiplicity of infection (MOI). Bacteriophage. 2016;6
(3):e1220348.
[54] Bach SJ, et al. Effect of bacteriophage DC22 on
Escherichia coli O157: H7 in an artificial rumen sys-
tem (Rusitec) and inoculated sheep. 2003;52
(2):89–101.
[55] Majewska J, et al. Oral application of T4 phage
induces weak antibody production in the gut and in
the blood. 2015;7(8):4783–4799.
[56] Chen L-K, et al. Potential of bacteriophage ΦAB2 as
an environmental biocontrol agent for the control of
multidrug-resistant Acinetobacter baumannii.
2013;13(1):1–10.
[57] Sutherland IW, et al. The interaction of phage and
biofilms. 2004;232(1):1–6.
[58] Kishor C, et al. Phage therapy of staphylococcal
chronic osteomyelitis in experimental animal model.
Indian J Med Res. 2016;143(1):87–94. .
[59] Wang Y, et al. Intranasal treatment with bacterioph-
age rescues mice from Acinetobacter
baumannii-mediated pneumonia. 2016;11
(5):631–641.
[60] Yin S, Huang G, Zhang Y, et al. Phage Abp1 rescues
human cells and mice from infection by pan-drug
resistant Acinetobacter Baumannii. Cell Physiol
Biochem. 2017;44(6):2337–2345. .
[61] EARS-NET., European antimicrobial resistance sur-
veillance network (EARS-Net). 2014. EARS-Net
Report,Quarters 1–4. Dublin. 2014. This is a perma-
nent publication in PDF, thus access date not require
[62] Kateete DP, et al. High prevalence of methicillin
resistant Staphylococcus aureus in the surgical units
of Mulago hospital in Kampala, Uganda. BMC Res
Notes. 2011;4(1):326.
[63] Seni J, et al. Molecular characterization of
Staphylococcus aureus from patients with surgical
site infections at Mulago Hospital in Kampala,
Uganda. PLoS One. 2013;8(6):e66153.
[64] Pitout JD, Laupland KB. Extended-spectrum
beta-lactamase-producing Enterobacteriaceae: an
emerging public-health concern. Lancet Infect Dis.
2008;8(3):159–166.
[65] EFSA Panel on Biological Hazards. Scientific Opinion
on Carbapenem resistance in food animal ecosystems.
J EFSA. 2013;11(12):3501.
[66] Nordmann P, Dortet L, Poirel LJTIMM. Carbapenem
resistance in Enterobacteriaceae: here is the storm!
2012;18(5):263–272.
[67] CDC, Antibiotic resistance threats in the United States.
2013.
[68] Rose T, et al. Experimental phage therapy of burn
wound infection: difficult first steps. 2014;4(2):66.
[69] Sarker S, et al. Oral phage therapy of acute bacterial
diarrhea with two coliphage preparations:
a randomized trial in children from Bangladesh.
2016;4:124–137.
[70] Verstappen KM, et al. The effectiveness of bacterio-
phages against methicillin-resistant Staphylococcus
aureus ST398 nasal colonization in pigs. PLoS One.
2016;11(8):e0160242.
[71] Rodríguez-Rubio L, et al. Bacteriophage
virion-associated peptidoglycan hydrolases: potential
new enzybiotics. Crit Rev Microbiol. 2013;39
(4):427–434. .
[72] Young R. Phage lysis: do we have the hole story yet?
Curr Opin Microbiol. 2013;16(6):790–797.
[73] Haddad Kashani H, et al. Recombinant endolysins as
potential therapeutics against antibiotic-resistant
<span class=“named-content genus-species”
id=“named-content-1”>Staphylococcus aureus</
span>: current status of research and novel delivery
strategies. 2018;31(1):e00071–17.
[74] García P, Rodríguez L, Rodríguez A, et al. Food bio-
preservation: promising strategies using bacteriocins,
bacteriophages and endolysins. Trends Food
SciTechnol. 2010;21(8):373–382. .
[75] Borysowski J, Weber-Dąbrowska B, Górski A, et al.
Bacteriophage endolysins as a novel class of antibac-
terial agents. Exp Biol Med (Maywood). 2006;231
(4):366–377. .
[76] Kusuma C, Jadanova A, Chanturiya T, et al.
Lysostaphin-resistant variants of Staphylococcus
ALEXANDRIA JOURNAL OF MEDICINE 17
aureus demonstrate reduced fitness in vitro and in
vivo. Antimicrob Agents Chemother. 2007;51
(2):475–482. .
[77] Viertel T, Ritter K, Horz HP. Viruses versus
bacteria-novel approaches to phage therapy as a tool
against multidrug-resistant pathogens. J Antimicrob
Chemother. 2014;69(9):2326–2336.
[78] Mao J, et al. Chimeric Ply187 endolysin kills
Staphylococcus aureus more effectively than the par-
ental enzyme. 2013;342(1):30–36.
[79] Yang H, Yu J, Wei H. Engineered bacteriophage
lysins as novel anti-infectives. 5(542):2014.
[80] Donovan D, Foster-Frey J. LambdaSa2 prophage endo-
lysin requires Cpl-7-binding domains and amidase-5
domain for antimicrobial lysis of streptococci. FEMS
Microbiol Lett. 2008;287(1):22–33.
[81] Lang LH. FDA approves use of bacteriophages to be
added to meat and poultry products. Gastroenterology.
2006;131(5):1370.
[82] de Oliveira AC, Damasceno QS. [Surfaces of the hos-
pital environment as possible deposits of resistant
bacteria: a review]. Rev Esc Enferm USP. 2010;44
(4):1118–1123.
[83] French GL, et al. Tackling contamination of the hos-
pital environment by methicillin-resistant
Staphylococcus aureus (MRSA): a comparison
between conventional terminal cleaning and hydro-
gen peroxide vapour decontamination. J Hosp Infect.
2004;57(1):31–37. .
[84] Ssekatawa K, et al. A systematic review: the current
status of carbapenem resistance in East Africa. BMC
Res Notes. 2018;11(1):629. .
[85] D’Accolti M, et al. Efficient removal of hospital patho-
gens from hard surfaces by a combined use of bacter-
iophages and probiotics: potential as sanitizing
agents. 2018;11:1015.
