Respirable Bacteriophages for the Treatment of Bacterial Lung Infections

19
Respirable Bacteriophages for the Treatment of Bacterial Lung Infections Susan Hoe, PhD, 1 Diana D. Semler, MSc, 2 Amanda D. Goudie, MSc, 2 Karlene H. Lynch, PhD, 2 Sadaf Matinkhoo, BSc, 1 Warren H. Finlay, PhD, 1 Jonathan J. Dennis, PhD, 2 and Reinhard Vehring, Dr-Ing 1 Abstract This review article discusses the development of respiratory therapeutics containing bacteriophages indicated for lung infections, specifically those that have become increasingly difficult to treat because of antibiotic re- sistance. Recent achievements and remaining problems are presented for each step necessary to develop a bacteriophage-containing dosage form for respiratory drug delivery, including selection of appropriate bacte- riophages for therapy, processing and purification of phage preparations, formulation into a stable, solid dosage form, and delivery device selection. Safety and efficacy studies in animals and human subjects are also reviewed. Key words: bacteriophages, respiratory drug delivery, bacterial lung infections, antibiotic resistance, phage therapy Introduction O ne of the most significant achievements of modern medicine is the control of infections. Clinical procedures such as surgery, transplantation, and chemotherapy are not feasible without effective control of bacterial infections. An- tibiotics were discovered and put into clinical practice in the first half of the 20 th century and contributed to a significant increase in life expectancy. (1) Antibiotics are substances that can kill or inhibit the growth of bacteria by interfering with bacterial functions or growth processes. Antibiotics can be categorized based on their specificity. Broad-spectrum antibiotics can be used against a wide range of bacteria, but even narrow-spectrum antibiotics typically target a whole class of bacteria, such as Gram-negative or Gram-positive. Due to the relatively nonspecific mechanisms of action of some broad-spectrum antibiotics, bacterial strains are often not identified before therapy is initiated. However, the nonspecific activity also results in some of the commonly observed antibiotic side effects, e.g., diarrhea, associated with the disruption of beneficial human micro- flora. Arguably the greatest weakness of antibiotics is that their effectiveness is diminished by the emergence of bacte- rial resistance. Within two decades after the discovery of penicillin, it was recognized that bacterial resistance would impact the efficacy of antibiotics. Sir Alexander Fleming, among others, warned early—and to no avail—against im- proper use of penicillin, e.g., using insufficient doses or ter- minating treatment prematurely. The emergence of resistant bacterial strains has been accelerated by a variety of factors, such as the overuse and misuse of antibiotics in clinical practice and in patients’ hands, and indiscriminate applica- tion of antibiotics in animal husbandry and fish farming. It has typically taken between 3 and 30 years following the introduction of a new antibiotic to observe the onset of resistance. (2) Bacterial resistance has become a significant problem in the treatment of infections of the respiratory system. Re- fractory or chronic infections are common in patients with compromised lungs, such as cystic fibrosis (CF) patients. Their lungs are colonized by a variety of bacteria, often ini- tially by Staphylococcus aureus or Haemophilus influenzae and later typically by bacteria of the Burkholderia cepacia complex (Bcc) or by Pseudomonas aeruginosa. (3) Many of these bacteria have high intrinsic antimicrobial resistance or have acquired mutations that result in resistance to a broad range of anti- biotics. (4,5) Similarly, highly resistant bacterial strains of Klebsiella pneumoniae, Streptococcus pneumoniae, (6) and En- terobacter spp. are encountered in hospital emergency care units and are a leading cause of mortality due to nosocomial pneumonia. Some of these bacterial species have now be- come resistant to all carbapenem antibiotics (7) via the transfer 1 Department of Mechanical Engineering, University of Alberta, Edmonton, Alberta, T6G 2G8, Canada. 2 Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada. JOURNAL OF AEROSOL MEDICINE AND PULMONARY DRUG DELIVERY Volume 26, Number 0, 2013 ª Mary Ann Liebert, Inc. Pp. 1–19 DOI: 10.1089/jamp.2012.1001 1

Transcript of Respirable Bacteriophages for the Treatment of Bacterial Lung Infections

Respirable Bacteriophages for the Treatmentof Bacterial Lung Infections

Susan Hoe, PhD,1 Diana D. Semler, MSc,2 Amanda D. Goudie, MSc,2 Karlene H. Lynch, PhD,2

Sadaf Matinkhoo, BSc,1 Warren H. Finlay, PhD,1 Jonathan J. Dennis, PhD,2 and Reinhard Vehring, Dr-Ing1

Abstract

This review article discusses the development of respiratory therapeutics containing bacteriophages indicatedfor lung infections, specifically those that have become increasingly difficult to treat because of antibiotic re-sistance. Recent achievements and remaining problems are presented for each step necessary to develop abacteriophage-containing dosage form for respiratory drug delivery, including selection of appropriate bacte-riophages for therapy, processing and purification of phage preparations, formulation into a stable, solid dosageform, and delivery device selection. Safety and efficacy studies in animals and human subjects are also reviewed.

Key words: bacteriophages, respiratory drug delivery, bacterial lung infections, antibiotic resistance, phagetherapy

Introduction

One of the most significant achievements of modernmedicine is the control of infections. Clinical procedures

such as surgery, transplantation, and chemotherapy are notfeasible without effective control of bacterial infections. An-tibiotics were discovered and put into clinical practice in thefirst half of the 20th century and contributed to a significantincrease in life expectancy.(1) Antibiotics are substances thatcan kill or inhibit the growth of bacteria by interfering withbacterial functions or growth processes.

Antibiotics can be categorized based on their specificity.Broad-spectrum antibiotics can be used against a wide rangeof bacteria, but even narrow-spectrum antibiotics typicallytarget a whole class of bacteria, such as Gram-negative orGram-positive. Due to the relatively nonspecific mechanismsof action of some broad-spectrum antibiotics, bacterialstrains are often not identified before therapy is initiated.However, the nonspecific activity also results in some of thecommonly observed antibiotic side effects, e.g., diarrhea,associated with the disruption of beneficial human micro-flora. Arguably the greatest weakness of antibiotics is thattheir effectiveness is diminished by the emergence of bacte-rial resistance. Within two decades after the discovery ofpenicillin, it was recognized that bacterial resistance wouldimpact the efficacy of antibiotics. Sir Alexander Fleming,

among others, warned early—and to no avail—against im-proper use of penicillin, e.g., using insufficient doses or ter-minating treatment prematurely. The emergence of resistantbacterial strains has been accelerated by a variety of factors,such as the overuse and misuse of antibiotics in clinicalpractice and in patients’ hands, and indiscriminate applica-tion of antibiotics in animal husbandry and fish farming. Ithas typically taken between 3 and 30 years following theintroduction of a new antibiotic to observe the onset ofresistance.(2)

Bacterial resistance has become a significant problem inthe treatment of infections of the respiratory system. Re-fractory or chronic infections are common in patients withcompromised lungs, such as cystic fibrosis (CF) patients.Their lungs are colonized by a variety of bacteria, often ini-tially by Staphylococcus aureus or Haemophilus influenzae andlater typically by bacteria of the Burkholderia cepacia complex(Bcc) or by Pseudomonas aeruginosa.(3) Many of these bacteriahave high intrinsic antimicrobial resistance or have acquiredmutations that result in resistance to a broad range of anti-biotics.(4,5) Similarly, highly resistant bacterial strains ofKlebsiella pneumoniae, Streptococcus pneumoniae,(6) and En-terobacter spp. are encountered in hospital emergency careunits and are a leading cause of mortality due to nosocomialpneumonia. Some of these bacterial species have now be-come resistant to all carbapenem antibiotics(7) via the transfer

1Department of Mechanical Engineering, University of Alberta, Edmonton, Alberta, T6G 2G8, Canada.2Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada.

JOURNAL OF AEROSOL MEDICINE AND PULMONARY DRUG DELIVERYVolume 26, Number 0, 2013ª Mary Ann Liebert, Inc.Pp. 1–19DOI: 10.1089/jamp.2012.1001

1

of genes that encode a highly effective b-lactamase en-zyme.(8) In addition, it is likely that at least 5% of the morethan 400,000 multidrug-resistant tuberculosis cases per yearworldwide are caused by extensively drug-resistant Myco-bacterium tuberculosis,(9) which is practically untreatable indeveloping countries.(10) Some of this high-level antibioticresistance is attributable to the fact that several pulmonarypathogens are capable of adopting a biofilm lifestyle,(11) inwhich the bacteria are encased in a self-synthesized polymericmatrix that serves to protect them from antibiotic activity andimmune responses. The exact mechanisms producing biofilmresistance remain undiscovered, but may be through a com-bination of bacterial adaptations, the net effect being thatbiofilm bacteria are nearly impossible to clear even usingcombinations of conventional chemical antibiotics.

More than 20 classes of antibiotics were introduced forclinical use between 1940 and 1962. However, only two havebeen added after this time, and very few are currently indevelopment. Given the recent increase in the use and

overuse of antibiotics, it is not surprising that now manybacterial species have developed resistance against morethan one antibiotic. A recent study by Coates et al. estimatesthat 20 new classes of antibiotics would have to be dis-covered and developed in the next five decades to controlbacterial infections at the current level.(12) Because it appearsunlikely that this goal can be accomplished, even if strongereconomic incentives for the development of new antibioticswere put in place, a search for alternatives to antibioticsseems warranted.

One alternative, phage therapy, has received renewed at-tention.(13,14) Bacteriophages, or phages, are viruses that in-fect and kill bacteria.(15) Figure 1 gives an overview of someselected milestones in the discovery of bacteriophages andtheir application to the treatment of respiratory infections.Phages were discovered independently by Frederick Twortand Felix d’Herelle at the beginning of the 20th century.D’Herelle soon thereafter initiated experimental treatment ofbacterial infections with phage ‘‘cocktails’’ (preparations

FIG. 1. A historical over-view of respiratory phagetherapy.

2 HOE ET AL.

consisting of a collection of different phages active againstmultiple bacterial strains or species), despite the fact thatlittle was known about phage particles at the time.(15,16) Earlyphage therapy was quite successful, leading to the devel-opment of many commercial phage preparations in the 1920sand 1930s by companies such as Eli Lilly & Company andwidespread use of phage therapy in Europe and the UnitedStates. However, early clinical trials also suffered frompoorly characterized phage preparations and a lack of un-derstanding of phage biology. As a result, the efficacy ofphage treatment was called into question, and phage therapyin the West was largely abandoned with the advent of che-mical antibiotics.(17) Nevertheless, phage research intensifiedin the 1940s as phages were recognized by Max Delbruck,among others, as powerful tools for the exploration of ge-netics. The American Phage Group, coordinated by Del-bruck, contributed largely to the scientific understanding ofphages that emerged in the 1940s and 1950s. In 1969, theNobel Prize in Physiology or Medicine was awarded to MaxDelbruck, Alfred D. Hershey, and Salvador E. Luria, ‘‘fortheir discoveries concerning the replication mechanism andthe genetic structure of viruses.’’(18) Phage research becamean exact science with rigorous quantitative methods. Some ofthe earlier failures of phage therapy could now be explained.It was realized that phages typically have a narrow bacterialhost range, and that this host range specificity depends on anexplicit match between the bacteriophage and bacterial sur-faces or surface structures.

The first reports of successful inhalation therapy usingphages emerged around 1960.(19,20) Table 1 outlines selectedstudies of inhalation phage therapy in humans. Delacostesuccessfully treated patients with refractory cough using anebulized phage cocktail.(19) Hoeflmayr nebulized a phagecocktail concomitantly with antimicrobial agents.(20) En-dotoxins were intentionally left in the preparation to stimulatean immune response in the patients. The treatment was suc-cessful in 90% of patients with chronic bronchitis. In EasternEurope, in particular in Russia, Poland, and Georgia, phagetherapy has been continuously applied and further developedsince the 1920s.(21) Phage therapy research centers include theHirszfeld Institute of Immunology and Experimental Ther-apy(22) in Wroc1aw, Poland, and the Eliava Institute(23) inTbilisi, Georgia. In Georgia, phage therapy is an accepted andwidely used mode of treatment. Commercial phage products,including phage cocktails that are available to the publicwithout prescription, are manufactured and approved inGeorgia and Russia.(21) The prospects for respirable phages asan alternative to antibiotics appear promising.

In this article, we provide an overview of the individualsteps that need to be taken to develop a bacteriophage dos-age form for administration to the respiratory system. Thefirst sections review the selection of adequate phages, theiramplification, and purification. Then recent advances in theformulation of phage preparations into stable, respirabledosage forms are discussed and delivery device options arereviewed. Finally, the efficacy and safety of phage therapy asestablished by recent clinical trials are summarized.

Bacteriophage Selection

The first steps in the development of a phage therapeuticare the isolation and selection of safe and efficacious

Ta

bl

e1.

Se

le

ct

ed

St

ud

ie

so

fB

ac

te

rio

ph

ag

eT

he

ra

py

fo

rt

he

Tr

ea

tm

en

to

fR

esp

ir

at

or

yD

ise

ase

sin

Hu

ma

ns

Res

earc

hg

rou

pY

ear

Con

ten

tR

esp

irat

ory

dis

ease

trea

ted

Nu

mbe

rof

pat

ien

tstr

eate

dR

ecor

ded

resu

lts

oftr

eatm

ent

Ref

eren

ce

Eli

ava

Inst

itu

te(T

bil

isi,

Geo

rgia

)B

efo

re19

47S

.au

reu

sp

hag

eP

ulm

on

ary

S.

aure

us

infe

ctio

n45

lun

go

per

atio

np

atie

nts

trea

ted

wit

hp

hag

ean

der

yth

rom

yci

n

24%

re-i

nfe

cted

wit

hp

leu

ral

infe

ctio

n,

agai

nst

67%

inco

ntr

ol

gro

up

(ery

thro

my

cin

on

ly)

Ch

anis

hv

ili(1

45

)

Ho

eflm

ayr

1957

–196

1C

ock

tail

of

180–

200

ph

ages

Acu

tean

dch

ron

icb

ron

chit

is29

pat

ien

ts55

.1%

ach

iev

edfu

llre

cov

ery

,34

.8%

ach

iev

edcl

inic

alim

pro

vem

ent

Ho

eflm

ayr(2

0)

Mel

adze

etal

.19

82S

.au

reu

sp

hag

eP

ulm

on

ary

S.

aure

us

infe

ctio

n22

3p

atie

nts

(45

rece

ived

i.v

.p

hag

e)82

%ac

hie

ved

full

reco

ver

y,

com

par

edw

ith

64%

of

pat

ien

tsre

ceiv

ing

anti

bio

tics

(n=

117)

Su

lak

vel

idze

(14

6)

Iose

lian

iet

al.

