You are on page 1of 10

[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.

80]

Review Article

Filamentous bacteriophage: A prospective


platform for targeting drugs in
phage‑mediated cancer therapy
ABSTRACT Pankaj Garg
A new modality of targeting therapeutic drugs based on the use of bacteriophage (virus), as an emerging tool for specific targeting
Department of
and for vaccine development, has been an area of interest for genetic and cancer research. The approach is based on genetic Chemistry, GLA
manipulation and modification in the chemical structure of a filamentous bacteriophage that facilitates its application not only for University, Mathura,
in vivo imaging but also for therapeutic purpose, as a gene delivery vehicle, as drug carriers, and also as an immunomodulatory Uttar Pradesh, India
agent. Filamentous bacteriophage on account of its high surface holding ability with adaptable genetic engineering properties can
For correspondence:
effectively be used in loading of chemical and genetic drugs specifically on to the targeted lesion location. Moreover, the specific
Dr. Pankaj Garg,
peptides/proteins exhibited on the phage surface can be applied directly as self‑navigating drug delivery nanovehicles. The present Department of
review article has been framed with an objective to summarize the importance of bacteriophage in phage cancer therapy and to Chemistry, GLA
understand the possible future prospective of this approach in developing new tools for biotechnological and genetic research, University, 17‑km
especially in phage ‑mediated cancer therapy. Importantly, the peptides or proteins emerging from the surface of a nano carrier will Stone, NH‑2,
Mathura‑Delhi Road,
make the expense of such peptides economically more effective as compared to other immunological tools, and this seems to be
Mathura ‑ 281 406,
a potential approach for developing a new nanodrug carrier platform. Uttar Pradesh, India.
E‑mail: pankaj.garg@
gla.ac.in
KEY WORDS: Antibody/peptide–drug conjugate, drug delivery vehicle, phage display technology, phage nanoparticle

INTRODUCTION or cytotoxic drugs chemically conjugated on to the


phage coat for therapeutic purposes.
There is a need for intensive research on
bacteriophage and its role in cancer treatment, both Moreover, the peptides or proteins can also be
in diagnosis and therapy as reported by researchers directly self‑assemble themselves into the phage
worldwide.[1-4] A filamentous bacteriophage is imitative nanoparticles that can further be used
considered to be a dependable performer in as a self‑guided nanomissile for drug delivery
antibody engineering and is gaining importance operations [Figure 1a and b]. The present review
in nanobiotechnology and cancer research. The signifies the potential of bacteriophages as an
possible specificity of bacteriophage for certain effective agent for targeted delivery by utilizing
cell and tissue components makes it a crucial them as antibacterial and more importantly
candidate to fight against cancer.[5-7] The specificity antitumor cancer cell nanomedicines.[9,10]
of the target is related to the proteins or peptides
exhibits on the phage coat, which can further be THE STRUCTURE AND BIOLOGICAL MECHANISM OF
conjugated with other therapeutic nanoparticles, FILAMENTOUS PHAGE
endowing them with much specific targeting and
drug‑loading capacity. The genetic and chemical Filamentous bacteriophages are viruses capable of
modification of these phages opens a modular target infecting a variety of Gram‑negative bacteria. The
drug‑carrying platform of nanometric dimensions, Access this article online
where the targeting moieties (biomolecules) and This is an open access journal, and articles are distributed under the terms of the Website: www.cancerjournal.net
Creative Commons Attribution‑NonCommercial‑ShareAlike 4.0 License, which DOI: 10.4103/jcrt.JCRT_218_18
the conjugated drugs can be delivered at will.[8] allows others to remix, tweak, and build upon the work non‑commercially, as
The complete consignment may be assumed as a Quick Response Code:
long as appropriate credit is given and the new creations are licensed under the
wrapped gene linked with bacteriophage genome identical terms.
for gene delivery applications or as imaging agents For reprints contact: reprints@medknow.com

Cite this article as: Garg P. Filamentous bacteriophage: A prospective platform for targeting drugs in phage-mediated cancer
therapy. J Can Res Ther 2019;15:S1-10.

© 2019 Journal of Cancer Research and Therapeutics | Published by Wolters Kluwer - Medknow S1
[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

a
Figure 2: The biological structure of bacteriophage virus with single
strand genome, encapsulated by a protein coat. The positions of phage
coat proteins p‑VIII and minor coat proteins pVII, pIX, pIII, pVI are
marked with their intergenic regions as g8, g7, g9, g3, g6, respectively

BACTERIOPHAGE INTERACTION WITH CANCER CELL! HOW


IDEAL IT MAY BE?