[86] Ho YH, et al. Application of bacteriophage-containing
aerosol against nosocomial transmission of
carbapenem-resistant acinetobacter baumannii in an
intensive care unit. PLoS One. 2016;11(12):e0168380. .
[87] Akimkin V, Shestopalov N, Shumilov V, et al. ICPIC,
meeting abstracts from international conference on
prevention & infection control (ICPIC 2017), biolo-
gical disinfection with bacteriophages. Antimicrob
Resist Infect Control. 2017;6(3):52.
[88] O’flaherty S, et al. Potential of the polyvalent
anti-Staphylococcus bacteriophage K for control of
antibiotic-resistant staphylococci from hospitals.
2005;71(4):1836–1842.
[89] Brown TL, et al. The formulation of bacteriophage in
a semi solid preparation for control of propionibacter-
ium acnes growth. PLoS One. 2016;11(3):e0151184. .
[90] Beckner M, Ivey ML, Phister TG. Microbial contam-
ination of fuel ethanol fermentations. 2011;53
(4):387–394.
[91] Silva J, Sauvageau D. Bacteriophages as antimicrobial
agents against bacterial contaminants in yeast fer-
mentation processes. J Biotechn biofuels. 2014;7
(1):123.
[92] Fish R, et al. Bacteriophage treatment of intransigent
diabetic toe ulcers: a case series. 2016;25(Sup7):S27–
S33.
[93] Leszczyński P, et al. Successful eradication of methi-
cillin-resistant Staphylococcus aureus (MRSA) intest-
inal carrier status in a healthcare worker–case report.
Folia Microbiol (Praha). 2006;51(3):236–238. .
[94] Marza JAS, et al. Multiplication of therapeutically
administered bacteriophages in Pseudomonas aerugi-
nosa infected patients. Burns. 2006;32(5):644–646. .
[95] Fadlallah A, Chelala E, Legeais J-MJTOOJ. Corneal
infection therapy with topical bacteriophage
administration. 2015;9:167.
[96] Wright A, et al. A controlled clinical trial of
a therapeutic bacteriophage preparation in chronic
otitis due to antibiotic-resistant Pseudomonas aeru-
ginosa; a preliminary report of efficacy. 2009;34
(4):349–357.
[97] Khawaldeh A, et al. Bacteriophage therapy for refrac-
tory Pseudomonas aeruginosa urinary tract infection.
J Med Microbiol. 2011;60(Pt 11):1697–1700. .
[98] Letkiewicz S, et al. Eradication of Enterococcus fae-
calis by phage therapy in chronic bacterial prostatitis
—case report. 2009;54(5):457.
[99] Roach DR, et al. Bacteriophage-encoded lytic enzymes
control growth of contaminating Lactobacillus found
in fuel ethanol fermentations. 2013;6(1):20.
[100] Khatibi PA, et al. Saccharomyces cerevisiae
expressing bacteriophage endolysins reduce
Lactobacillus contamination during fermentation.
2014;7(1):104.
[101] Obeso JM, et al. Use of logistic regression for predic-
tion of the fate of Staphylococcus aureus in pasteur-
ized milk in the presence of two lytic phages. Appl
Environ Microbiol. 2010;76(18):6038–6046. .
[102] Lee Y-D, Park J-H. Characterization and application
of phages isolated from sewage for reduction of
Escherichia coli O157:H7 in biofilm. Lebensmittel-
Wissenschaft + [i.e. und] Tech. 2015;60(no. 1):571–-
577–2015.
[103] Tomat D, Mercanti D, Balagué C, et al. Phage biocon-
trol of enteropathogenic and Shiga toxin-producing
Escherichia coli during milk fermentation. Lett Appl
Microbiol. 2013;57(1):3–10. .
[104] Guenther S, Loessner MJ. Bacteriophage biocontrol of
Listeria monocytogenes on soft ripened white mold
and red-smear cheeses. J App Bacteri. 2011;1
(2):94–100.
[105] Vonasek EL, et al. Incorporating phage therapy into
WPI dip coatings for applications on fresh whole and
cut fruit and vegetable surfaces. 2018;83(7):1871–1879.
[106] Leverentz B, et al. Biocontrol of Listeria monocytogenes
on fresh-cut produce by treatment with lytic bacterio-
phages and a bacteriocin. 2003;69(8):4519–4526.
[107] Guenther S, et al. Virulent bacteriophage for efficient
biocontrol of Listeria monocytogenes in ready-to-eat
foods. J App environ microbio. 2009;75(1):93–100.
[108] Deasy T, et al. Isolation of a virulent Lactobacillus
brevis phage and its application in the control of beer
spoilage. 2011;74(12):2157–2161.
[109] Augustine J, Bhat SG. Biocontrol of Salmonella
Enteritidis in spiked chicken cuts by lytic bacterio-
phages ΦSP-1 and ΦSP-3. J basic microbio. 2015;55
(4):500–503.
[110] Patel J, et al. Inactivation of Escherichia coli O157: H7
attached to spinach harvester blade using
bacteriophage. 2011;8(4):541–546.
[111] Jun JW, et al. Eating oysters without risk of vibriosis:
application of a bacteriophage against Vibrio para-
haemolyticus in oysters. 2014;188:31–35.
[112] Bandara N, et al. Bacteriophages BCP1-1 and BCP8-2
require divalent cations for efficient control of
Bacillus cereus in fermented foods. 2012;31(1):9–16.
18 K. SSEKATAWA ET AL.
[113] Lone A, et al. Development of prototypes of bioactive
packaging materials based on immobilized bacterio-
phages for control of growth of bacterial pathogens in
foods. 2016;217:49–58.
[114] Kim KP, Klumpp J, Loessner MJ. Enterobacter saka-
zakii bacteriophages can prevent bacterial growth in
reconstituted infant formula. Int J Food Microbiol.
2007;115(2):195–203.
[115] Huang C, et al. Isolation, characterization, and appli-
cation of a novel specific Salmonella bacteriophage in
different food matrices. Food Res Int.
2018;111:631–641.
[116] Gouvêa DM, et al. Acetate cellulose film with bacter-
iophages for potential antimicrobial use in food
packaging. 2015;63(1):85–91.