1980

Co

ckta

ilo

fp

hag

esfo

rS

.au

reu

s,S

trep

toco

ccu

s,P

rote

us,

and

E.

coli

Pu

lmo

nar

yS

.au

reu

sin

fect

ion

45p

atie

nts

Su

cces

sfu

lly

trea

ted

wit

han

tib

ioti

csS

ula

kv

elid

ze

Hir

szfe

ldIn

stit

ute

of

Imm

un

olo

gy

and

Ex

per

imen

tal

Th

erap

y(W

roc1

aw,

Po

lan

d)

1987

–199

9S

.au

reu

s,K

lebs

iell

a,P

rote

us,

P.

aeru

gin

osa,

or

E.

coli

ph

ages

Mu

cop

uru

len

tch

ron

icb

ron

chit

is,

lary

ng

itis

,rh

init

is,

bro

nch

op

neu

mo

nia

,p

leu

riti

s

377

pat

ien

ts31

3(8

3%)

ach

iev

edfu

llre

cov

ery

wit

hn

od

etec

tab

leb

acte

ria

51(1

3.5%

)ac

hie

ved

mar

ked

imp

rov

emen

tw

ith

som

ed

etec

ted

bac

teri

a

Web

er-D

abro

wsk

aet

al.(1

47

)

BACTERIOPHAGES FOR BACTERIAL LUNG INFECTIONS 3

bacteriophages. Phages are found in vast quantities andabundant variety in all environments that harbor bacteria.Phages specific to a certain bacterial strain usually can beisolated from samples containing the host bacteria, suchas soil or sewage. Phages are obligate parasites of bacteria.They have been shown to coevolve with their bacterialhosts.(24) Consequently, phages specific to a mutated bacte-rium have been observed to emerge soon after mutation ofthe bacterial host.(25)

An effective phage must: (1) successfully bind to the sur-face of the bacterium; (2) transfer its genetic material into thehost bacterium; (3) synthesize progeny virions; and (4) lysethe bacterial cell.

This process of infection is referred to as the ‘‘lytic lifecycle’’ as shown in Figure 2. One lytic cycle is completed inabout 15–60 min and produces typically 100–200 new pha-ges.(21,26–32) In phage therapy, the concentration of phagesin vivo will theoretically grow rapidly if the bacterial host ispresent in sufficient quantity. When used therapeutically, thehigh interaction specificity between a phage and the bacterialcell prevents the viral particles from destroying human cells.However, this also limits the usefulness of certain phagesdue to their extreme selectivity. In selecting a phage, or set ofphages for clinical use, the most important criterion is bac-terial host specificity. Phage host ranges are limited due tofactors such as the inability to bind the host or to transferDNA, the cleavage of phage DNA by host proteins, or bac-terial abortive infection systems.(33) The narrow host range ofphages is beneficial, because it avoids the adverse effects onthe normal flora observed following broad-spectrum antibi-otic treatment.(25) A disadvantage of this specificity, how-ever, is that a panel of phages must be screened on theinfecting bacterium prior to administration in order to assesswhich phages are the most effective in lysing the bacterialcells.(16)

To mitigate this issue, phages with expanded bacterialhost ranges can be isolated or developed using classical ormolecular techniques.(34,35) In addition, multiple phages maybe used together in a cocktail formulation, and this is espe-cially useful for polymicrobial infections where multiplebacterial pathogens are present (as might be found in the CFlung).(34) This can be achieved by selecting phages that infect

the most common bacterial species or strains associated witha particular infection, or by first determining which bacterialspecies or strains are involved in the infection and selectingphages for each. A CF patient, for example, might be treatedwith a cocktail of phages capable of infecting a number ofdifferent P. aeruginosa, Burkholderia cenocepacia, and S. aureusstrains, because these are common pathogens found in thelungs of CF patients.(36) Phage cocktails can also be used toreduce the chance of bacterial resistance occurring, becausephages specific for two different bacterial receptors wouldrequire that a single target cell undergo simultaneous mu-tation of two different receptors, a highly improbable event.

Virulent versus temperate phages

Two other important criteria for phage selection are anobligately lytic lifestyle and the absence of genes that encodevirulence factors. Phages can be classified as either obligatelylytic (also referred to as virulent) or temperate. Obligatelylytic phages follow the lytic life cycle, causing lysis of thebacterial host and release of progeny virions at the end ofeach infection cycle.(37) In contrast, temperate phages mayeither lyse the bacterial host and release virions or, alterna-tively, the phage DNA may be incorporated into the cell aspart of the bacterial chromosome or as a plasmid, remainingdormant as a ‘‘prophage’’ through successive bacterial celldivisions until triggered to reenter the lytic life cycle.(37)

During lysogeny, the host cell is not killed by the phage andbecomes resistant to infection by all other related phages.(38)

The integrated prophage genome may occasionally containsequences that encode virulence proteins such as adhesins ortoxins.(39) In the case of Vibrio cholerae (in which virulencefactors, cholera toxin, and the toxin-coregulated pilus arefound to be prophage-encoded), these genes can have dra-matic effects on the pathogenicity of the bacterial host.(40,41)

Temperate phages may also transfer bacterial virulencegenes via specialized transduction, a process in which hostDNA is packaged into progeny virions along with phageDNA following induction.(42)

Genome sequencing and analysis can be used to assessboth the lifestyle (obligately lytic or temperate) and the genecontent of a phage prior to clinical use. Temperate phageshave characteristic sequences that lytic phages lack, includ-ing a repressor gene, operator sequences to which the re-pressor protein binds, and (in most cases) an integrasegene.(38) It should be noted that, in rare cases, phages withthese sequences have been identified that do not form stablelysogens (lysogenized bacteria).(43,44) Therefore, assessmentof phage lifestyle should include both genomics analysis andscreening for lysogens or turbid plaques (characteristic oftemperate phages).(42) Virulence genes can also be identifiedthrough bioinformatics analysis and may show similarity toproteins that mediate cell adherence, immune evasion, toxinproduction, or other processes.(39)

The concerns discussed above usually lead to a recom-mendation to select lytic phages for therapeutic applications.However, several cases have been identified in which phageswith temperate lifestyles or virulence genes have been shownto be effective in phage therapy applications. For instance,most phages of Clostridium difficile and the Bcc appear to betemperate.(45,46) Out of necessity, phages used (and shown tobe effective) in infection models of these bacterial species areFIG. 2. Life cycle of lytic bacteriophages.

4 HOE ET AL.

either temperate or putatively temperate.(47–50) Temperatephages have also been shown to be effective against S. au-reus, even in a case where the phage carried putative leu-kocidin and enterotoxin A genes.(51–53) Instead of discardingphages encoding proteins involved in lysogeny or virulence,genetic modification may be an option to improve safetyprior to clinical use. Lytic mutants of temperate phages havebeen shown to be effective against Escherichia coli in a mousemodel,(54) S. aureus in human volunteers and a mouse woundmodel,(55) and B. cenocepacia in an invertebrate model.(48)

These phages may be spontaneous mutants (with a charac-teristic clear plaque morphology) or generated by random ortargeted mutagenesis of the operator or repressor se-quences.(42,48,55) Although the use of obligately lytic phagesprevents lysogenic conversion,(56) virulence genes can beremoved from the temperate phage genome such that theydo not introduce problematic genetic material upon recom-bination with the bacterial chromosome during infection.

It has been shown that some phages can penetrate biofilmmatrices, or possess surface-exposed enzymes that are ca-pable of degrading the polymeric matrix components ofbacterial biofilms.(57,58) Such polysaccharide depolymerasesor glycanases are capable of allowing some phages access tobacterial host cells deeper within biofilms, thereby enhancinglysis and degradation of the bacterial biofilm.(59,60) The genesencoding such phage biofilm-degrading enzymes can betransferred to and expressed by other phages in order toenhance phage penetration of biofilms.

In addition to removing lysogeny and virulence genes,or adding genes encoding enzymes capable of enhancingbiofilm degradation, other phage components can be genet-ically manipulated prior to therapeutic use.(61) Such modi-fications include bacterial host range expansion(35,62) andthe introduction of antibacterial toxin genes.(63–65) En-gineered phages have been shown to be active against sev-eral species in mouse models, including E. coli, S. aureus, andP. aeruginosa.(53,64–66)

Potential host effects

Phages are considered to be biological entities and, whenused as therapeutic agents, have the potential to interact withor stimulate the host’s immune system. However, phages asantibacterial agents are not unique in this respect. Severalapproved protein-based pharmaceuticals also have the abil-ity to stimulate an immune response, and chemical antibi-otics that achieve their effects through bacterial lysis willcause the release of bacterial cell wall components, includingendotoxin. Current dogma suggests that phage particles inan animal or human host are cleared by the reticuloendo-thelial system within a few days.(54,67,68) However, the im-munogenicity of phage particles is likely highly variable,partially depending on where the phages are initially ad-ministered (bloodstream versus body surface). For example,Carmody et al. were able to provide a small therapeutic effectto a pulmonary bacterial infection via intraperitoneal phagedelivery, suggesting that some phages can easily penetratedifferent body cavities without activating an anti-phage im-mune response.(47) Within the lung, phages showed ampli-fication and, at the same time, markers of pathogen-inducedinflammation were reduced. Similarly, Merril et al. were ableto evolve long-circulating phages that were presumably less

immunogenic than parental types, suggesting that somephages may be relatively invisible to an immune re-sponse.(54) Although little rigorous information exists aboutimmune stimulation by phages in vivo, the paucity of suchdata suggests that phage lysis of bacteria in an animal orhuman host is of little consequence and, at least, does notworsen the host’s condition. Several recently completedphage therapy clinical trials suggest that the presence ofphages in different body cavities does not enhance the dis-ease state due to immune activation.(16,69) In support of thisview, several phage-related products have now passed someregulatory standards and have been classified by the U.S.Food and Drug Administration as GRAS (Generally Re-garded As Safe), have been registered with the U.S. En-vironmental Protection Agency, or have been approved foruse by the U.S. Department of Agriculture.(70,71)

Summary

By using conventional screening techniques together withgenetic modification, one can select phages for clinical usethat are bacterial host-specific, obligately lytic, free of viru-lence genes, and highly active in vivo. There is little existingdata on the human or animal immune response to phages,but also little evidence of phages having a negative effect ondisease state.

Bacteriophage Preparation

As discussed, there are already many phages availablethat are capable of infecting a wide range of antibiotic-resistant bacteria, and more are continually being isolatedand characterized. The procedures for growing phages arewell described elsewhere; a summary of these preparationsteps is provided in Figure 3.

Phages are obtained from environmental or biologicalsamples, such as soil, river water, sewage effluent, and fecalmatter. One or a panel of bacterial strains is used to propa-gate any desired phages in the sample before filter sterili-zation to remove particulate and cellular matter. Samples areoften mixed with a culture medium along with the bacterialhost, and the entire preparation is incubated to allow phagesto bind to potential hosts. If host-specific phages are presentin the sample, the bacterial cells will be lysed to releaseprogeny virions. The lysate is then centrifuged or passedthrough a membrane filter, or both, to elute the phages in asterilized sample. This sample is then plated against bacterialstrains to isolate the single desired phage.(16,72–74)

Once phages have been isolated, they can then be grownto the desired concentration. This is known as phage am-plification. As with enrichment, the phage is cultured withthe host bacterial strain or strains in a suitable medium andincubated. This can be done in liquid culture or on agarplates, followed by an overlay of sterile water or suspensionmedium to suspend the phages in solution. Separation of thephage from cellular debris is often carried out by centrifu-gation and membrane filtration (Fig. 3).

Early phage preparations were often just filtered lysatesthat still contained bacterial endo- and exotoxins at the pointof human delivery.(75) There are no reports of serious sideeffects due to endotoxins in the early literature covering ex-perimental bacteriophage therapy. Nevertheless, current ex-pectations regarding the safety of biological preparations

BACTERIOPHAGES FOR BACTERIAL LUNG INFECTIONS 5

require removal of bacterial endotoxin from phage prepara-tions before proceeding with any other modifications (i.e.,spray-drying, freeze-drying, nebulizing, etc.). Endotoxin is apart of the cell wall of Gram-negative bacteria known aslipopolysaccharide. The lipid component is associated withtoxicity, whereas the polysaccharide component is respon-sible for eliciting an immune response.(76)

Lipopolysaccharide is typically present in initial phagepreparations once phages have lysed bacterial host cells.There are a number of commercially available products thatremove endotoxin from phage samples. For large-scalepreparations, ion-exchange or gel filtration chromatographymay be used.(76–78) Another method of removing endotoxininvolves a series of phage dialysis and purification stepsusing an organic solvent such as octyl or butyl alcohol.(79)

Because laboratory-scale methods of bacteriophagegrowth and purification tend to be labor-intensive and time-consuming, commercial production of phage preparationsrequires optimization of manufacturing steps during scale-up. The production process for live attenuated virus vaccineshas been developed to commercial stage and is now a rou-tine operation.(80) Many of the processing steps used in virusvaccine production are similar to the ones needed for bac-teriophage manufacturing.(72) Specific improvements forphage manufacturing processes can be found in the patentliterature: Bujanover proposed the use of a semisolid culturemedium instead of liquid medium for incubation with thehost bacterial strain.(81) This is followed by a repeating pro-cess of dilution, centrifugation, and extraction of supernatantto achieve a bacteriophage titer of at least 1010 plaque-forming units (PFU)/mL in the final extract. Lenherr andBartsch outlined a fully automated purification process.(73)

A prepurification step involves passing the unprocessedpostamplification phage and bacteria preparation through arotating membrane prefilter with a low-pressure pump. Thefiltrate is subjected to subsequent filtration steps with pro-

gressively finer pore size. The phages are then washed off thefinal filter and collected.