b Current modalities for the treatment of cancer include


Figure 1: (a) Chemically/genetically modified bacteriophage surgery, chemotherapy, radiotherapy, hormonal therapy,
incorporated with nanocarriers such as liposomes and other and other advanced treatment methods including
nanoparticles, utilizing them as self‑driven nanovehicles for immunotherapy (tumor treatment using monoclonal
gene/drug delivery applications. (b) Bacteriophage fused coat antibodies [MoAbs], peptides antisense nucleotides, etc.),
protein/peptide incorporated with liposomes and other polymeric
dendritic cell‑based interventions, and immunogenic cell
nanoparticles self‑assembling them to form a phage mimetic
death inducers are some of the emerging nanobiotechnology
nanocapsules for successful delivery of gene/drug
techniques for cancer treatment. In the present scenario for
a personalized medicinal approach, bacteriophage being a
most characterized class of bacteriophages is the Ff class
nanoparticle can ideally be suited as a unique component for
which includes M13, f1, and fd strains. A filamentous
various molecular therapies suggested and can be explored
phage particle posses a single‑stranded, closed DNA as a promising nanobiotechnological tool for the treatment
genome which includes 11 genes and is encased in a long of cancer.[17‑19]
capsid protein cylinder with a diameter of 7 nm and a
length of 800–2000 nm. The bacteriophage coat contains The therapeutic potential of bacteriophage lies itself
five different proteins [Figure 2]. Approximately 2700 in its structure. The minute and homogeneous size of
reprints of the major coat protein – pVIII [marked as g8 bacteriophage (nanosize) makes it the most promising
in Figure 2] – cover the complete surface of the phage nanoparticle for drug delivery as well as for other purposes
particle, and the remaining four minor coat proteins including phage display and cancer targeting. Moreover,
pVII, pIX, pIII and pVI [marked as g7, g9, g3, and g6 in bacteriophages are the most heavily populated species in the
Figure 2] cover five copies per particle.[11,12] All the proteins biosphere that leads to its licensing for human use due to its
bestowed to the structural stability of a phage particle, nonpathogenic nature and inert properties. Large amounts
but pIII protein is additionally responsible for host cell of their detection naturally in the saliva, serum, and stool
recognition and infection. pIII is the largest and most sample confirm their findings and justify their observations
complicated among all the phage proteins and is composed as biocompatible.[20,21]
of three distinct domains. The N1 terminal domain is
responsible for initiating the translocation of viral DNA Studies by different scientific groups relating to the interaction
into Escherichia coli, the second N2 domain relates with of bacteriophage with cancer cells have been reported in
host cell identification, while the third C‑terminal domain support. Benhar [22] successfully studied the potential of
is responsible for the integration of pIII on to the phage bacteriophage as a targeted drug delivery vehicle by utilizing
coat.[13,14] them as antitumor nanomedicine. The phages were genetically
modified successfully to develop selective target specificity on
Infection in host initiate by the linkage of the phage the phage minor protein coat. A large payload of a drug was then
through the N‑terminal of the pIII ends to the male chemically conjugated to these targeted phages by means of a
E. coli cell. The coat proteins are then disbanded on the cleavable bond.[23,24] The result obtained shows growth inhibition
cytoplasmic membrane, and the viral single‑ stranded with positive toxicity and immunogenicity, thus underscoring
circular DNA is translocated into the cytoplasm, where it the potential of targeted drug‑carrying nanomedicine platform
is replicated by bacterial enzymes and converted into a for further development.[25,26] Related studies by other scientific
double‑stranded replicate form molecule. This molecule, in groups in support reported the in vivo and in vitro demonstration
fact, acts as a template for synthesis of all the phage‑related of bacteriophage binding to the cancer cells. Studies relating to
proteins.[15,16] the binding mechanism of bacteriophage T4 to melanoma cells