[117] Gouvêa DM, et al. Absorbent food pads containing
bacteriophages for potential antimicrobial use in
refrigerated food products. 2016;67:159–166.
[118] Pereira C, et al. Application of phage therapy during
bivalve depuration improves Escherichia coli
decontamination. 2017;61:102–112.
[119] Ryan EM, et al. Synergistic phage-antibiotic combina-
tions for the control of Escherichia coli biofilms in
vitro. 2012;65(2):395–398.
[120] Kusmiatun A, Rusmana I, Budiarti S.
Characterization of bacteriophage specific to bacillus
pumilus from Ciapus River in Bogor, West Java,
Indonesia. HAYATI J Biosci. 2015;22(1):27–33.
[121] Kvachadze L, et al. Evaluation of lytic activity of
staphylococcal bacteriophage Sb-1 against freshly iso-
lated clinical pathogens. 2011;4(5):643–650.
[122] Sillankorva S, Neubauer P, Azeredo JJBB.
Pseudomonas fluorescens biofilms subjected to
phage phiIBB-PF7A. 2008;8(1):79.
[123] Jung L-S, et al. Evaluation of lytic bacteriophages for
control of multidrug-resistant Salmonella
Typhimurium. 2017;16(1):1–9.
[124] Alves DR, et al. Development of a high-throughput
ex-vivo burn wound model using porcine skin, and its
application to evaluate new approaches to control
wound infection. J Frontiers cellular infec microbio.
2018;8:196.
[125] Ghosh A, et al. Combined application of essential oil
compounds and bacteriophage to inhibit growth of
Staphylococcus aureus in vitro. 2016;72(4):426–435.
[126] Lungren MP, et al. Bacteriophage K for reduction of
Staphylococcus aureus biofilm on central venous
catheter material. 2013;3(4):e26825.
[127] Carson L, Gorman S, Gilmore B. The use of lytic
bacteriophages in the prevention and eradication of
biofilms of Proteus mirabilis and Escherichia coli.
2010;59(3):447–455.
[128] Nouraldin AAM, et al. Bacteriophage-antibiotic
synergism to control planktonic and biofilm produ-
cing clinical isolates of Pseudomonas aeruginosa.
2016;52(2):99–105.
[129] Vipra AA, et al. Antistaphylococcal activity of bacter-
iophage derived chimeric protein P128. 2012;12
(1):41.
[130] Chhibber S, Nag D, Bansal SJBM. Inhibiting biofilm
formation by Klebsiella pneumoniae B5055 using an
iron antagonizing molecule and a bacteriophage.
2013;13(1):1–8.
[131] Abdulamir AS, et al. The potential of bacteriophage
cocktail in eliminating Methicillin-resistant
Staphylococcus aureus biofilms in terms of different
extracellular matrices expressed by PIA, ciaA-D and
FnBPA genes. 2015;14(1):49.
[132] Didamony GE, Askora A, Shehata AA. Isolation and
characterization of T7-Like Lytic Bacteriophages
Infecting multidrug resistant pseudomonas aerugi-
nosa isolated from Egypt. Curr Microbiol. 2015;70
(6):786–791.
[133] Niculae C-M, et al. The 12th Edition of the Scientific
Days of the National Institute for Infectious Diseases
“Prof. Dr. Matei Bals” and the 12th National Infectious
Diseases Conference. 99. Neguț AC, Săndulescu O,
Streinu-Cercel A, et al. Inuence of bacteriophages on
sessile Gram-positive and Gramnegative bacteria. . in
BMC Infectious diseases. 2016. BioMed Central.
[134] Woo J, Ahn JJAOM. Assessment of synergistic com-
bination potential of probiotic and bacteriophage
against antibiotic-resistant Staphylococcus aureus
exposed to simulated intestinal conditions. 2014;196
(10):719–727.
[135] Morris J, et al. Evaluation of bacteriophage
anti-biofilm activity for potential control of orthope-
dic implant-related infections caused by
Staphylococcus aureus. 2019;20(1):16–24.
[136] Fu W, et al. Bacteriophage cocktail for the prevention
of biofilm formation by Pseudomonas aeruginosa on
catheters in an in vitro model system. 2010;54
(1):397–404.
[137] Olszak T, et al. In vitro and in vivo antibacterial
activity of environmental bacteriophages against
Pseudomonas aeruginosa strains from cystic fibrosis
patients. 2015;99(14):6021–6033.
[138] Guang-Han O, et al. Experimental phage therapy for
Burkholderia pseudomallei infection. 2016;11(7):
e0158213.
[139] Chhibber S, Gupta P, Kaur SJBM. Bacteriophage as
effective decolonising agent for elimination of
MRSA from anterior nares of BALB/c mice.
2014;14(1):212.
[140] Abdulamir AS, et al. Novel approach of using
a cocktail of designed bacteriophages against gut
pathogenic E. colifor bacterial load biocontrol.
2014;13(1):39.
[141] Furusawa T, et al. Phage therapy is effective in
a mouse model of bacterial equine keratitis. 2016;82
(17):5332–5339.
[142] Yen M, Cairns LS, Camilli AJNC. A cocktail of three
virulent bacteriophages prevents Vibrio cholerae
infection in animal models. 2017;8(1):1–7.
[143] Augustine J, Gopalakrishnan MV, Bhat SG.
Application of ΦSP-1 and ΦSP-3 as a therapeutic
strategy against Salmonella Enteritidis infection
using Caenorhabditis elegans as model organism.
J FEMS Microbio Letters. 2014;356(1):113–117.
[144] Vieira A, et al. Phage therapy to control
multidrug-resistant Pseudomonas aeruginosa skin
infections: in vitro and ex vivo experiments. 2012;31
(11):3241–3249.
[145] Kumari S, Harjai K, Chhibber Sjtjoiidc. Topical treat-
ment of Klebsiella pneumoniae B5055 induced burn
wound infection in mice using natural products.
2010;4(6):367–377.
[146] Wei C, et al. Developing a bacteriophage cocktail for
biocontrol of potato bacterial wilt. Virol Sin. 2017;32
(6):476–484. .