Summary

Bacteriophage preparation, from raw environmentalsamples to purified endotoxin-free phage product, can beaccomplished using established biotechnology processes.Similar production processes for viral vaccines have beenscaled up and are routinely used in vaccine manufacturing.

Formulation

After the selection of an appropriate phage and prepara-tion of phage stock, the next step in the development of arespirable dosage form is to incorporate the phage into astable, inhalable formulation.

The reported shelf life of phage preparations varies, andalthough many phages can be stored for years without anysignificant loss, not all phages are suited to long-term storagewithout further stabilization measures. Clark (1962) testedthe effect of 2 years of different storage conditions on thepreservation of 14 different phages.(82) He found that mostphages survived better when stored at 4�C. At this temper-ature, 10 phages survived best when freeze-dried, four whensuspended in liquid, and three when stored in 50% glycerol.More recently, it has been reported that lactococcal phagesare stored best at - 30�C in a broth supplemented with cal-cium, but are less stable when freeze-dried.(83) However, Bccand P. aeruginosa phages KS4-M and FKZ, respectively, canbe stored without significant titer loss after lyophilizing in alactose/lactoferrin matrix.(84)

Storage conditions for phage preparations will, therefore,depend on the individual phage, or phages, that they con-tain. A variety of commercial phage preparations in Georgiaare marketed as liquid suspensions with a shelf life of 2 yearswhen stored at 2–15�C(85) and are also available in a

FIG. 3. Processing steps inthe manufacture of bacterio-phage preparations.

6 HOE ET AL.

Ta

bl

e2.

St

ud

ie

sIn

ve

st

ig

at

in

gB

ac

te

rio

ph

ag

eS

ta

bil

it

y,

Disc

usse

din

‘‘F

or

mu

la

tio

n’’

Ref

eren

ceV

aria

ble

stu

die

dP

hag

este

sted

Pre

par

atio

nC

omm

ents

Cla

rk(8

2)

Tem

per

atu

re,

soli

d/

liq

uid

stat

eIn

clu

des

ph

ages

agai

nst

Bac

illu

s,E

.co

li,

P.

aeru

gin

osa,

and

S.

aure

us

Pre

par

edin

bro

th,

bro

thw

ith

50%

gly

cero

l,d

ried

,o

rly

op

hil

ized

;st

ore

dat

4�C

or

26�C

for

2y

ears

Gre

ater

surv

ival

(<1

log

10

tite

rlo

ss)

wh

enst

ore

dat

4�C

,an

dw

hen

spra

y-d

ried

;P

.ae

rug

inos

ain

bro

thsh

ow

edn

oti

ter

loss

wh

enst

ore

dat

4�C

Gar

cıa

etal

.(14

8)

Tem

per

atu

reF

H5

andF

A72

(S.

aure

us

ph

ages

)P

rep

ared

inU

HT

wh

ole

mil

k,

and

sto

red

at4,

22,

37,

and

72�C

for

up

to8

hr

No

tite

rlo

ssat

4�C

/8

hr;

0.1–

0.15

log

10

tite

rlo

ssat

37�C

/8

hr;

no

surv

ival

at72

�C/

1m

inC

ald

eira

and

Pea

bo

dy

(88

)T

emp

erat

ure

MS

2(E

.co

lip

hag

e)S

ub

ject

edto

20–1

00�C

tem

per

atu

rera

ng

e,fo

r2

min

atea

chte

mp

erat

ure

po

int;

also

sub

ject

edto

55�C

con

dit

ion

for

40m

in

Ph

age

rem

ain

ed10

0%so

lub

leat

60�C

,20

%so

lub

leat

65�C

,an

d0%

solu

ble

at70

�C;

ph

age

was

90%

solu

ble

afte

r40

min

at55

�CB

rier

set

al.(8

9)

Tem

per

atu

re,

ion

icst

ren

gth

,p

HF

KZ

(P.

aeru

gin

osa

ph

age)

Th

een

zym

atic

acti

vit

yo

fa

pro

tein

inF

KZ

was

mea

sure

din

pH

5.0–

8.0,

20–3

20m

Mio

nic

stre

ng

th,

and

25–9

0�C

con

dit

ion

s

Op

tim

alp

Han

dio

nic

stre

ng

thra

ng

ew

ere

pH

6.2–

6.8

and

140–

200

mM

;h

eati

ng

for

10m

inat

60�C

red

uce

dac

tiv

ity

to*

60%

of

rela

tiv

eo

pti

mu

m;

at90

�C,

acti

vit

yre

du

ced

furt

her

to*

10%

Kn

ezev

icet

al.(9

2)

Tem

per

atu

re,

pH

d,J-

1,r

-1,

001A

(P.

aeru

gin

osa

ph

ages

)A

dso

rpti

on

rate

of

ph

age

toh

ost

bac

teri

alce

llm

easu

red

afte

rex

po

sure

to7,

18,

26,

37,

and

44�C

for

30m

inP

hag

ev

iab

ilit

ym

easu

red

afte

rex

po

sure

top

H1.

2,3,

5,7,

and

9

90–9

5%p

hag

ead

sorp

tio

nac

hie

ved

for

all

fou

rp

hag

esat

37–4

4�C

;d

and

r-1

rem

ain

75%

via

ble

atp

H5,

60–7

0%v

iab

leat

pH

3;J-

1an

d00

1Are

mai

n50

%v

iab

leat

pH

5,15

%v

iab

leat

pH

3L

ang

let

etal

.(94

)p

HM

S2

(E.

coli

ph

age)

Pre

par

edin

0.4

MK

Cl

atp

H2.

5,3.

9,an

d6.

7at

20�C

No

tite

rlo

ssat

pH

6.7,

1lo

g1

0ti

ter

loss

atp

H3.

9,3

log

10

tite

rlo

ssat

pH

2.5

Go

lsh

ahi

etal

.(96

)Io

nic

stre

ng

thK

S4-

M(B

CC

ph

age)

Pre

par

edin

ah

yp

oto

nic

(105

mO

sm)

and

iso

ton

ic(2

82–2

90m

Osm

)n

ebu

lize

rso

luti

on

;n

ebu

lize

dfr

om

Par

iL

CS

tar

and

Par

ieF

low

neb

uli

zers

Hy

po

ton

icso

luti

on

(in

itia

lti

ter

2.5

·10

8P

FU

)h

adan

emit

ted

ph

age

cou

nt

of

1.78

·10

7P

FU

.Is

oto

nic

solu

tio

n(i

nit

ial

tite

r5.

108

PF

U/

mL

)h

adan

emit

ted

ph

age

cou

nt

of

108

PF

U.

7

lyophilized form with 1.5 years of shelf life at unspecifiedstorage conditions.(86) Given that phages are protein struc-tures, they are potentially susceptible to factors that areknown to cause protein denaturation, including temperature,pH, interfacial adsorption, and ionic strength. Table 2 pro-vides a summary of the studies discussed in this section.

Temperature

It is well known that the application of high temperaturesleads to thermal denaturation of proteins, a fact that can beused for sterilization or pasteurization purposes. Phagesshow typical behavior in response to short-term temperatureexposure: S. aureus-specific phages FH5 and FA72 werefound to maintain initial titer ( > 4 · 105 PFU/mL) after sus-pension in ultra-high-temperature–processed, whole-fat milkwhen stored at 4�C for 8 hr, showed approximately 20–25%titer loss at 37�C, and showed complete loss at 72�C.(87)

Caldeira and Peabody demonstrated thermal stability of E.coli-specific MS2 phages in storage media, from 20 to 60�C,after 2-min exposure to temperature.(88) Briers et al. identifiedthe peptidoglycan hydrolase gp181 as an important part ofphage FKZ infectivity against P. aeruginosa, and found thatits activity gradually declined from 100% to 0% when ex-posed to temperatures of 25–90�C for 60 min.(89) The study ofphage survival is well established in the dairy industry,where the presence of phages can inactivate the bacterialcultures required for fermentation. For example, the Lacto-coccus lactis phage P008 experienced 6 log10 titer loss whenheld at 72�C for 30 min(90); the heat-resistant variant phageP680 experienced a 2 to 3 log10 titer loss when held at 90�Cfor 10 min.(91) Short-term temperature exposure is not aconcern in the manufacturing of phage preparations, becausehigh-temperature exposure is unnecessary or avoidable in allprocessing steps. However, temperature excursions duringtransport or administration may need to be considered.

pH

The pH is an important formulation parameter that iscommonly controlled in feedstock preparation for furtherprocessing steps such as freeze-drying. In general, phages arewell adapted to pH ranges typically encountered in livingsystems. Four P. aeruginosa-specific phages were found toachieve 100% survival at pH 7 and 80–100% survival at pH 9,but declined in phage activity when pH was decreased to1.5.(92) However, formulation pH may affect the charge car-ried on protein surfaces, resulting in an alteration to elec-trostatic and hydrophobic interactions, and a change inprotein conformation.(93) For MS2 bacteriophages, pH con-ditions close to or below the isoelectric point were shownto favor phage aggregation, associated with a greater than1 log10 loss in PFU count.(94)

Ionic strength

The ionic strength of a formulation may affect the os-motic pressure on bacteriophages in a suspension; if thedifference in osmotic pressure inside and outside the phageis too great, the phages may be destroyed by osmoticshock.(95) This may be of particular concern if the ionicstrength changes rapidly, for example, during removal ofwater in drying processes. The peptidoglycan hydrolase

gp181 used in phage FKZ infectivity maintained optimalactivity at a pH of 6.2 and ionic strength of 140 mM; vari-ation in pH and ionic strength in either direction (pH 5–8,20–320 mM) only produced a significant decline in enzy-matic activity.(89) Golshahi et al. observed a 74% drop inKS4-M bacteriophage titer when prepared in a hypotonicsuspension (105 mOsm), compared against an isotonic (282–290 mOsm) suspension.(96)

Interfacial adsorption

When presented with an interface, e.g., during filtration orin the process of dry particle formation, proteins may unfoldto assume the conformation of lowest energy; its hydro-phobic regions are exposed to the hydrophobic phase (air ororganic solvent) and its hydrophilic regions (charged andpolar groups) to the hydrophilic phase (aqueous or polarsolvent). This has ramifications for the design of a stablebacteriophage formulation, where the phages may need to beprotected from the liquid–air interface. Pharmaceutical for-mulations often use nonionic surfactants to prevent interfa-cial adsorption of proteins.(97)

In addition to thermal and chemical stresses, phages maybe susceptible to damage by mechanical stresses inducedduring processing. In particular, shear stress from high-speed mixing, filtration, and centrifugation is a potentialsource of denaturation.(98) Short-term and long-term stresson bacteriophages during processing and storage apply tophages regardless of their mode of delivery, and standardtechniques for stabilization such as cryoprotection and de-siccoprotection may be applied. For respirable dosage forms,additional criteria must be met. The final dosage form mustbe suitable for efficient inhalation and delivery to the ap-propriate disease target.

Summary

Formulation of stable phage dosage forms appears feasi-ble, given that over-the-counter phage preparations are al-ready available. Formulation variables to consider includepH, temperature, osmotic pressure, ionic strength, and me-chanical stress. However, formulation of biological activesinto a dosage form suitable for respirable delivery remainsan active area of research.

Phage Delivery

The efficacy of phage delivery to an infected respiratorytract is reliant upon a number of factors common to respi-rable dosage forms. Two significant factors are their aero-dynamic behavior under inhalational airflow and theintended target for delivery.

Aerodynamic particle characteristics

Aerodynamic particle diameter is often used in in vitroaerosol studies to estimate regional deposition in the respi-ratory tract. The actual deposition pattern of particles in theairways is obviously also affected by device and patientfactors, such as the breathing profile and individual airwaygeometry. As depicted in Figure 4, for typical breathingpatterns and normal airways, it is generally accepted thatparticles with an aerodynamic diameter between 1 and 5 lmare respirable, whereas most particles larger than 5 lm are

8 HOE ET AL.

expected to deposit in the extrathoracic airways, and parti-cles smaller than 1 lm (but larger than 100 nm) are liable tobe lost in exhalation.(99) From aerodynamic particle diametermeasurements, the parameters of fine particle dose (mass ofdelivered particles smaller than 5 lm), mass median aero-dynamic diameter (MMAD; diameter at which 50% of thedelivered mass is smaller than this value), and geometricstandard distribution can be used along with delivered doseto predict the amount of aerosol delivered into the lung andthe delivery efficiency.(100)

Delivery target

While in vitro studies of aerosol particle size distributionoften attempt to correlate fine particle dose to lung deposi-tion, the architecture and airways surface liquid composi-tion of an acute or chronically diseased lung weakensthis in vitro–in vivo correlation further. If we were to take CFas an example, features of the disease, such as bronchiec-tasis, peribronchial thickening, mucus plugging, and air-trapping,(101) would be expected to alter the aerodynamicprofile of the pulmonary system, as shown in Figure 4, notonly due to morphological changes, but also as a result ofchanges in inhalation profile.

The results of chest radiography studies in CF patientsshow a more complicated range of disease presentation,making it difficult to mathematically model the diseasedrespiratory system.(100) Kaza et al. identified diffuse bilateraldisease from chest radiographs in 62% of the adult patientsstudied (n = 68), compared with 28% with predominant up-per lobe disease and 7% with lower lobe disease.(102) Incomparison, a small study of 17 children younger than 4years of age found six patients with the greatest disease inthe right upper lobe and another six in the right lower lobe,but the left upper lobe was also qualitatively categorized ashaving the least amount of disease in six subjects.(103)

Another study of 60 children (aged 6–10 years of age) foundbronchiectasis and mucous plugging to be distributed simi-larly between the left and right, and upper and lower,lobes.(104)

Nevertheless, numerical models have been devised to assistin vitro prediction of aerosol deposition in CF patients. In oneexample, deposition of coarse particles ( > 5 lm MMAD) by im-paction was predicted to be two to three times higher in the largeapical and basal airways (generations 1 through 12) of CF pa-tients than in those of healthy subjects.(105) However, to keep themodel workable, assumptions were made in their simulation:disease was assumed to progress from the small to large airways,sedimentation and impaction were the only deposition mecha-nisms included, and the disease was more severe at the apex.

Based on other aerosol formulations in CF, and due touncertainties in predicting deposition patterns in diseasedlungs and considerable patient variability (both intersubjectand temporal intrasubject variability), it seems advisable todesign bacteriophage dosage forms with an MMAD smallerthan approximately 5 lm for lung infections, and an MMADlarger than approximately 5 lm for infections of the extra-thoracic airways.

At the time of this publication, three formulation tech-niques have been used to manufacture a respirable phagedosage form: suspension of phages in a liquid medium,freeze-drying, and spray-drying. These formulations havethen been loaded into inhalation devices, intended for de-livery into the human respiratory tract. This section willoutline the phage formulations that have been successfullyproduced and the outcomes of the in vitro aerosol testingperformed for these formulations (see Table 3 for a summaryof the studies discussed in this section).

Suspension formulations

So far, the production of liquid suspension for directphage delivery into the respiratory tract has been for nebu-lization. To the best of our knowledge, no work has beenpublished reporting suspensions of bacteriophage in pro-pellants for metered dose inhaler delivery. Animal studiesand experimental clinical work have adopted nasal andpleural instillation and insufflation of phage suspensions as amethod of delivery.(47,106,107)

Commercial nebulizers for inhalational drug deliveryemploy different types of aerosol dispersion,(108,109) includ-ing jet (air-blast) nebulization, vibrating mesh plate nebuli-zation, ultrasonic nebulization, as well as colliding jets. Withjet nebulization, compressed air is passed through a gap in athin tube, leading to a Venturi effect that creates a pressuregradient that draws the nebulizer solution or suspension upa feed tube. Droplets are generated via a viscosity-inducedinstability,(100) whereby nonlinear growth of surface wavesand subsequent drop breakup and filtering by impaction onbaffles occur. Vibrating mesh nebulization may be carriedout by two methods: (1) a piezoelectric crystal vibrates athigh frequency, transferring the vibration to a transducerhorn that pushes the liquid through a mesh plate (‘‘passive’’);or (2) a piezoelectric element directly vibrates a mesh plate,causing micrometer-range vertical displacement of the meshthat generates droplets from the liquid drawn through(‘‘active’’).(110,111) Ultrasonic nebulization does away with themesh plate altogether, and involves the oscillation of a pie-zoelectric crystal transducer, which transfers the oscillationenergy to the liquid. Droplets are formed by the breakup ofsurface capillaries or the collapse of cavitation bubbleswithin the liquid.(109,111)

FIG. 4. Common delivery device options [metered doseinhaler (MDI), dry powder inhaler (DPI), or nebuliser], anddelivery targets in healthy and diseased lungs.

BACTERIOPHAGES FOR BACTERIAL LUNG INFECTIONS 9

Nebulization was the mode of delivery in the early reportson phage inhalation reviewed in the introduction.(19,20)

However, few device and formulation details are given, andinformation on possible titer loss during nebulization cannotbe derived from these publications. Phages have been neb-ulized to test the efficacy of biological respirator filters,alongside aerosolized bacteria, fungi, and viruses (e.g.,Balazy et al.,(112) Eninger et al.(113)). However, the focus ofthese studies is typically on particles in the most penetratingsize range (a few hundred nanometers in diameter) andnormally involves evaporated nebulized aerosols usuallyproduced by the standard Collison nebulizer, which iscommonly used in laboratory studies in occupational hy-giene. However, such submicrometer particle sizes are notoptimal for respiratory delivery due to their low efficiency ofdeposition. In addition, the Collison nebulizer does notmatch current commercial nebulizer designs in its ability todeliver therapeutic aerosols to the human respiratory tract.For example, Liu et al. used the six-jet Collison nebulizer toaerosolize D29 phages (for M. tuberculosis).(114) With an initialphage titer of 108 PFU/mL, the phage suspension wasaerosolized at 10 L/min, into a chamber for 10 min, beforethe aerosol was sampled at 12.5 L/min for 5 min in a liquidimpinger. Regardless of the suspension medium (PBS orsaline), the sampling medium (PBS, saline, or deionizedwater), or relative humidity in the chamber (25, 55, or 85%),there was a 2 to 3 log10 titer loss upon collection of theaerosolized phage, supporting the conclusion that the Colli-son nebulizer may not be the best choice for phage lungdelivery.(114)

A study by Golshahi et al. reported on nebulizer suspen-sions designed with the intent to deliver phages to the re-spiratory tract.(96) The phage selected for the study was KS4-M, which is selective for one strain of the highly antibiotic-resistant Bcc. KS4-M was prepared to an initial phage titer of2.15 – 1.63 · 108 PFU/mL. Two commercial nebulizers wereused in this study: the Pari LC Star jet nebulizer, and the ParieFlow vibrating mesh nebulizer.(96) The eFlow system is ofparticular interest, given that it is has been suggested that itsactive vibration nebulization subjects the liquid to less shearstress,(115) a feature that could potentially reduce phage titerloss from nebulization. Each nebulizer was loaded with2.5 mL of the KS4-M suspension and aerosolized under asimulated inhalation profile generated by a breathing ma-chine (tidal volume = 800 mL, respiratory rate = 14 breaths/min, duty cycle = 0.5). No significant difference in total neb-ulizer output was found between the eFlow and LC Starsystems, with both producing about 108 PFU. This equates toless than 1 log10 titer loss, which is an ideal outcome for arespiratory delivery method; although the vibration meshnebulizer did not appear to give an advantage in phagesurvival, both nebulizers demonstrate a clear advantage overthe Collison nebulizer used by Eninger et al.(113) There werealso no significant differences between the two nebulizers forthe KS4-M droplet size distributions, with a mass meangeometric diameter of 5–6 lm.

The distribution of these nebulized droplets in the lungwas predicted by the incorporation of particle size distribu-tion and breathing pattern data into a numerical respiratorytract deposition model. Alveolar deposition was approxi-mately 3 · 107 PFU for both systems. These distribution datawere then used in a follow-on model of airway surface liquid

Ta

bl

e3.

Ae

ro

so

lP

ha

ge

Fo

rm

ul

at

io

nS

tu

die

sD

isc

usse

din

‘‘P

ha

ge

De

liv

er

y’’

Ref

eren

ceP

hag

eF

orm

ula

tion

Sta

bili

tyD

eliv

ery

effi

cen

cy

Liu

etal

.(11

4)

D29

(M.

tube

rcu

losi

sp

hag

e)S

usp

ensi

on

insi

x-j

etC

oll

iso

nn

ebu

lize

r—

2–3

log

10

tite

rlo

ssaf

ter

neb

uli

zati

on

Go

lsh

ahi

etal

.(96

)K

S4-

M(B

CC

ph

age)

Su

spen

sio

nin

jet

and

vib

rati

ng

mes

hn

ebu

lize

rs—

<1

log

10

tite

rlo

ssaf

ter

neb

uli

zati

on

Wal

bec

k(1

36

)E

.co

liO

157:

H7-

spec

ific

ph

ages

Sp

ray

-dri

edp

ow

der

7lo

g1

0ti

ter

loss

afte

rsp

ray

-dry

ing

;v

ery

hig

hin

let

tem

per

atu

re(2

00–2

20�C

)an

do

utl

ette

mp

erat

ure

(90–

130�

C)

use

d

Go

lsh

ahi

etal

.(84

)K

S4-

M(B

CC

ph

age)

;F

KZ

(P.

aeru

gin

osa

ph

age)

Ly

op

hil

ized

po

wd

er(w

ith

60:4

0%

w/

wla

cto

se:l

acto

ferr

inex

cip

ien

ts)

Aft

erly

op

hil

izat

ion

,2

log

10

tite

rlo

ss(K

S4-

M),

1lo

g1

0ti

ter

loss

(FK

Z);

afte

r3

mo

nth

sst

ora

ge

at4�

Co

r21

�C,

no

tite

rlo

ssfo

rK

S4-

Mo

rF

KZ

1.2

log

10

tite

rlo

ss(K

S4-

M),

0.84

log

10

tite

rlo

ss(F

KZ

)af

ter

dis

per

sio

nfr

om

Aer

oli

zer

DP

Iat

60L

/m

inA

lfad

hel

etal

.(12

8)

S.

aure

us-

spec

ific

ph

age

Ly

op

hil

ized

po

wd

er(w

ith

HP

MC

and

man

nit

ol

exci

pie

nts

)A

fter

lyo

ph

iliz

atio

n,

1lo

g1

0ti

ter

loss

;re

frig

erat

ion

at4�

Cre

sult

edin

1lo

g1

0ti

ter

loss

afte

r1

mo

nth

,3

log

10

tite

rlo

ssaf

ter

12m

on

ths

Mat

ink

ho

oet

al.(1

37

)K

S4-

M(B

CC

ph

age)

;F

K2/

D3

(P.

aeru

gin

osa

ph

ages

)S

pra

y-d

ried

po

wd

er(w

ith

76:1

9%

w/

wtr

ehal

ose

:leu

cin

ep

lus

exci

pie

nts

)<

1lo

g1

0ti

ter

loss

afte

rsp

ray

dry

ing

;<

0.15

log

10

tite

rlo

ssaf

ter

3m

on

ths

70%

lun

gd

ose

frac

tio

naf

ter

dis

per

sio

nfr

om

Aer

oli

zer

DP

Iat

60L

/m

in

10 HOE ET AL.

in the lungs to estimate KS4-M concentration within thedifferent lung generations, dependent on mucus productionand velocity. For both the LC Star and eFlow nebulizers,concentration decreased with conducting airway generation,from 1.9–2.2 · 107 PFU/mL (generation 1) to 6.0–7 · 106

PFU/mL (generation 13). It should be noted that althoughthese deposition predictions are promising, the numericalmodels are based on healthy patient lung architecture.

Dry powder formulations

In designing an inhalable dry powder formulation, animportant factor for consideration is the ability of the deliv-ery device to entrain and deaggregate the powder, whichrequires a dispersible powder. Most dry powder inhalers(DPIs) are of the passive type, requiring the patient’s inha-lation effort to generate a sufficient air flow rate to dispersethe dry powder into the respiratory tract.

Although milling is commonly used to produce powdersfor inhaled aerosol formulations, destructive forces in millsmay result in reduced survival of phage formula-tions.(84,116,117) Instead, lyophilization and spray-drying havebeen used to produce dry powder formulations intended forinhalation therapy. To the best of our knowledge, only twopublished articles have extended bacteriophage dry powderformulation studies as far as in vitro aerosol testing: oneusing a lyophilized dosage form and the other a spray-driedpowder.

Lyophilization (freeze-drying). Lyophilization has longbeen used for the preservation of commercial protein for-mulations, including vaccines, antihemophilic factors, hu-man growth hormone, and interferons.(118) Lyophilizationhas also been used to preserve phages; however, phagesexhibit different stabilities (short- and long-term) dependingon the formulation contents and method of lyophiliza-tion,(119–121) and there are few data specifically dealing withlyophilization of phages infecting problematic respiratorybacterial species such as P. aeruginosa and the BCC.

Traditional lyophilization involves three stages: freezing,primary drying, and secondary drying. During the freezingstage, the solvent crystallizes amongst solid amorphous orcrystalline solute. Solid solvent sublimation occurs duringprimary drying, and moisture desorption during secondarydrying, leaving a solid porous ‘‘cake’’ of the solute.(122) Ex-cipients such as polyethylene glycol (PEG), trehalose, man-nitol, and sucrose are commonly added as cryoprotectants,protein stabilizers, and bulking agents, to give the cakepharmaceutically acceptable physical properties.(122,123) Re-lated to traditional lyophilization, spray freeze-drying,whereby primary freezing occurs upon spraying into coldliquid, has been proposed.(124) In addition, atmosphericspray freeze-drying, where primary freezing occurs uponspraying into cold gas in order to ease commercial scale-upissues, has been used to prepare aerosol powders of bacte-ria(125) with 90% survival rates in the prepared aerosolpowder.

Using traditional lyophilization, Golshahi et al. carriedout a study on freeze-dried phage formulations designedfor respiratory delivery.(84) The phages KS4-M (for a Bccstrain) and FKZ (for P. aeruginosa) were each formulatedwith lactose and lactoferrin. Lactose served as a carrier and

bulking agent.(84) Lactoferrin, a protein abundant in airwaysurface liquid, has been shown to inhibit P. aeruginosa biofilmformation.(126,127) In addition, lactose and lactoferrin wereselected as cryoprotectants and stabilizers for the phages. A60:40 % w/w ratio of lactose to lactoferrin was selected onthe basis of its optimal aerodynamic particle size distributionand protection in preliminary studies. Viability tests postlyophilization demonstrated a 2 log10 titer loss for KS4-Mand a 1 log10 loss for FKZ. The authors also reported nofurther phage loss after a milling procedure in a mixer mill(without the use of beads) to deagglomerate the lyophilizedpowders.

The lyophilized powders were also subjected to in vitroaerosol testing; 10 mg of powder was dispersed froman Aerolizer DPI into the Alberta idealized throat and filterat a vacuum air flow rate of 60 L/min, where the mass ofpowder recovered from the filter was representative of re-spirable dose. The results showed an acceptable titer dropfrom capsule dose to respirable dose, with the maximumloss observed being 1.2 log10 for KS4-M, and 0.84 log10 forFKZ. Respirable doses of 3.4 – 2.5 · 106 for KS4-M and1.9 – 0.6 · 107 for FKZ were achieved.