[147] Kocharunchitt C, Ross T, McNeil DL. Use of bacter-
iophages as biocontrol agents to control Salmonella
ALEXANDRIA JOURNAL OF MEDICINE 19
associated with seed sprouts. Int J Food Microbiol.
2009;128(3):453–459.
[148] Elhalag K, et al. Potential use of soilborne lytic Podoviridae
phage as a biocontrol agent against Ralstonia
solanacearum. J Basic Microbiol. 2018;58(8):658–669. .
[149] Soleimani-Delfan A, et al. Isolation of Dickeya dadan-
tii strains from potato disease and biocontrol by their
bacteriophages. 2015;46(3):791–797.
[150] Tan D, et al. Vibriophages differentially influence
biofilm formation by Vibrio anguillarum strains.
Appl Environ Microbiol. 2015;81(13):4489–4497.
[151] Khairnar K, et al. Novel bacteriophage therapy for control-
ling metallo-beta-lactamase producing Pseudomonas aer-
uginosainfection in Catfish. BMC Vet Res. 2013;9(1):264. .
[152] Christiansen RH, et al. Effect of bacteriophages on the
growth of flavobacterium psychrophilum and development
of phage-resistant strains. Microb Ecol. 2016;71(4):845–859.
[153] Stalin N, Srinivasan P. Characterization of Vibrio
parahaemolyticus and its specific phage from shrimp
pond in Palk Strait, South East coast of India.
Biologicals. 2016;44(6):526–533.
[154] Kittler S, et al. Effect of bacteriophage application on
Campylobacter jejuni loads in commercial broiler
flocks. 2013;79(23):7525–7533.
[155] Lau GL, Sieo CC, Tan WS, et al. Characteristics of
a phage effective for colibacillosis control in poultry.
J Sci Food Agric. 2012;92(13):2657–2663. .
[156] Belgini DRB, et al. Culturable bacterial diversity from
a feed water of a reverse osmosis system, evaluation of
biofilm formation and biocontrol using phages. World
J Microbiol Biotechnol. 2014;30(10):2689–2700. .
[157] Maal KB, et al. Isolation and identification of two novel
Escherichia Coli bacteriophages and their application in
wastewater treatment and coliform’s phage therapy.
2015;8:3. J Jundishapur j microbio.
[158] Bull JJ, Levin BR, DeRouin T, et al. Dynamics of success
and failure in phage and antibiotic therapy in experi-
mental infections. BMC Microbiol. 2002;2(1):35.
20 K. SSEKATAWA ET AL.
... Furthermore, phages are not toxic to mammals or humans and can be used for patients with antibiotic allergies [15]. Additionally, compared with the cost of producing novel antibiotics, generating phage preparations is inexpensive, it is easy to isolate new phages from a wide range of sources [16,17], and phages have the efficacy to penetrate and destroy bacterial biofilms [18]. Phage therapy has been evaluated with multi-drug resistant oral bacteria during animal and human studies and has been found to be safe [19]. ...
Article
Full-text available
Abstract: Background and Objectives: Enterococcus faecalis (E. faecalis) is a primary pathogen responsible for dental abscesses, which cause inflammation and pain when trapped between the crown and soft tissues of an erupted tooth. Therefore, this study aims to use specific phages as an alternative method instead of classical treatments based on antibiotics to destroy multidrug-resistant E. faecalis bacteria for treating dental issues. Materials and Methods: In the current study, twenty-five bacterial isolates were obtained from infected dental specimens; only five had the ability to grow on bile esculin agar, and among these five, only two were described to be extensive multidrug-resistant isolates. Results: Two bacterial isolates, Enterococcus faecalis A.R.A.01 [ON797462.1] and Enterococcus faecalis A.R.A.02, were identified biochemically and through 16S rDNA, which were used as hosts for isolating specific phages. Two isolated phages were characterized through TEM imaging, which indicated that E. faecalis_phage-01 had a long and flexible tail, belonging to the family Siphoviridae, while E. faecalis_phage-02 had a contractile tail, belonging to the family Myoviridae. Genetically, two phages were identified through the PCR amplification and sequencing of the RNA ligase of Enterococcus phage vB_EfaS_HEf13, through which our phages shared 97.2% similarity with Enterococcus phage vB-EfaS-HEf13 based on BLAST analysis. Furthermore, through in silico analysis and annotations of the two phages' genomes, it was determined that a total of 69 open reading frames (ORFs) were found to be involved in various functions related to integration excision, replication recombination, repair, stability, and defense. In phage optimization, the two isolated phages exhibited a high specific host range with Enterococcus faecalis among six different bacterial hosts, where E. faecalis_phage-01 had a latent period of 30 min with 115.76 PFU/mL, while E. faecalis_phage-02 had a latent period of 25 min with 80.6 PFU/mL. They were also characterized with stability at wide ranges of pH (4-11) and temperature (10-60 • C), with a low cytotoxic effect on the oral epithelial cell line at different concentrations (1000-31.25 PFU/mL). Conclusions: The findings highlight the promise of phage therapy in dental medicine, offering a novel approach to combating antibiotic resistance and enhancing patient outcomes. Further research and clinical trials will be essential to fully understand the therapeutic potential and safety profile of these bacteriophages in human populations.
... 41 Antibiotics may be replaced by lytic phages to treat bacterial illnesses that do not respond to traditional antibiotic therapy. 42,43 Total viable count for environmental isolates Angwa Rukuba_1, Old JUTH_1, Student Village Hostel_2, and Old JUTH_2 had the highest bacterial count, whereas Angwa Rukuba_2 and Student Village Hostel_1 had the lowest bacterial load count. This confirms that pathogenic bacteria can be isolated from the environment, which supports the findings of Obayiuwana et al. 44 carried out in wastewater obtained from pharmaceutical facilities; a significant colony of bacteria was discovered in Nigeria's Lagos and Ogun States. ...