The phage powders were stored at 21 – 2% relative hu-midity and temperatures of 4�C and 21�C for 3 months aspart of a short stability study. There was no evidence of titerloss for either KS4-M or FKZ, indicating that phage activitywas preserved. Long-term stability results were not reported.

Alfadhel et al. did perform a 12-month phage stabilitystudy on a formulation designed for nasal delivery.(128) In-cluded in the formulation besides an S. aureus-specific phagewas hydroxypropylmethylcellulose (HPMC), to alter vis-cosity and increase residence time in the nasal cavity, andmannitol, as a lyoprotectant and bulking agent. Active phagetiter was decreased by 1 log10 after lyophilization. Duringstorage of the freeze-dried powder at 4�C, there was a further1 log10 loss after 1 month, another 1 log10 loss after 2 months,and a total of 3 log10 loss from the beginning of refrigerationto the end of the 12-month period.(128) Ultimately, the ac-ceptability of this magnitude of titer loss depends on itsvariability, the necessary delivered dosage, and whether aformulation with sufficiently high initial titer can be practi-cably prepared to offset the loss.

Furthermore, the addition of mannitol did not appear toaffect phage survival, with the formulations containingHPMC (1 or 2% w/v) as the only excipient performingsimilarly to those with both HPMC and mannitol. A man-nitol-only phage formulation was found to exhibit 6 log10

titer loss just after lyophilization. These results pointed to-ward HPMC acting to maintain protein conformation, andthe authors suggested the stability of amorphous HPMC tobe crucial (the glass transition temperature for lyophilizedHPMC was approximately 187�C).(128) Although the formu-lations tested in this study were intended for nasal delivery,they demonstrate that high-molecular-weight polymers mayhave a future role in maintaining phage activity during ly-ophilization and storage.

Spray-drying. Spray-drying is an established techniquefor the manufacture of pharmaceutical products.(129) Asshown in Figure 5, a liquid feedstock is atomized, typicallyusing atomization gas in a twin-fluid nozzle, and mixed withdrying gas in a drying chamber. The atomized droplets

BACTERIOPHAGES FOR BACTERIAL LUNG INFECTIONS 11

rapidly evaporate, and dry particles with a respirable parti-cle diameter are produced, separated from the gas stream,e.g., in a cyclone, and collected.

When drying biological material, the design of the spray-drying process(130,131) must be conducted in a way thatminimizes stresses, including shear, temperature, and des-iccation stress.(132) Low-temperature spray-drying processesfor heat-sensitive biological pharmaceutical ingredients havebeen developed.(133–135)

Walbeck submitted a patent application detailing the useof a rotary atomization spray-dryer system for the produc-tion of E. coli O157:H7-specific phages in dry powder.(136)

The process used an inlet temperature of 200–220�C and anoutlet temperature of 90–130�C. The authors predicted thepowders to contain 2–4 · 109 PFU/mL; the actual phage titerof the spray-dried powders was 1.1–1.8 · 102 PFU/mL, a 7log10 titer loss.(136) Given that existing literature has estab-lished that some phages are thermally labile at temperaturesbelow 100�C (as described above), it is unsurprising thatsuch high outlet temperatures would result in significant ti-ter losses.

Matinkhoo et al. presented a study on spray-dried bac-teriophage powder formulated for inhalation from aDPI.(137) The authors selected the Bcc phage KS4-M andthe P. aeruginosa phage cocktail FKZ/D3 for study. Eachphage or phage cocktail was formulated with trehalose forprotection against thermal stress and desiccation, andleucine as a dispersibility enhancer in the ratio 76:19, withother excipients in the remaining 5%. Given the consid-erations to be made about phage inactivation from thermalstress, a relatively low spray-dryer outlet temperature of40–45�C was selected. A vibrating mesh atomizer with4 lm mesh orifice diameter was used to produce 7 lmdroplets.(137)

The authors reported less than 1 log titer loss after spray-drying for all tested phages, regardless of formulation com-position.(137) The FKZ/D3 cocktail was particularly resilient,with less than 0.5 log10 titer loss in all cases. These results notonly indicate that inactivation by thermal stress was largelyprevented, but also that the other factors for inactivation, likeshear stress or desiccation stress, were not significant influ-ences for the chosen formulations. Also reported was a lessthan 0.15 log10 titer loss for FKZ/D3 spray-dried powderafter 3 months of refrigerated storage. However, as with thefreeze-drying studies, long-term stability results were notreported, but would be necessary to advance the phageformulations into clinical trials.(137)

Matinkhoo et al. also subjected the spray-dried powders toin vitro aerosol testing, using an Aerolizer DPI and the same

methodology as described for the lyophilized powderproduced by Golshahi et al.,(84) but with 25–28 mg of powderloaded into the capsule.(137) The respirable dose was *70%of loaded powder mass, regardless of phage type. Phagetiters for the respirable dose were *107 PFU for FKZ/D3,and *108 PFU for KS4-M. Separate particle size measure-ments with the Andersen Cascade Impactor showed theKS4-M and FKZ/D3 in leucine-trehalose-casein formulationsto have an MMAD of < 3 lm, suggesting that the powder ishighly dispersible and capable of reaching the smaller air-ways.(137) An efficacious dose for local phage treatment oflung infections has not been established, but the titers re-ported in this study are two to three orders of magnitudehigher than the minimum titer in commercial phage prepa-rations for P. aeruginosa infections distributed in Georgia.(85)

Summary

The feasibility of efficiently delivering phages to the lunghas been demonstrated using different processing and deviceoptions. More work in developing a stable respirable dosageform is needed, and long-term stability of such dosage formsstill needs to be evaluated.

Reports on Safety and Efficacy in Animals

The study of aerosol phage therapy in animals has been arelatively recent addition to the body of research beingconducted in phage therapy, and further studies need to beconducted. The aerosol phage therapy animal studiesreviewed here (Carmody et al.,(47) Debarbieux et al.,(138)

Morello et al.,(106) Alemayehu et al.(59)) all follow a similarprotocol. Mice are infected and treated via an intranasal in-stillation of the liquid. Intranasal inhalation requires theliquid containing either the bacteria or phage to be drippedonto the nares of the mouse, and from there the droplets ofthe liquid are inhaled into the lungs.

Although intranasal instillation is a much easier deliveryroute to use in animal studies because there are no additionalaerosol generators or delivery systems required, it should benoted that it does not accurately mimic aerosol drug delivery inclinical environments, and as such it is difficult to extrapolatethe results of these studies to a prediction of aerosol therapyefficacy. Indeed, a previous study has shown that intranasalinstillation of bacteria does not mimic aerosol particle deposi-tion within the lungs of mice: Halperin et al. demonstrated thatmice given Bordetella pertussis respiratory infections via twomethods, either intranasal inhalation or a whole-body expo-sure chamber, had very different bacterial lung counts.(139) Themice receiving intranasal instillation showed a far higher de-gree of variability in bacterial load in comparison with thosereceiving aerosolized bacteria (a 1,000-fold variability as com-pared with a five-fold variability, respectively). The mice re-ceiving intranasal instillation also showed a far greatervariability in bacterial distribution within the lungs (right lung,43–84%; left lung, 16–57%) in comparison with those receivingaerosols via the whole-body exposure chamber (right lung, 60–68%; left lung, 32–40%).(139) This may also translate into highervariability when treating mice in phage therapy studies, and itwould be of interest to see additional studies performed using awhole-body exposure chamber or nose-only inhalation cham-ber, in order to observe the effect of aerosol phage deliverymethod on treatment efficacy.

FIG. 5. Schematic of a spray-dryer for respirable powders.

12 HOE ET AL.

Debarbieux et al. demonstrated the efficacy of aerosolphage therapy via intranasal instillation for treating acuterespiratory infections using a bioluminescent strain of P.aeruginosa PAK.(138) Provided that the phage titer was highenough and treatment was given soon enough, the phagetreatment proved effective. Untreated mice all died within48 hr of infection, whereas those treated 2 hr after infectionwith a multiplicity of infection (MOI) of 10 all survived to theend of the trial (12 days). Twenty-four hours after treatment,the bacterial load in treated mice was already 6 log10 lowerthan in untreated mice; 48 hr after treatment, the inflamma-tion levels in the phage-treated mice (as indicated by tumornecrosis factor-a and interleukin-6) had decreased to baselinelevels, whereas levels remained high in untreated mice.(138)

Morello et al. tested the efficacy of phage therapy via in-tranasal instillation in treating a P. aeruginosa strain isolatedfrom a CF patient.(106) Mice were treated 2 hr after infectionwith a high enough phage titer to provide a 95% survivalrate (MOI 100). These mice survived to the end of the 16-daytrial as opposed to untreated mice, which had 0% survival by2 days post infection. In addition to survival and an ap-proximately 3 log10 reduction in bacterial lung titer 20 hrafter infection, treated mice also had significantly reducedinflammation and significantly reduced lung damage (asdemonstrated through histology). Interestingly, immuno-histochemistry performed 20 hr after infection revealed thatuntreated mice had bacteria distributed through multipleareas of the lung, including the macrophages, extracellularspace, and alveoli, whereas treated mice were found to havefew bacteria remaining in the lungs, and these were mainlylocalized in lung macrophages.(106) This demonstrates thatthe immune system works in conjunction with the action ofphages to remove bacteria from the lungs.

Debarbieux et al. and Morello et al. both demonstrated thatphages could protect mice from P. aeruginosa infection. Allmice receiving phage either 1 day (Debarbieux et al.) or 4days (Morello et al.) prior to infection were protected fromdeath.(106,138) Although this effect may not be as clinicallysignificant as infection treatment, it demonstrates the efficacyof phage usage to prevent infection.

Additionally, Alemayehu et al. demonstrated that a phagecocktail of FNH-4 and FMR299-2 delivered 2 hr after infec-tion via intranasal instillation was able to clear P. aeruginosarespiratory infections in mice.(59) Two separate biolumines-cent strains of P. aeruginosa (MR299 and NH57388A) weredelivered intranasally, and the bioluminescence within thelungs was measured at 2-hr intervals up to 8 hr after infec-tion. The luminescence within the lungs (which is propor-tional to bacterial concentration) showed a significantdecrease in phage-treated mice, whereas untreated miceshowed a three-fold increase in luminescence intensity.(59)

Carmody et al. studied phage treatment via intranasalinstillation in a respiratory infection of B. cenocepaciaAU0728.(47) Phages were administered 24 hr after infectionvia either an intranasal or intraperitoneal route. The bacterialload was determined within the lungs of all mouse groups48 hr after treatment. In contrast to the three studiesreviewed above (Debarbieux et al.,(138) Morello et al.,(106)

Alemayehu et al.,(59)), phage delivered intranasally did notproduce a significant reduction in bacterial load within thelungs (approximately 1 log10 reduction), whereas phagedelivered intraperitoneally did significantly reduce the

bacterial load (approximately a 2 log10 reduction), eventhough a higher phage titer was isolated from mice treatedintranasally. The authors suggest that these results indicatethat phage therapy in lung infections is more effective whenadministered systemically rather than topically.(47) There areseveral reasons why these results may have been obtained.With respect to the reference described above (Halperinet al.(139)), intranasal phage application does not give phageaerosol deposition in the lungs. As we have observed the lossof aerosolized phages out of the lung into the peritoneum(unpublished data), it is not inconceivable that phages cancross from the peritoneum into the lungs. In this case, itappears that peritoneal injection of high titers of phages wasmore effective than the ineffective intranasal deposition ofphages on the mouse nares.

In either case, the 1 to 2 log10 drop in bacterial numbersobserved by Carmody et al.(47) may not be an overly effectivetreatment if the bacterial load is high, and the load is notreduced far enough for the human immune system to dealwith the remainder of the infection. One could argue that, ineither condition, phage therapy in this study was not effi-cacious. It is also interesting that more than one phage wasnot tested, as we have observed notable differences in phageactivity in vivo, as compared with their in vitro activity. Fi-nally, due to the controversial nature of these findings, it isunfortunate that these experiments were not repeated withother bacterial species (such as P. aeruginosa) in order todetermine if intraperitoneal phage administration wouldhave a similar effect with a proven and published phagetherapy candidate.(23,138)

Summary

Animal studies of phage therapy have been limited tointranasal delivery. Although these studies have demon-strated varying levels of efficacy in reducing bacterial loadand increasing survival, work is still needed to investigatethe safety and efficacy of phage aerosol delivery into thelungs.

Reports on Safety and Efficacy in Humans

Although there have been studies investigating aerosolphage formulations (see Suspension formulations and Drypowder formulations), there are relatively few reports ofaerosol phage therapy in humans. However, case studieshave shown promising results (see Table 1). In Eastern Eur-ope, it is often applied to infections with antibiotic-resistantstrains, and typically 80–90% of cases are cured. This in-cludes phage treatment for respiratory tract infections.(23)

Slopek et al. reported treatment success between 82 and 92%for different patient groups with respiratory infections trea-ted with oral or locally administered phages.(107) Successfultreatment of a pediatric CF patient was also reported recentlyby Kvachadze et al.(140) In this case, phage cocktails wereadministered to the lung via a nebulizer. However, no con-trolled clinical trials have been performed to evaluate aerosolphage delivery to patients. Evidence for the clinical safety ofphage therapy in humans may be found in studies on oraland topical phage treatment. Four recent trials will be re-viewed (Bruttin and Brussow,(141) Merabishvili et al.,(16)

Rhoads et al.,(142) Wright et al.(69)). One of these trials (Wrightet al.) also addresses phage therapy efficacy.