Article
Full-text available
Background: Escherichia coli O157:H7, causing a severe public health concern, is currently an emerging food-borne pathogen. Bacteriophages (phages), to this effect, can provide alternative measures for their control. This study aimed at identifying and characterizing environmental Escherichia coli O157:H7, the phages infecting them, and some selected pathogenic bacteria. Materials and Methods: The bacteria and phages were isolated from six (6) different locations within Jos, Nigeria. The bacterial strains were identified using the Bruker matrix-assisted laser desorption ionization-time of flight mass spectrometry system, and their antibiotic susceptibility profiles were determined. The phages were screened against Escherichia coli O157:H7 and some selected pathogenic bacteria and examined for their physiological and morphological characteristics. Results: The mean bacterial viable counts indicated that although Angwa Rukuba_1 had the highest count of 7.69 ± 8.89 log10 CFU/mL, Student Village Hostel_1 had the lowest count of 7.52 ± 2.65 log10 CFU/mL. Bacillus spp. had the highest occurrence (30%), followed by Escherichia coli (28%), Pseudomonas aeruginosa (26%), Klebsiella spp. (8%), Proteus spp. (4%), and Staphylococcus aureus (4%). The results indicated that all the isolates were resistant to ceftazidime, whereas others were either susceptible or resistant to the eight common antibiotics used. The four bacteriophages designated as E. coli phage Anayo (ECPA)_1, ECPA_2, ECPA_3, and ECPA_4 had mean titers ranging from 9.80 ± 38.55 to 11.85 ± 13.45 log10 PFU/mL. The phages displayed broad activity against Escherichia coli O157:H7 and some selected pathogenic bacteria. The phages were relatively stable over a wide range of temperatures (45–55°C) for varying time intervals, NaCl concentrations (0.5–15%), chloroform (10%), and pH values (2–9) after 24 h of exposure. Conclusions: This study indicated that the Escherichia phages have activity on Escherichia coli O157:H7 strains as well as on other bacterial pathogens used. The experimental phages obtained in this study can be exploited in the formulation of phage cocktails for alternative therapy and bio-control of Escherichia coli O157:H7 because of their lytic activity and stability to different storage conditions.
... A lower MOI value still showed the reduction though it could not completely inhibit the bacterial host growth. More concentration of bacteriophages means more cells can be lysed, yielding rapid lytic activity 24 . However, these phages differed from phage ETEC-S3, where MOI higher than 0.001 could not completely inhibit the growth of the cells. ...
Article
Full-text available
Microbial food spoilage and foodborne disease are the main challenges in the food industry regarding food shelf life. Current preservation methods are frequently associated with changes in organoleptic characteristics and loss of nutrients. For this reason, bacteriophage offers an alternative natural method as a biocontrol agent that can reduce bacterial contamination in food without altering the organoleptic properties. This study was conducted to isolate and characterize bacteriophage from soil to control food spoilage bacteria, such as Bacillus cereus and Bacillus subtilis, and foodborne pathogenic bacteria, such as enterotoxigenic Escherichia coli (ETEC) and enterohemorrhagic E. coli (EHEC). Isolation was done by agar overlay assay method, and phages BC-S1, BS-S2, ETEC-S3, and EHEC-S4 were recovered. The host range of all isolated phages tended to be narrow and had high specificity towards the specific bacteria. The phage efficiency were measured where ETEC-S3 showed no effectivity against B. cereus and EHEC-S4 showed low efficiency against Enteropathogenic E. coli (EPEC). Morphology analysis was conducted for phage BC-S1 and ETEC-S3 with Transmission Electron Microscopy (TEM), and it is shown to belong to the Caudovirales order. Phages BC-S1 and BS-S2 significantly reduced the host bacteria when applied to the cooked rice and pasteurized milk samples with miMOI of 0.1. While phage ETEC-S3 at miMOI of 0.001 and phage EHEC-S4 at miMOI of 1 also showed a significant reduction when applied to chicken meat and lettuce samples at storage temperatures of 4 °C and 28 °C. The highest bacterial reduction of 100% was shown by phage BC-S1 on pasteurized milk samples and reduction up to 96.06% by phage ETEC-S3 on chicken meat samples at 28 °C incubation.
... The systematic reviews show more confidence for the short-term adoption of phage therapy for topical applications, and in bio-preservation, biodecontamination and bio-sanitization (Ssekatawa et al., 2021). They express more confidence in phage therapy through individualised treatment with phages matched to the bacteria, but with an expectation that in the longer-term phage therapy can be used in the early stages of infection and on a larger scale, reducing the up-front use of antibiotics, helping to preserve them (Suh et al., 2022). ...
Article
Full-text available
Bacteriophages (phages) have a long history of use in Eastern Europe and are poised for wider exploitation as novel antimicrobials in the context of anti-microbial resistance. Integrating phages into mainstream medicine requires an in-depth understanding of phages and of regulatory, economic and practical frameworks. Here we summarise insights from a UK perspective into therapeutic phage development and detail our progress towards being able to use phages for UTIs. Phages are of interest as new medicines to target bacterial infections that are currently difficult to treat with the available therapies, and protect the medicine that protect us, by preventing the use of last line antibiotics. A pressing need has arisen for phage products to be able to treat urinary tract infections (UTIs) caused by E. coli and Klebsiella pneumoniae. Clinical trials data are needed to ensure the safety, efficacy and clinical benefits of phage treatment according to modern criteria, motivate interest from clinicians and investment from the pharmaceutical industry and thus widen access to phages. We therefore aim to conduct a human clinical trial in participants with recurrent UTIs. We have established a UTI phage cocktail (combination) that we are opti-mising through a robust analysis of the phage genomes and phenotypes. On the genome front, we implement our graph-based framework to probe the genetic relationships between phages in the absence of a common marker. We describe here our repurposed ecological framework where we contextualise phage traits such as functionality in relevant physiological conditions. Ultimately , we hope to combine these approaches and correlate phage traits with therapeutic efficacy and more easily predict which phages should be developed as treatments. Human trials can be informed by data from large scale animal trials and we show how our recent work on swine and poultry pathogens informs phage
... The development of bacteriophage-based interventions is raising global attention (Ssekatawa et al., 2021). Bacteriophages (phages) are viruses that infect bacteria and cause their lysis (Kortright et al., 2019). ...