BACTERIOPHAGES FOR BACTERIAL LUNG INFECTIONS 13

The first of the recent phage administration human safetytrials in the English literature was performed by Bruttin andBrussow (2005).(141) Fifteen volunteers were given either aplacebo, a low-phage titer preparation (103 PFU/mL), or ahigh-phage titer preparation (105 PFU/mL) orally on 2 con-secutive days followed by 5 days without phage adminis-tration. This was repeated for the following 2 weeks witheach patient receiving a different phage preparation or pla-cebo each week. Throughout the study, five adverse effectswere reported; however, there was no difference in thenumber of adverse effects in the placebo, low-phage titer,and high-phage titer groups, and the effects were deemed tobe unrelated to the study. Phages were never recovered fromstool samples prior to phage administration or when theplacebo was administered. However, phages were recoveredfrom stool samples in a dose-dependent manner after phageadministration, demonstrating that the phages were able tosurvive transit through the gastrointestinal system. Interest-ingly, the presence of phage T4 did not correlate with a de-crease in natural E. coli counts in the stool samples,suggesting that the phage did not affect the natural micro-flora of the volunteers, albeit as measured by numbers ofonly one bacterial species. Throughout the study, there wasno change in serum alanine aminotransferase and aspartateaminotransferase levels, which are liver enzymes that wouldhave indicated an increase in liver toxicity. There was also nodetected immune response to the phages, demonstrating thesafety of phage administration.(141)

A phase I safety trial was performed by Rhoads et al. toensure that the application of WPP-201, a phage cocktailcontaining eight lytic phages targeting S. aureus, P. aerugi-nosa, and E. coli, would not pose any health risks to patientswith venous leg ulcers.(142) Thirty-nine patients completedthe 24-week trial, with every patient receiving the samecompression therapy and wound dressings, with the excep-tion that approximately half received WPP-201 applied to-pically, whereas the controls received only a sterile salineapplication. Phage application occurred weekly for 12 weeks,while dressings were changed three times weekly for thesame duration. Patients were monitored weekly for the first12 weeks of the study, and data collected included woundsize and severity, vital signs, and adverse events, which mayor may not have been related to the phage application (ad-verse events included a variety of infections, injuries, medicalprocedures, pain, and cardiovascular issues). Wound bacte-rial counts, blood chemistry, and blood cell counts wereperformed every other week as well. Two additional as-sessments were made on weeks 16 and 24. There were nostatistically significant differences between the test andcontrol groups in either the number of adverse events ex-perienced or the frequency with which wounds healed. Thisphase I clinical study did not set out to evaluate phage ef-ficacy, but instead appraised the safety of phage application.With this objective successfully completed, the researcherscan now proceed to phase II studies, which will evaluate theefficacy of WPP-201.(142)

Merabishvili et al. developed and began evaluating BFC-1,a phage cocktail consisting of three phages that targetedP. aeruginosa and S. aureus strains isolated from burn woundsof patients in the Burn Centre of the Queen Astrid MilitaryHospital in Belgium.(16) The cocktail was sprayed onto theburn wounds of eight patients using a micromister. None of

these patients was reported to experience any adverse eventsresulting from the phage application.(16)

Chronic otitis caused by P. aeruginosa can be problematicto treat, as P. aeruginosa is highly antibiotic-resistant. Wrightet al. conducted phase I/II trials to evaluate the treatment ofthis infection using Biophage-PA, a cocktail of six phagesspecific for P. aeruginosa.(169) The 24 volunteers in this study,each with a history of chronic otitis lasting 2 to 58 years, hadP. aeruginosa infections sensitive to Biophage-PA prior toparticipation. A single dose of either Biophage-PA or placebowas applied directly into the ear of each participant. Themedian bacterial counts significantly decreased in phage-treated volunteers on days 21 and 42 (swabs were taken ondays 0, 7, 21, and 42), whereas there was no significant de-crease in placebo-treated volunteers. This study demon-strated the advantage of administering a self-replicatingtreatment, as the mean phage count isolated on days 7, 21,and 42 was 1.27 · 108 PFU, whereas the initial phage countadministered was 6 · 105 PFU. The phage persisted at the siteof the infection for an average of 23.1 days, and the phagewas cleared in cases where the infection was successfullytreated. Each case was also scored on a visual analogue scale(VAS) by the patient and the physician. Patients scored theinfection on discomfort, itchiness, wetness, and odor,whereas the physician scored the infection on inflammation,ulceration/granulation/polyps, discharge quantity, dis-charge type, and odor. When the patient VAS scores fromdays 7, 21, and 42 were averaged, there was a significantdecrease from days 0 for discomfort, itchiness, and wetness,as well as overall patient VAS scores, whereas placebo-treated volunteers did not show any significant reductions.When the physician VAS scores from days 7, 21, and 42 wereaveraged, there was a significant decrease in inflammation,ulceration/granulation/polyps, discharge type, and odor, aswell as overall physician VAS scores, whereas the placebogroup did not show any significant reductions. As with theother clinical trials reviewed, none of the adverse events re-ported during the course of the trial was deemed to be re-lated to phage treatment. This study provided sufficientevidence of phage therapy efficacy to warrant the design of aphase III clinical trial.(69)

It is interesting to note that these studies delivered a rangeof phage titers to the infected sites and, in the phase I/II trial,only one phage dose was required for treatment.(69) Thishighlights a unique difference in the pharmacokinetics ofphage therapy in contrast with traditional chemical antibi-otics. Traditional antibiotics must be administered at regularintervals as they are metabolized and excreted. In some sit-uations, similar delivery schedules for phages are used. Thisis known as passive phage therapy, because the treatmentdoes not rely on phage replication within the host bacteri-um.(143) In some situations, a single treatment is sufficientbecause phages can replicate at the site of infection, allowingfor the continued release of new antibacterial progeny. Thisis known as active phage therapy.

The bacterial density at the site of infection may also affectthe treatment strategy. Active phage therapy is effective inareas of high bacterial density, whereas passive phage ther-apy may be effective in areas of reduced bacterial density.(30)

Interestingly, mathematical modeling has demonstrated thatthe timing of phage administration is also very impor-tant.(143) As expected, delayed administration of phages may

14 HOE ET AL.

be detrimental to treatment; however, treating an infectiontoo early (e.g., low bacterial numbers) may also be detri-mental. This, again, relates to the density of bacterial hostsavailable at the site of an infection. A very low densitybacterial load may be more treatable once it has replicated tothe point at which the density is high enough for effectiveactive phage therapy. The studies reviewed above (Bruttinand Brussow,(141) Merabishvili et al.,(16) Rhoads et al.,(142)

Wright et al.(69)) demonstrate, however, that regardless of thephage delivery strategy used, phages are safe for use inphage therapy applications.

Summary

Recent trials of oral and topical phage therapy have shownthe safety of phage therapy in humans. Experimental phagetherapy has been reported to be efficacious in older studiesand in a recent phase I/II trial; however, there have not beencontrolled clinical trials for aerosol phage therapy.

Conclusion

The current experience with phage treatment of bacterialinfections indicates that it is likely that safe and efficaciousphage therapeutics can be developed. Phages have the po-tential to become a much-needed tool in the fight againstextensively antibiotic-resistant bacteria, including thosecausing respiratory infections. However, many pharmaco-kinetic questions remain open, and the ideal route of deliveryfor lung infections has not yet been determined. Althoughthere is evidence to show the safety and efficacy of phagetherapy, there is a real need for in vitro and in vivo studiesspecifically investigating aerosol phage delivery to the lung.Local administration of inhalable phages is an attractiveprospect, as it may provide advantages, in particular forinfections that are walled off from the systemic circulatorysystem, such as in CF patients.

Initial studies probing the feasibility of inhalable phagedosage forms appear promising, and compatibility withnebulization and dry powder delivery has been shown. Mostchemistry, manufacturing, and controls questions, except forlong-term storage stability, either have been addressed suc-cessfully or can be answered using the extensive develop-ment history with viral vaccine manufacturing.

The future of phage therapy in the West will depend oneconomic considerations as phage products move into moreexpensive late-stage clinical trials. Whether it will becomeattractive to industry to advance these much needed thera-peutics to the commercial stage will be decided by patent-ability, market projections, potential financial incentives bygovernmental or nonprofit organizations, and by the pre-scribed regulatory pathway for these products. Currently,there are no specific regulatory guidelines for the develop-ment and approval of bacteriophage therapeutics in Europeand North America. Specific guidelines or clarifications re-garding current applicable regulations would be desirable toremove development risks for sponsors. The regulatoryframework should take into account that bacteriophagestrains will likely have to be updated rapidly and may haveto be adjusted to locally prevalent bacterial strains,(144)

similar to the modifications required in the case of attenuatedviral influenza vaccines. For this reason, a standard formu-lation and delivery system for bacteriophages that can be

proven to be generally safe and suitable for a wide variety ofinterchangeable phage strains appears highly desirable.

Author Disclosure Statement

The authors declare that no conflicts of interest exist.

References

1. Riley JC: Rising Life Expectancy: A Global History. CambridgeUniversity Press, Cambridge; 2001.

2. Palumbi S: Evolution—humans as the world’s greatestevolutionary force. Science. 2001;293:1786–1790.

3. Hassett DJ, Korfhagen TR, Irvin RT, Schurr MJ, Sauer K,Lau GW, Sutton MD, Yu H, and Hoiby N: Pseudomonasaeruginosa biofilm infections in cystic fibrosis: insights intopathogenic processes and treatment strategies. Expert OpinTher Targets. 2010;14:117–130.

4. Mandsberg LF, Ciofu O, Kirkby N, Christiansen LE, Poul-sen H, and Hoiby N: Antiobiotic resistance in Pseudomonasaeruginosa strains with increased mutation frequency due toinactivation of the DNA oxidative repair system. Anti-microb Agents Chemother. 2009;53:2483–2491.

5. Seed KD, and Dennis JJ: Isolation and characterization ofbacteriophages of the Burkholderia cepacia complex. FEMSMicrobiol Lett. 2005;251:273–280.

6. Reinert RR: The antimicrobial resistance profile of Strepto-coccus pneumoniae. Clin Microbiol Infect. 2009;15:7–11.

7. Yong D, Toleman MA, Giske CG, Cho HS, Sundman K, LeeK, and Walsh TR: Characterization of a new metallo-b-lactamase gene, blaNDM-1, and a novel erythromycin esterasegene carried on a unique genetic structure in Klebsiellapneumoniae sequence type 14 from India. Antimicrob AgentsChemother. 2009;53:5046–5054.

8. Karthikeyan K, Thirunarayan M, and Krishnan P: Coex-istence of blaOXA-23 with blaNDM-1 and armA in clinical iso-lates of Acinetobacter baumannii from India. J AntimicrobChemother. 2010;65:2253–2254.

9. World Health Organization: Multidrug and extensivelydrug-resistant TB (M/XDR-TB). 2010 global report onsurveillance and response. Geneva, Switzerland; 2010.

10. Jassal M, and Bishai WR: Extensively drug-resistant tu-berculosis. Lancet Infect Dis. 2009;9:19–30.

11. Tomlin KL, Coll OP, and Ceri H: Interspecies biofilms ofPseudomonas aeruginosa and Burkholderia cepacia. Can J Mi-crobiol. 2001;47:949–954.

12. Coates ARM, Halls G, and Hu Y: Novel classes of antibioticsor more of the same? Br J Pharmacol. 2011;163:184–194.

13. Abedon ST, and Thomas-Abedon C: Phage therapy phar-macology. Curr Pharm Biotechnol. 2010;11:28–47.

14. Burrowes B, Harper DR, Anderson J, McConville M, andEnright MC: Bacteriophage therapy: potential uses in thecontrol of antibiotic-resistant pathogens. Expert Rev AntiInfect Ther. 2011;9:775–785.

15. Parisien A, Allain B, Zhang J, Mandeville R, and Lan CQ:Novel alternatives to antibiotics: bacteriophages, bacterialcell wall hydrolases, and antimicrobial peptides. J ApplMicrobiol. 2008;104:1–13.

16. Merabishvili M, Pirnay J, Verbeken G, Chanishvili N,Tediashvili M, Lashkhi N, Glonti T, Krylov V, Mast J, VanParys L, Lavigne R, Volckaert G, Mattheus W, Verween G,De Corte P, Rose T, Jennes S, Zizi M, De Vos D, and Va-neechoutte M: Quality-controlled small-scale production ofa well-defined bacteriophage cocktail for use in humanclinical trials. PLoS One. 2009;4:e4944.

BACTERIOPHAGES FOR BACTERIAL LUNG INFECTIONS 15

17. Housby JN, and Mann NH: Phage therapy. Drug DiscovToday. 2009;14:536–540.

18. Hanawalt PC: Density matters: the semiconservative rep-lication of DNA. Proc Natl Acad Sci USA. 2004;101:17889–17894.

19. Delacoste P: Considerations sur le traitement des affectionsrespiratoires banales au moyen de bacteriophages [Con-siderations on the treatment of common respiratory dis-eases by means of bacteriophages]. Rev Med SuisseRomande. 1959;79:552–563.

20. Hoeflmayr J: Inhalationstherapie mit Bakteriophagen beitherapieresistenten Infektionen [Inhalation therapy withbacteriophages for treatment-resistant infections]. For-tschritte der biologischen Aerosol-Forschung in den Jahren1957–1961 [Advances in Biological Aerosols Research in theYears 1957–1961]. Stuttgart, Germany; pp. 403–409, 1962.

21. Abedon ST, Kuhl SJ, Blasdel BG, and Kutter EM: Phagetreatment of human infections. Bacteriophage. 2011;1:66–85.

22. Gorski A, Miedzybrodzki R, Borysowski J, Weber-Dab-rowska B, qobocka M, Fortuna W, Letkiewicz S, ZimeckiM, and Filby G: Bacteriophage therapy for treatment ofinfections. Curr Opin Investig Drugs. 2009;10:766–774.

23. Kutateladze M, and Adamia R: Phage therapy experienceat the Eliava Institute. Med Mal Infect. 2008;38:426–430.

24. Buckling A, and Rainey PB: Antagonistic coevolution be-tween a bacterium and a bacteriophage. Proc R Soc Lond B.2002;269:931–936.

25. Thiel K: Old dogma, new tricks—21st century phage ther-apy. Nat Biotechnol. 2004;22:31–36.

26. Abedon ST, Herschler TD, and Stopar D: Bacteriophagelatent-period evolution as a response to resource avail-ability. Appl Environ Microbiol. 2001;67:4233–4241.