Article
Full-text available
Salmonella spp. is a relevant foodborne pathogen with worldwide distribution. To mitigate Salmonella infections, bacteriophages represent an alternative to antimicrobials and chemicals in food animals and food in general. Bacteriophages (phages) are viruses that infect bacteria, which interact constantly with their host. Importantly, the study of these interactions is crucial for the use of phages as a mitigation strategy. In this study, experimental coevolution of Salmonella Enteritidis (S. Enteritidis) and a lytic phage was conducted in tryptic soy broth for 21 days. Transfer to fresh media was conducted daily and every 24 hours, 2 mL of the sample was collected to quantify Salmonella OD 600 and phage titter. Additionally, time-shift experiments were conducted on 20 colonies selected on days 1, 12, and 21 to evaluate the evolution of resistance to past (day 1), present (day 12), and future (day 21) phage populations. The behavior of the dynamics was modeled and simulated with mathematical mass-action models. Bacteria and phage from days 1 and 21 were sequenced to determine the emergence of mutations. We found that S. Enteritidis grew for 21 days in the presence and absence of the phage and developed resistance to the phage from day 1. Also, the phage was also able to survive in the media for 21 days, however, the phage titer decreased in approx. 3 logs PFU/mL. The stability of the lytic phage population was consistent with the leaky resistance model. The time-shift experiments showed resistance to phages from day 1 of at least 85% to the past, present, and future phages. Sequencing of S. Enteritidis showed mutations in genes involved in lipopolysaccharide biosynthesis genes rfbP and rfbN at day 21. The phage showed mutations in the tail phage proteins responsible for recognizing the cell surface receptors. These results suggest that interactions between bacteria and phage in a rich resource media generate a rapid resistance to the infective phage but a fraction of the population remains susceptible. Interactions between Salmonella and lytic phages are an important component for the rational use of phages to control this important foodborne pathogen.
Article
Full-text available
Bacteriophages are currently considered one of the most promising alternatives to antibiotics under the ‘One Health’ approach due to their ability to effectively combat bacterial infections. This study aimed to characterize Vibrio species in hatchery water samples collected from an aquaculture farm and investigate the biocontrol potential of their bacteriophages. Vibrio spp. (n = 32) isolates confirmed by LNA probe-based qPCR were used as hosts. Three Vibrio phages were isolated. IKEM_vK exhibited a broad host range, infecting V. harveyi (n = 8), V. alginolyticus (n = 2), V. azureus (n = 1), and V. ordalii (n = 1). IKEM_v5 showed lytic activity against V. anguillarum (n = 4) and V. ordalii (n = 1), while IKEM_v14 was specific to V. scophtalmi (n = 4). The morphological appearance of phages and their lytic effects on the host were visualized using scanning electron microscopy (SEM). All three phages remained relatively stable within the pH range of 6–11 and up to 60 °C. The lytic activities and biofilm inhibition capabilities of these phages against planktonic Vibrio cells support their potential applications in controlling vibriosis in aquaculture systems.
Article
In the food industry, despite the widespread use of interventions such as preservatives and thermal and non-thermal processing technologies to improve food safety, incidences of foodborne disease continue to happen worldwide, prompting the search for alternative strategies. Bacteriophages, commonly known as phages, have emerged as a promising alternative for controlling pathogenic bacteria in food. This review emphasizes the potential applications of phages in biological sciences, food processing, and preservation, with a particular focus on their role as biocontrol agents for improving food quality and preservation. By shedding light on recent developments and future possibilities, this review highlights the significance of phages in the food industry. Additionally, it addresses crucial aspects such as regulatory status and safety concerns surrounding the use of bacteriophages. The inclusion of up-to-date literature further underscores the relevance of phage-based strategies in reducing foodborne pathogenic bacteria's presence in both food and the production environment. As we look ahead, new phage products are likely to be targeted against emerging foodborne pathogens. This will further advance the efficacy of approaches that are based on phages in maintaining the safety and security of food.
Article
Full-text available
Fish is an important dietary component and commodity of trade for Uganda. Currently, the currently grappling to meet the fish food and nutritional needs of its rapidly growing population and at the same time effectively harness the economic opportunities of its existing local, regional and international markets to support national socio-economic development. The country must produce an additional 1,000,000 mt of fish per annum above the 750,000 mt of fish currently being produced from both the fisheries and aquaculture to realize its National objectives for the sectors. Environmentally sustainable commercial aquaculture has been identified as the most feasible option for sustainably an additional 1,000,000 mt of fish per annum. However, this objective cannot be comprehensively achieved without addressing threats to aquaculture development arising from biosecurity and biosafety control, ecosystem health and climate-change. The loss of production, access to markets and negative impacts on public health and environmental sustainability that may accrue unless the aforementioned are addressed will negate growth that has been realized from the public and private sector investments that have so far been made into the sector. Establishing an aquaculture Monitoring, Surveillance and Control helps address these concerns by generating data and information to facilitate evidence-based decision making for aquatic animal disease control, environmental management, and public health and on national zoo-sanitary status to assure markets.
Article
Full-text available
Background: Despite significant advancements in surgical protocols and biomaterials for orthopedics, peri-prosthetic joint infection (PJI) remains a leading cause of implant failure. Staphylococcus aureus nasal colonization is an established risk factor for PJI, with methicillin-sensitive S. aureus a leading cause of orthopedic implant-related infections. The purpose of these in vitro studies was to investigate the antibacterial activity of a tailored bacteriophage cocktail against planktonic and biofilm-associated S. aureus. Methods: The S. aureus strains (n = 30) were screened for their susceptibility to a library of S. aureus-specific bacteriophage (n = 31). Five bacteriophage preparations that demonstrated bactericidal activity against >90% of S. aureus strains tested were combined as a StaPhage cocktail and assessed for their antibacterial activity toward planktonic and biofilm-associated S. aureus, with biofilms established on three-dimensional-printed porous titanium scaffolds. Results: StaPhage treatment immediately after bacterial inoculation inhibited growth of S. aureus by >98% in eight hour cultures when multiplicity of infection of phages to bacteria was greater than 1:1 (p < 0.01). Viable bacterial numbers within biofilms on titanium surfaces were significantly reduced (6.8 log10 to 6.2 log10 colony forming units [CFU]; p < 0.01) after exposure to the StaPhage cocktail, in vitro. No significant reduction was observed in biofilms exposed to 100 times the minimal inhibitory concentration of cefazolin (log10 6.81 CFU). Conclusions: Combined, these data demonstrate the in vitro efficacy of S. aureus-specific bacteriophage cocktails against S. aureus growing on porous titanium and warrant further in vivo studies in a clinically relevant animal model to evaluate the potential application of bacteriophage in the management of PJI caused by S. aureus.