27. Bade EG: Asymmetric transcription of bacteriophage Mu-1.J Virol. 1972;10:1205–1207.

28. Carlton RM: Phage therapy: past history and future pros-pects. Arch Immunol Ther Exp. 1999;47:267–274.

29. Hadas H, Einav M, Fishov I, and Zaritsky A: BacteriophageT4 development depends on the physiology of its hostEscherichia coli. Microbiology. 1997;143:179–185.

30. Levin BR, and Bull JJ: Population and evolutionary dy-namics of phage therapy. Nat Rev Microbiol. 2004;2:166–173.

31. Levin BR, Stewart FM, and Chao L: Resource-limitedgrowth, competition, and predation: a model and experi-mental studies with bacteria and bacteriophage. Am Nat.1977;111:3–24.

32. Wolfson J, Dressler D, and Magazin M: Bacteriophage T7DNA replication: a linear replicating intermediate. ProcNatl Acad Sci USA. 1972;69:499–504.

33. Labrie SJ, Samson JE, and Moineau S: Bacteriophage re-sistance mechanisms. Nat Rev Microbiol. 2010;8:317–327.

34. Kelly D, McAuliffe O, Ross RP, O’Mahony J, and Coffey A:Development of a broad-host-range phage cocktail forbiocontrol. Bioeng Bugs. 2011;2:31–37.

35. Pouillot F, Bois H, and Iris F: Genetically engineered viru-lent phage banks in the detection and control of emergentpathogenic bacteria. Biosecur Bioterror. 2010;8:155–169.

36. Coutinho HDM, Falcao-Silva VS, and Goncalves GF: Pul-monary bacterial pathogens in cystic fibrosis patients andantibiotic therapy: a tool for the health workers. Int ArchMed. 2008;1:24.

37. Campbell A: The future of bacteriophage biology. Nat RevGenet. 2003;4:471–477.

38. Oppenheim AB, Kobiler O, Stavans J, Court DL, and Ad-hya S: Switches in bacteriophage lambda development.Annu Rev Genet. 2005;39:409–429.

39. Boyd EF, and Brussow H: Common themes among bacte-riophage-encoded virulence factors and diversity amongthe bacteriophages involved. Trends Microbiol. 2002;10:521–529.

40. Karaolis DKR, Somara S, Maneval DR, Johnson JA, andKaper JB: A bacteriophage encoding a pathogenicity island,a type-IV pilus and a phage receptor in cholera bacteria.Nature. 1999;399:375–379.

41. Waldor MK, and Mekalanos JJ: Lysogenic conversion by afilamentous phage encoding cholera toxin. Science. 1996;272:1910–1913.

42. Griffiths AJF, Miller JH, Suzuki DT, Lewontin RC, andGelbart WM: An Introduction to Genetic Analysis, 7th ed.W.H. Freeman and Company, New York, NY; pp. 229–230,348–350, 2000.

43. Gill JJ, Summer EJ, Russell WK, Cologna SM, CarlileTM, Fuller AC, Kitsopoulos K, Mebane LM, ParkinsonBN, Sullivan D, Carmody LA, Gonzalez CF, LiPumaJJ, and Young R: Genomes and characterization of pha-ges Bcep22 and BcepIL02, founders of a novel phagetype in Burkholderia cenocepacia. J Bacteriol. 2011;193:5300–5313.

44. Lynch KH, Stothard P, and Dennis JJ: Characterization ofDC1, a broad/host/range Bcep22-like podovirus. ApplEnviron Microbiol. 2012;78:889–891.

45. Lynch KH, and Dennis JJ: Genomic analysis and modifi-cation of Burkholderia cepacia complex bacteriophages:Cangene Gold Medal Lecture. Can J Microbiol. 2012;58:221–235.

46. Meader E, Mayer MJ, Gasson MJ, Steverding D, Carding SR,and Narbad A: Bacteriophage treatment significantly re-duces viable Clostridium difficile and prevents toxin produc-tion in an in vitro model system. Anaerobe. 2010;16:549–554.

47. Carmody LA, Gill JJ, Summer EJ, Sajjan US, Gonzalez CF,Young RF, and LiPuma JJ: Efficacy of bacteriophage ther-apy in a model of Burkholderia cenocepacia pulmonary in-fection. J Infect Dis. 2010;201:264–271.

48. Lynch KH, Seed KD, Stothard P, and Dennis JJ: Inactivationof Burkholderia cepacia complex phage KS9 gp41 identifiesthe phage repressor and generates lytic virions. J Virol.2010;84:1276–1288.

49. Ramesh V, Fralick JA, and Rolfe RD: Prevention of Clos-tridium difficile-induced ileocecitis with bacteriophage.Anaerobe. 1999;5:69–78.

50. Seed KD, and Dennis JJ: Experimental bacteriophagetherapy increases survival of Galleria mellonella larvae in-fected with clinically relevant strains of the Burkholderiacepacia complex. Antimicrob Agents Chemother. 2009;53:2205–2208.

51. Capparelli R, Parlato M, Borriello G, Salvatore P, and Ian-nelli D: Experimental phage therapy against Staphylococcusaureus in mice. Antimicrob Agents Chemother.2007;51:2765–2773.

52. Matsuzaki S, Yasuda M, Nishikawa H, Kuroda M, UjiharaT, Shuin T, Shen Y, Jin Z, Fujimoto S, Nasimuzzaman MD,Wakiguchi H, Sugihara S, Sugiura T, Koda S, Muraoka A,and Imai S: Experimental protection of mice against lethalStaphylococcus aureus infection by novel bacteriophagejMR11. J Infect Dis. 2003;187:613–624.

53. Paul VD, Sundarrajan S, Rajagopalan SS, Hariharan S,Kempashanaiah N, Padmanabhan S, Sriram B, and

16 HOE ET AL.

Ramachandran J: Lysis-deficient phages as novel thera-peutic agents for controlling bacterial infection. BMC Mi-crobiol. 2011;11:195.

54. Merril CR, Biswas B, Carlton R, Jensen NC, Creed GJ, ZulloS, and Adhya S: Long-circulating bacteriophage as anti-bacterial agents. Proc Natl Acad Sci USA. 1996;93:3188–3192.

55. Rapson ME, Burden FA, Glancey LP, Hodgson DA, andMann NH: Anti-bacterial agents. Patent Application 11/963,899, December 25, 2008.

56. Brussow H, Canchaya C, and Hardt W: Phages and theevolution of bacterial pathogens: from genomic rearrange-ments to lysogenic conversion. Microbiol Mol Biol Rev.2004;68:560–602.

57. Glonti T, Chanishvili N, and Taylor PW: Bacteriophage-derived enzyme that depolymerizes the alginic acid cap-sule associated with cystic fibrosis isolates of Pseudomonasaeruginosa. J Appl Microbiol. 2010;108:695–702.

58. Leiman PG, Battisti AJ, Bowman VD, Stummeyer K,Muhlenhoff M, Gerardy-Schahn R, Scholl D, and MolineuxIJ: The structures of bacteriophages K1E and K1-5 explainprocessive degradation of polysaccharide capsules andevolution of new host specificities. J Mol Biol. 2007;371:836–849.

59. Alemayehu D, Casey PG, McAuliffe O, Guinane CM,Martin JG, Shanahan F, Coffey A, Ross RP, and Hill C:Bacteriophages jMR299-2 and jNH-4 can eliminate Pseu-domonas aeruginosa in the murine lung and on cystic fibrosislung airway cells. MBio. 2012;3:e00029–e00012.

60. Lu TK, and Collins JJ: Dispersing biofilms with engineeredenzymatic bacteriophage. Proc Natl Acad Sci USA. 2007;104:11197–11202.

61. Moradpour Z, and Ghasemian A: Modified phages: novelantimicrobial agents to combat infectious diseases. Bio-technol Adv. 2011;29:732–738.

62. Mahichi F, Synnott AJ, Yamamichi K, Osada T, and Tanji Y:Site-specific recombination of T2 phage using IP008 longtail fiber genes provides a targeted method for expandinghost range while retaining lytic activity. FEMS MicrobiolLett. 2009;295:211–217.

63. Hagens S, and Blasi U: Genetically modified filamentousphage as bactericidal agents: a pilot study. Lett Appl Mi-crobiol. 2003;37:318–323.

64. Hagens S, Habel A, von Ahsen U, von Gabain A, and BlasiU: Therapy of experimental Pseudomonas infections with anonreplicating genetically modified phage. AntimicrobAgents Chemother. 2004;48:3817–3822.

65. Westwater C, Kasman LM, Schofield DA, Werner PA, DolanJW, Schmidt MG, and Norris JS: Use of genetically en-gineered phage to deliver antimicrobial agents to bacteria:an alternative therapy for treatment of bacterial infections.Antimicrob Agents Chemother. 2003;47:1301–1307.

66. Lu TK, and Collins JJ: Engineered bacteriophage targetinggene networks as adjuvants for antibiotic therapy. ProcNatl Acad Sci USA. 2009;106:4629–4634.

67. Uchiyama J, Maeda Y, Takemura I, Chess-Williams R,Wakiguchi H, and Matsuzaki S: Blood kinetics of fourintraperitoneally administered therapeutic candidatebacteriophages in healthy and neutropenic mice. MicrobiolImmunol. 2009;53:301–304.

68. Reyes A, Semenkovich NP, Whiteson K, Rohwer F, andGordon JI: Going viral: next-generation sequencing appliedto phage populations in the human gut. Nat Rev Microbiol.2012;10:607–617.

69. Wright A, Hawkins CH, Anggard EE, and Harper DR: Acontrolled clinical trial of a therapeutic bacteriophagepreparation in chronic otitis due to antibiotic-resistantPseudomonas aeruginosa; a preliminary report of efficacy.Clin Otolaryngol. 2009;34:349–357.

70. Kutter E, De Vos D, Gvasalia G, Alavidze Z, Gogokhia L,Kuhl S, and Abedon ST: Phage therapy in clinical practice:treatment of human infections. Curr Pharm Biotechnol.2010;11:69–86.

71. Sulakvelidze A, and Barrow P. Phage therapy in animalsand agribusiness. In: E Kutter and A Sulakvelidze, (eds).Bacteriophages: Biology and Application. CRC Press, BocaRaton, FL; pp. 335–380, 2005.

72. Gill JJ, and Hyman P: Phage choice, isolation, and preparationfor phage therapy. Curr Pharm Biotechnol. 2010;11:2–14.

73. Lenherr H, and Bartsch R: Process and system for the in-dustrial scale purification of bacteriophages intended forbacteriophage therapy. Patent Application 2011/0281329A1, November 17, 2011.

74. Soothill J, Hawkins C, and Harper D: Bacteriophage-con-taining therapeutic agents. Patent Patent 7,807,149 B2, Oc-tober 5, 2010.

75. Skurnik M, Pajunen M, and Kiljunen S: Biotechnologicalchallenges of phage therapy. Biotechnol Lett. 2007;29:995–1003.

76. Boratynski J, Syper D, Weber-Dabrowska B, qusiak-Szelachowska M, Pozniak G, and Gorski A: Preparation ofendotoxin-free bacteriophages. Cell Mol Biol Lett. 2004;9:253–259.

77. Wei GL, Liu XL, Li JH, Liu Y, and Shang ZH: Endotoxinremoval in some medicines and human serum albuminsolution by affinity membranes. Sepu-zh�ongguo huaxuehuı [Chin J Chromatogr]. 2002;2:108–114.

78. Wilson MJ, Haggart CL, Gallagher SP, and Walsh D: Re-moval of tightly bound endotoxin from biological products.J Biotechnol. 2001;88:67–75.

79. Boratynski J, Gorski A, Weber-Dabrowska B, and Szermer-Olearnik B: Method of obtaining bacteriophage preparation.Patent Application 2010/0227376 A1, September 9, 2010.

80. Ulmer JB, Valley U, and Rappuoli R: Vaccine manufactur-ing: challenges and solutions. Nat Biotechnol. 2006;24:1377–1383.

81. Bujanover S: Production of bacteriophage compositions foruse in phage therapy. Patent 7,588,929 B2, September 25, 2009.

82. Clark WA: Comparison of several methods for preservingbacteriophages. Appl Environ Microbiol. 1962;10:466–471.

83. Mullan WMA: Preparation and storage of high titre lacto-coccal lysates [Web page]. 2005; Available at http://www.dairyscience.info/preparation-and-storage-of-high-titre-lactococcal-lysates.html. Accessed October 31, 2011.

84. Golshahi L, Lynch KH, Dennis JJ, and Finlay WH: In vitrolung delivery of bacteriophages KS4-M and FKZ using drypowder inhalers for treatment of Burkholderia cepacia com-plex and Pseudomonas aeruginosa infections in cystic fibrosis.J Appl Microbiol. 2010;110:106–117.

85. JSC Biochimpharm. Our products [Web page]. 2010; Avail-able at http://www.biochimpharm.ge/index.php?option=com_content&view=category&layout=blog&id=1&Itemid=2&lang = en. Accessed June 26, 2012.

86. Advanced Biophage Technologies. Products [Web Page].Available at http://advancedbiophagetechnologies.com/products.html. Accessed June 26, 2012.

87. Garcıa P, Madera C, Martınez B, Rodrıguez A, andEvaristo Suarez J: Prevalence of bacteriophages infecting

BACTERIOPHAGES FOR BACTERIAL LUNG INFECTIONS 17

Staphylococcus aureus in dairy samples and their potential asbiocontrol agents. J Dairy Sci. 2009;92:3019–3026.

88. Caldeira JC, and Peabody DS: Thermal stability of RNAphage virus-like particles displaying foreign peptides. JNanobiotechnol. 2011;9:22.

89. Briers Y, Miroshnikov K, Chertkov O, Nekrasov A, Me-syanzhinov V, Volckaert G, and Lavigne R: The structuralpeptidoglycan hydrolase gp181 of bacteriophage jKZ.Biochem Biophys Res Commun. 2008;374:747–751.

90. Muller-Merbach M, Neve H, and Hinrichs J: Kinetics of thethermal inactivation of the Lactococcus lactis bacteriophageP008. J Dairy Res. 2005;72:281–286.

91. Atamer Z, and Hinrichs J: Thermal inactivation of the heat-resistant Lactococcus lactis bacteriophage P680 in moderncheese processing. Int Dairy J. 2010;20:163–168.

92. Knezevic P, Obreht D, Curcin S, Petrusic V, Kostansjek, andPetrovic O: Phages of Pseudomonas aeruginosa: response toenvironmental factors and in vitro ability to inhibit bacterialgrowth and biofilm formation. J Appl Microbiol. 2011;111:245–254.

93. Chi EY, Krishnan S, Randolph TW, and Carpenter JF:Physical stability of proteins in aqueous solution: mecha-nism and driving forces in nonnative protein aggregation.Pharm Res. 2003;20:1325–1336.

94. Langlet J, Gaboriaud F, and Gantzer C: Effects of pH onplaque forming unit counts and aggregation of MS2 bac-teriophage. J Appl Microbiol. 2007;103:1632–1638.

95. Furiga A, Pierre G, Glories M, Aimar P, Roques C, Caus-serand C, and Berge M: Effects of ionic strength on bacte-riophage MS2 behavior and their implications for theassessment of virus retention by ultrafiltration membranes.Appl Environ Microbiol. 2011;77:229–236.

96. Golshahi L, Seed KD, Dennis JJ, and Finlay WH: Towardmodern inhalational bacteriophage therapy: nebulisation ofbacteriophages of Burkholderia cepacia complex. J AerosolMed Pulm Drug Deliv. 2008;21:351–359.

97. Manning MC, Chou DK, Murphy BM, Payne RW, andKatayama DS: Stability of protein pharmaceuticals: anupdate. Pharm Res. 2010;27:544–575.

98. Branston S, Stanley E, Ward J, and Keshavarz-Moore E: Studyof robustness of filamentous bacteriophages for industrialapplications. Biotechnol Bioeng. 2011;108:1468–1472.

99. Jaafar-Maalej C, Andrieu V, Elaissari A, and Fessi H: As-sessment methods of inhaled aerosols: technical aspectsand applications. Expert Opin Drug Deliv. 2009;6:941–959.

100. Finlay WH: Mechanics of Inhaled Pharmaceutical Aerosols: AnIntroduction. Academic Press, San Diego, CA; 2001.

101. Eichinger M, Heussel CP, Kauczor HU, Tiddens H, andPuderbach M: Computed tomography and magnetic reso-nance imaging in cystic fibrosis lung disease. J Magn ResonImaging. 2010;32:1370–1378.

102. Kaza V, Katz MF, Cumming S, Frost AE, and Safdar Z:Correlation of chest radiograph pattern with genotype, age,and gender in adult cystic fibrosis. Chest. 2007;132:569–574.

103. Davis SD, Fordham LA, Brody AS, Noah TL, Retsch-BogartGZ, Qaqish BF, Yankaskas BC, Johnson RC, and LeighMW: Computed tomography reflects lower airway in-flammation and tracks changes in early cystic fibrosis. Am JRespir Crit Care. 2007;175:943–950.

104. Brody AS, Klein JS, Molina PL, Quan J, Bean JA, and Wil-mott RW: High-resolution computed tomography in youngpatients with cystic fibrosis: distribution of abnormalitiesand correlation with pulmonary function tests. J Pediatr.2004;145:32–38.

105. Brown JS, and Bennett WD: Deposition of coarse particlesin cystic fibrosis: model predictions versus experimentalresults. J Aerosol Med. 2004;17:239–248.

106. Morello E, Saussereau E, Maura D, Huerre M, Touqui L,and Debarbieux L: Pulmonary bacteriophage therapy onPseudomonas aeruginosa cystic fibrosis strains: first stepstowards treatment and prevention. PLoS One. 2011;6:e16963.

107. Slopek S, Weber-Dabrowska B, Dabrowski M, and Ku-charewicz-Krukowska A: Results of bacteriophage treat-ment of suppurative bacterial infections in the years 1981–1986. Arch Immunol Ther Exp. 1987;35:569–583.

108. Finlay WH: Pharmaceutical aerosol sprays for drug deliv-ery to the lungs. In: N Ashgriz, (ed). Handbook of Atomiza-tion and Sprays: Theory and Applications. Springer, Boston,MA; pp. 899–908, 2011.

109. Knoch M, and Finlay WH: Nebulizer technologies. In: MJRathbone, J Hadgraft, MS Roberts, and ME Lane, (eds).Modified-Release Drug Delivery Technology, 2nd ed. InformaHealthcare, New York, NY; pp. 613–622, 2008.

110. Eslamian M, and Ashgriz N: Swirl, T-jet and vibrating-mesh atomizers. In: N Ashgriz, (ed). Handbook of Atomiza-tion and Sprays: Theory and Applications. Springer Press,Boston, MA; pp. 755–773, 2011.

111. Xu Z, and Hickey AJ: The physics of aerosol droplet andparticle generation from inhalers. In: HDC Smyth and AJHickey, (eds). Controlled Pulmonary Drug Delivery. Springer,New York, NY; 75–100, 2011.

112. Ba1azy A, Toivola M, Adhikari A, Sivasubramani SK, Re-ponen T, and Grinshpun SA: Do N95 respirators provide 95%protection level against airborne viruses, and how adequateare surgical masks? Am J Infect Control. 2006;34:51–57.

113. Eninger RM, Hogan CJ, Biswas P, Ashikari A, Reponen T,and Grinshpun SA: Electrospray versus nebulization foraerosolization and filter testing with bacteriophage parti-cles. Aerosol Sci Technol. 2009;43:298–304.

114. Liu K, Wen Z, Li N, Yang W, Wang J, Hu L, Dong X, Lu J,and Li J: Impact of relative humidity and collection mediaon mycobacteriophage D29 aerosol. Appl Environ Micro-biol. 2012;78:1466–1472.

115. Lass JS, Sant A, and Knoch M: New advances in aerosoliseddrug delivery: vibrating membrane nebuliser technology.Expert Opin Drug Deliv. 2006;3:693–702.

116. Nichols MEK, Stanislaus T, Keshavarz-Moore E, andYoung HA: Disruption of leaves and initial extraction ofwildtype CPMV virus as a basis for producing vaccinesfrom plants. J Biotechnol. 2002;92:229–235.

117. Sotome I, Nei D, Tsuda M, Hossen MS, Takenaka M, Oka-dome H, and Isobe S: Decontamination effect of milling by ajet mill on bacteria in rice flour. Biocontrol Sci. 2011;16:79–83.

118. Schwegman JJ, Hardwick LM, and Akers MJ: Practicalformulation and process development of freeze-driedproducts. Pharm Dev Technol. 2005;10:151–173.

119. Jonczyk E, K1ak M, Miedzybrodzki R, and Gorski A: Theinfluence of external factors on bacteriophages. Folia Mi-crobiol (Praha). 2011;56:191–200.

120. Schade AL, and Caroline L: The preparation of a polyvalentdysentery bacteriophage in a dry and stable form: I. Pre-liminary investigations and general procedures. J Bacteriol.1943;45:463–473.

121. Schade AL, and Caroline L: The preparation of a polyvalentdysentery bacteriophage in a dry and stable form: III. Sta-bility of the dried bacteriophage towards heat, humidity,age and acidity. J Bacteriol. 1944;48:243–251.

18 HOE ET AL.

122. Liu J: Physical characterization of pharmaceutical formu-lations in frozen and freeze-dried solid states: techniquesand applications in freeze-drying development. Pharm DevTechnol. 2006;11:3–28.

123. Puapermoonsiri U, Spencer J, and van der Walle CF: Afreeze-dried formulation of bacteriophage encapsulated inbiodegradable microspheres. Eur J Pharm Biopharm. 2009;72:26–33.

124. Finlay WH, Dennis JJ, Orszanska H, Seed KD, and LynchKH: Bacteriophage compositions for treatment of bacterialinfections. Patent Application 20110293671 A1, December1, 2011.

125. Wang ZL, Finlay WH, Peppler MS, and Sweeney LG:Powder formation by atmospheric spray-freeze drying.Powder Tech. 2006;170:45–52.

126. Ammons MCB, Ward LS, Dowd S, and James GA: Com-bined treatment of Pseudomonas aeruginosa biofilm withlactoferrin and xylitol inhibits the ability of bacteria to re-spond to damage resulting from lactoferrin iron chelation.Int J Antimicrob Agents. 2011;37:316–323.

127. Singh PK: Iron sequestration by human lactoferrin stimu-lates P. aeruginosa surface motility and blocks biofilm for-mation. Biometals. 2004;17:267–270.

128. Alfadhel M, Puapermpoonsiri U, Ford SJ, McInnes FJ, andvan der Walle CF: Lyophilised inserts for nasal adminis-tration harboring bacteriophage selective for Staphylococcusaureus: in vitro evaluation. Int J Pharm. 2011;416:280–287.

129. Vehring R: Expert review: Pharmaceutical particle engi-neering via spray drying. Pharm Res. 2008;25:999–1022.

130. Boraey MA, Matinkhoo S, and Vehring R: A new time andcost effective approach for the development of micropar-ticles for pulmonary drug delivery. [Abstract] Presented atRespiratory Drug Delivery Europe, Berlin, Germany, May3–8, 2011; Vol. 2, pp. 473–477.

131. Ivey J, and Vehring R: The use of modeling in spray dryingof emulsions and suspensions accelerates formulation andprocess development. Comput Chem Eng. 2010;34:1036–1040.

132. Peighambardoust SH, Golshan Tafti A, and Hesari J: Ap-plication of spray drying for preservation of lactic acidstarter cultures: a review. Trends Food Sci Technol. 2011;22:215–224.

133. Burger JL, Cape SP, Braun CS, McAdams DH, Best JA,Bhagwat P, Pathak P, Rebits LG, and Sievers RE: Stabiliz-ing formulations for inhalable powders of live-attenuatedmeasles virus vaccine. J Aerosol Med Pulm Drug Deliv.2008;21:25–34.

134. Feng AL, Boraey MA, Gwin MA, Finlay PR, Kuehl PJ, andVehring R: Mechanistic models facilitate efficient develop-ment of leucine containing microparticles for pulmonarydrug delivery. Int J Pharm. 2011;409:156–163.

135. Ohtake S, Martin RA, Yee L, Chen DX, Kristensen DD,Lechuga-Ballesteros D, and Truong-Le V: Heat-stablemeasles vaccine produced by spray drying. Vaccine. 2010;28:1275–1284.

136. Walbeck AK: Methods for drying bacteriophage and bac-teriophage containing compositions, the resulting drycompositions, and methods of use. Patent Application2009/0093041 A1, April 9, 2009.

137. Matinkhoo S, Lynch KH, Dennis JJ, Finlay WH, andVehring R: Spray-dried respirable powders containing

bacteriophages for the treatment of pulmonary infections. JPharm Sci. 2011;100:5197–5205.

138. Debarbieux L, Leduc D, Maura D, Morello E, Criscuolo A,Grossi O, Balloy V, and Touqui L: Bacteriophages can treatand prevent Pseudomonas aeruginosa lung infections.J Infect Dis. 2010;201:1096–1104.

139. Halperin S, Heifetz S, and Kasina A: Experimental respi-ratory infection with Bordetella pertussis in mice: com-parison of two methods. Clin Invest Med. 1988;11:297–303.

140. Kvachadze L, Balarjishvili N, Meskhi T, Tevdoradze E,Skhirtladze N, Pataridze T, Adamia R, Topuria T, Kutter E,Rhode C, and Kutateladze M: Evaluation of lytic activity ofstaphylococcal bacteriophage Sb-1 against freshly isolatedclinical pathogens. Microb Biotechnol. 2011;4:643–650.

141. Bruttin A, and Brussow H: Human volunteers receiv-ing Escherichia coli phage T4 orally: a safety test of phagetherapy. Antimicrob Agents Chemother. 2005;49:2874–2878.

142. Rhoads DD, Wolcott RD, Kuskowski MA, Wolcott BM,Ward LS, and Sulakvelidze A: Bacteriophage therapy ofvenous leg ulcers in humans: results of a phase I safetytrial. J Wound Care. 2009;18:240–243.

143. Payne RJH, and Jansen VAA: Pharmacokinetic principlesof bacteriophage therapy. Clin Pharmacokinet. 2003;42:315–325.

144. Verbeken G, Pirnaym JP, De Vos D, Jennes S, Zizi M, La-vigne R, Casteels M, and Huys I: Optimizing the Europeanregulatory framework for sustainable bacteriophage ther-apy in human medicine. Arch Immunol Ther Exp (Warsz).2012;60:161–172.

145. Chanishvili N, Chanishvili T, Tediashvili M, and BarrowPA: Phages and their application against drug-resistantbacteria. J Chem Technol Biotechnol. 2001;76:689–699.

146. Sulakvelidze A, Alavidze Z, and Glenn Morris Jr. J: Bac-teriophage therapy. Antimicrob Agents Chemother. 2001;45:649–659.

147. Weber-Dabrowska B, Mulczyk M, Gorski A, Boratynski J,Kusiak-Szelachowska M, and Syper D: Methods of poly-valent bacteriophage preparation for the treatment of bac-terial infections. Patent US 7, 232, 564 B2. june 19, 2007.

148. Garcıa P, Madeia C, Martınez B, Rodrıguez A, and EvaristoSuarez J: Prevalence of bacteriophages infecting Staphylo-coccus aureus in dairy samples and their potential as bio-control agents. J Dairy Sci. 2009;92:3019–3026.

Received on August 2, 2012in final form, December 9, 2012

Reviewed by:Brian Gilbert

Anthony Hickey

Address correspondence to:Dr. Reinhard Vehring

Department of Mechanical EngineeringUniversity of Alberta

Edmonton, AlbertaT6G 2G8

Canada

E-mail: [email protected]

BACTERIOPHAGES FOR BACTERIAL LUNG INFECTIONS 19