Article
Full-text available
Objective: In this systematic review, we present the molecular epidemiology and knowledge gaps of the carbapenem resistance in East Africa as well as the future probable research interventions that can be used to address the emergence of carbapenem resistance in the region. Results: The 17 articles which presented concrete information about the prevalence of carbapenem resistance in East Africa were reviewed. Tanzania exhibited the highest level of carbapenem resistance at 35% while DRC had the lowest level at 0.96%. Uganda was the only country with studies documenting CR obtained amongst hospital environment isolates with incidence ranging from 21% in Pseudomonas aeruginosa to 55% in Acinetobacter baumannii. Carbapenem resistance was more exhibited in A. baumannii (23%), followed by P. aeruginosa (17%), Klebsiella pneumoniae (15%), Proteus mirabilis (14%) and Escherichia coli (12%) mainly isolated from respiratory tract, blood, urine and wound/pus. The regional genetic determinants of carbapenem resistance detected were blaIMP, blaVIM-1 blaSPM-l, blaNDM-1, blaOXA-23 blaOXA-24, blaOXA-58 and blaKPC.
Article
Full-text available
Purpose Many hospital-acquired infections (HAIs) can be transmitted by pathogens contaminating hospital surfaces, not efficiently controlled by conventional sanitation, which can indeed contribute to the selection of MDR strains. Bacteriophages have been suggested as decontaminating agents, based on their selective ability to kill specific bacteria. However, there are no data on their stability in detergents and their potential use in routine sanitation. On the other hand, a probiotic-based sanitation system (Probiotic Cleaning Hygiene System, PCHS) was recently shown to stably reduce pathogens on treated surfaces. However, its action is not specific and slow, being based on competitive antagonism. This work aimed to assess the effectiveness of a combined use of phages and PCHS in removing HAI-associated pathogens from different hard surfaces. Materials and methods The decontamination ability of phages in PCHS was tested in vitro and in situ, against drug-susceptible or resistant Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa strains, and using bacterial densities similar to those detected on hospital surfaces. Results Phages targeted efficiently all tested bacteria, maintaining their full activity when added to the PCHS detergent. Notably, the combined use of phages and PCHS not only resulted in a rapid reduction (up to >90%) of the targeted pathogens, but also, due to the stabilizing effect of probiotics, the pathogens were maintained at low levels (>99%) at later times too, when instead the effect of phages tends to diminish. Conclusion These results suggest that a combined biological system might be successfully used in hospital sanitation protocols, potentially leading to effective and safe elimination of MDR pathogens from the hospital environment.
Article
Full-text available
Biofilm formation in wounds is considered a major barrier to successful treatment, and has been associated with the transition of wounds to a chronic non-healing state. Here, we present a novel laboratory model of wound biofilm formation using ex-vivo porcine skin and a custom burn wound array device. The model supports high-throughput studies of biofilm formation and is compatible with a range of established methods for monitoring bacterial growth, biofilm formation, and gene expression. We demonstrate the use of this model by evaluating the potential for bacteriophage to control biofilm formation by Staphylococcus aureus, and for population density dependant expression of S. aureus virulence factors (regulated by the Accessory Gene Regulator, agr) to signal clinically relevant wound infection. Enumeration of colony forming units and metabolic activity using the XTT assay, confirmed growth of bacteria in wounds and showed a significant reduction in viable cells after phage treatment. Confocal laser scanning microscopy confirmed the growth of biofilms in wounds, and showed phage treatment could significantly reduce the formation of these communities. Evaluation of agr activity by qRT-PCR showed an increase in activity during growth in wound models for most strains. Activation of a prototype infection-responsive dressing designed to provide a visual signal of wound infection, was related to increased agr activity. In all assays, excellent reproducibility was observed between replicates using this model.
Article
Full-text available
Management of prosthetic vascular graft infections caused by Pseudomonas aeruginosa can be a significant challenge to clinicians. These infections often do not resolve with antibiotic therapy alone due to antibiotic resistance/tolerance by bacteria, poor ability of antibiotics to permeate/reduce biofilms and/or other factors. Bacteriophage OMKO1 binding to efflux pump proteins in P. aeruginosa was consistent with an evolutionary trade-off: wildtype bacteria were killed by phage whereas evolution of phage-resistance led to increased antibiotic sensitivity. However, phage clinical-use has not been demonstrated. Here, we present a case report detailing therapeutic application of phage OMKO1 to treat a chronic P. aeruginosa infection of an aortic Dacron graft with associated aorto-cutaneous fistula. Following a single application of phage OMKO1 and ceftazidime, the infection appeared to resolve with no signs of recurrence.
Article
Full-text available
Bacterial contamination on seafood resulting from unhygienic food-handling practices causes foodborne diseases and significant revenue losses. Moreover, control measures are complicated by a high prevalence of antibiotic-resistant bacteria. Alternative measures such as the phage therapy, therefore, is considered as an environmental and consumer-friendly biological control strategy for controlling such bacterial contamination. In this study, we determined the effectiveness of a bacteriophage cocktail in controlling E. coli strains [JM 109, ATCC 13706 and the, extended spectrum beta-lactamase resistant strain (ATCC BAA 196)] and S. enterica subsp. enterica (ATCC 13311) as single and combined contaminants of the edible oysters. Five different E. coli-specific phages (belonging to the Siphoviridae family) and a Salmonella phage (belonging to the Tectiviridae family) were successfully isolated from sewage water samples taken from a local sewage treatment plan in the Sunshine Coast region of Australia. Phage treatments applied to the pathogens when they were presented on the oysters as either single or combined hosts, resulted in significant decrease of the number of these bacteria on edible oysters. Results obtained indicated that bacteriophages could have beneficial applications in oyster-processing plants in controlling pathogenic bacterial infestations. This study thus contributes towards ongoing international efforts into the effective use of bacteriophages for biological control purposes.
Article
A new podovirus RsPod1EGY Ralstonia phage (GenBank accession no MG711516) with a specific action against R. solanacearum phylotype IIa, sequevar I (race 3, biovar 2) was isolated from Egyptian soil. The potential efficacy of the isolated phage to be used as biocontrol agent was evaluated in vitro and under greenhouse conditions. The podovirus phage produced a plaque size of 3.0–4.0 mm in diameter and completed its infection cycle in 180 min after infection with a burst size of ∼27 virions per infected cell. On the basis of restriction endonuclease analysis, the genome size of the phage was about 41 kb of double‐stranded DNA. In vitro studies showed that RsPod1EGY is stable at higher temperatures (up to 60 °C), and at a wide pH range (5–9). SDS–PAGE analysis indicated the major structural protein to be approximately 32 kDa. Bacteriolytic activity of RsPod1EGY against R. solanacearum was detected at different multiplicity of infection (MOI). RsPod1EGY proved to be effective in reduction and prevention of formation of surface polysaccharides of R. solanacearum, during the exponential growth phase of the latter. Interestingly, RsPod1EGY was effective in suppression of R. solanacearum under greenhouse conditions. All Phage‐treated tomato plants showed no wilt symptoms or any latent infection during the experimental period, whereas all untreated plants have wilted by 10 days post‐infection. The lytic stability of RsPod1EGY phage at higher temperature as well as its effective suppression of wilting symptoms under greenhouse conditions would contribute to biocontrol the bacterial wilt disease in Egypt under field conditions.
Article
There is a significant unmet need to develop antimicrobial solutions to reduce the risk of contamination in fresh produce. Bacteriophages have been proposed as a potential approach for controlling foodborne pathogens. This study evaluated the combination of edible dip coatings with T7 bacteriophages on whole and cut produce. The evaluation includes an assessment of phage loading, phage storage stability, antimicrobial activity, and phage stability during simulated gastric digestion on sliced cucumbers, sliced apples, and whole cherry tomatoes. In this evaluation, phages coated on fresh produce using edible whey protein isolate (WPI) were compared with phages coated from an aqueous suspension (control coating). The results demonstrated that WPI coatings load more phages than the control and enhanced phage stability during cold storage (4 °C) for cut apples and whole cherry tomatoes. Phage stability decreased by 1 to 3 log(PFU) in a simulated gastric environment. Phage antimicrobial activity against Escherichia coli BL21 decreased 2 to 4 log(CFU) of bacteria on cut apples and whole cherry tomatoes, while no significant bacterial reduction was observed for sliced cucumbers. Overall, the results show that WPI dip coating provides phage loading, stability, and antimicrobial activity to produce surfaces compared to the control coating, and thus may be considered an effective approach for extending phage therapy on fresh produce. Practical Application The practical application is to prevent bacterial cross contamination of fresh produce by using a combination of edible coating with bacteriophages. The results demonstrate enhanced loading and stability of phages on fresh produce when used in combination with an edible coating.
Article
Application of bacteriophages to eliminate foodborne pathogens in food matrices is an emerging research field. In this study, a promising phage candidate specific for Salmonella strains was screened and its ability to decrease Salmonella counts in some food, such as milk, sausage, and lettuce, was investigated. A total of 58 Salmonella phages were isolated from a wastewater treatment plant, sewage near a river, farm ditch near a lake, and poultry house. Among them, phages LPST10, LPST18, and LPST23 were highly efficient in infecting Salmonella Typhimurium ATCC 14028. In particular, phage LPST10 could infect all the tested Salmonella Typhimurium and Salmonella Enteritidis strains with high efficiency. Bacterial challenge tests revealed that phage LPST10 and its combination with phages LPST18 and LPST23 could consistently inhibit the growth of multiple strains. Phage LPST10 presented a lysis time of about 50 min with a burst size of 101 PFU/CFU, exhibited two distinct phases in the one-step growth curve, and was stable at a pH range of 3–13 that corresponds to the pH of most of the foods (pH 3.5–7.5) and at temperatures between 30 °C and 60 °C. Transmission electron microscopy demonstrated that phage LPST10 belongs to the Siphoviridae family, with an icosahedral head with a diameter of 83.26 nm and tail length and width of approximately 144.89 nm and 10.9 nm, respectively. A significant decrease in the bacterial counts (0.92–5.12 log10 CFU/sample) and an increase in phage titers (0–2.96 log10 PFU/sample) were observed in different food matrices tested. These results demonstrated that phage LPST10 is a promising candidate for controlling Salmonella contamination in foods owing to its safety and effectiveness.
Article
Yersinia enterocolitica, the primary cause of yersiniosis, is one of the most important foodborne pathogens globally and is associated with the consumption of raw contaminated pork. In the current study, four virulent bacteriophages (phages), one of Podoviridae (fHe-Yen3-01) and three of Myoviridae (fHe-Yen9-01, fHe-Yen9-02, and fHe-Yen9-03), capable of infecting Y. enterocolitica were isolated and characterized. fHe-Yen9-01 had the broadest host range (61.3% of strains, 65/106). It demonstrated a latent period of 35 min and a burst size of 33 plaque-forming units/cell, and was found to have a genome of 167,773 bp with 34.79% GC content. To evaluate the effectiveness of phage fHe-Yen9-01 against Y. enterocolitica O:9 strain Ruokola/71, we designed an experimental model of the food market environment. Phage treatment after bacterial inoculation of food samples, including raw pork (4 °C, 72 h), ready-to-eat pork (26 °C, 12 h), and milk (4 °C, 72 h), prevented bacterial growth throughout the experiments, with counts decreasing by 1-3 logs from the original levels of 2-4 × 103 CFU/g or ml. Similarly, when artificially contaminated kitchen utensils, such as wooden and plastic cutting boards and knives, and artificial hands, were treated with phages for 2 h, bacterial growth was effectively inhibited, with counts decreasing by 1-2 logs from the original levels of ca 104 CFU/cm2or ml. To the best of our knowledge, this is the first report of the successful application of phages for the control of Y. enterocolitica growth in food and on kitchen utensils.