S2 Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019


[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

and their possible interaction between the phage capsid protein the production of new therapeutic drugs, as gene delivery
and also the β1 and β3 integrin receptors on the target cancer vehicle, as tumor targeting agents, or for other clinical
cells confirmed the potential of bacteriophage as an ideal drug purpose, demands consideration on phage immunogenicity
delivery platform.[27‑29] and its retention time while developing its efficacious
biological route. Similarly, there are questions arising on the
Bacteriophage interaction in antitumor therapies capabilities of bacteriophage while entering into the blood
The bacteriophage capability of affecting the mammalian circulatory system of higher organisms, behaving like highly
defense immune system has been explored in developing a immunogenic foreign antigenic particles and can interact
link between microbial strategies versus cancer targeting. with the integral immune system, inducing both humoral and
The pioneer study in this regard was conducted by Bloch,[30] cellular responses [Table 1].[42,43]
suggesting that bacteriophage possessed certain anticancer
activities and was reported to have the preferential PHAGE DISPLAY CONCEPT
accumulation in tumor tissues for the suppression of tumor
growth. A confirmatory study in this regard was forwarded by The phage display technology of bacteriophage has been
Kantoch and Mordarski, demonstrating that phages expressing reported to play an important part both in the diagnostic
Fab fragments of antibody, selectively assembled in the tumor and therapeutic applications for treating cancer, by helping
cells, induce both humoral and cellular immune responses, in finding the appropriate receptors on the target cells. The
resulting in the regression of solid tumor in mice.[31] tumor targeting peptides exhibiting on the surface of the
bacteriophage particle is an effective approach for targeting
In similar related studies conducted subsequently by drugs on to cancer cells with promising results.[44,45]
Dabrowska et al.,[32‑34] it was confirmed that T4 bacteriophage
interacting with β3‑integrins receptors at the surface of cancer The therapeutic potential of bacteriophages lies in their
cells, capable of modulating the function of human T‑cell, and structure itself. Its homogeneous and nanosize diameter makes
delayed tumor growth was observed. it the most promising and ideal candidate suited for drug
delivery vehicle as well as for other therapeutic purposes. The
Bacteriophage interaction with reactive oxygen species in development of phage display technology has proven out to
the suppression of cancer cells be a revolutionary step in the world of MoAbproduction and
The reactive oxygen species (ROS) is reported to be the cause for have open the way for new innovations.[46,47] The strength
the initiation and progression of cancer. High level of oxidative of the technology is based on a simple fact that a peptide
stress in the microenvironment of cancer cells results in the emerging on the surface of a filamentous bacteriophage is
promotion of cell proliferation and tumor progression. ROS physically connected to that particular DNA which encodes
suppression may, therefore, be helpful in regulating many it. The concept that a polypeptide fused on the surface of a
undesirable effects relating to the progression of cancer cells. The phage coat protein enabled the development of phage display
studies supporting the interaction between bacteriophage and technology.[48,49]
mammalian cell provide sufficient evidence for the inhibition
of ROS formation by bacteriophage.[35,36] It was reported that Phage display technology is a useful technology that
bacteriophage has the ability to reduce the production of ROS provides a format for obtaining libraries containing a large
by phagocyte in the presence of bacteria.[37,38] The mechanism number (millions or billions) of different peptides and proteins,
of this process is quite complex and involves both the including antibodies.[50,51] Smith working on filamentous
phage–phagocyte interaction and phage–lipopolysaccharide bacteriophage reported that the location, domain structure,
interaction. The role of bacteriophage in the suppression of and flexibility of the pIII bacteriophage minor coat protein
ROS production seems to be important in contributing to the might allow insertion of foreign polypeptides as fusion
beneficial effects of phage‑mediated therapy, especially in proteins to pIII. The display of the protein on the phage surface
patients suffering from a sepsis disease, where a high level of would make it accessible to antibodies, thus allowing the
ROS production plays a crucial role.[39,40] screening of a large library of pIII fusions against a specific
antibody.[52,53] Thus, using a recombinant DNA technology,
Bacteriophage immune response large peptide libraries could thus be built, with every phage
Bacteriophage is responsible for inducing strong antiphage displaying a unique random peptide. A direct physical linkage
humoral responses in the body, recognized as foreign is created between the displayed protein and its encoding gene.
exogenous antigens. Circulation of phages in the blood is The phage display technology has been suggested to use for
inactivated by macrophages of the reticuloendothelial cells, screening of peptide libraries, identifying peptide receptor
thus synergizing with antiphage antibodies.[41] ligands, identifying epitopes for MoAbs, selecting enzyme
substrates, and in screening of cloned antibody repertoires.
Continuous use of a phage as a therapy vehicle causes The most successful use of phage display technology reported
stimulation of memory cells with subsequent generation of has been in the isolation of MoAbs and its fragments using
antibodies. It is reported that utilization of phage either for large phage antibody libraries.[54,55]

Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019 S3


[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

Table 1: Effects of bacteriophage interaction against tumor targeting


Phage interaction Immunomodulatory effect References
Bacteriophage Bacteriophage unique anticancer properties reported to have a [22,23,26‑29]
interaction in antitumor preferential accumulation in the tumor tissues for the suppression
therapies of tumor growth. Phages are capable of expressing proteins and
Fab fragments of antibody which selectively assembled in the tumor
cells inducing certain immune responses. This has been further
confirmed by the bacterial interaction with β‑3 integrin receptors at
the surface of cancer cells capable of delaying the tumor growth
Phage‑phagocyte Bacteriophage ability to suppress the production of ROS by [36,38‑40]
interaction (interaction phagocytes, responsible for the undesirable effects for the
with ROS) progression of cancer cells. The mechanism involves both
phage‑phagocyte interactions and phage‑lipopolysaccharide
interaction and was reported to contribute for the beneficial effect on
the suppression of ROS in patients suffering from sepsis disease
Anti‑phage immunity Bacteriophage being recognized as a foreign exogenous antigen [41‑43]
response is responsible for the generation of antibodies in the body. The
repeated use of bacteriophage as a therapeutic vehicle induces
a strong antiphage humoral response in the body, synergizing
with the antiphage antibodies, resulting in their rapid clearance by
macrophages of the reticuloendothelial cells
Bacteriophage ‑ phage The direct linkage between phage phenotype and its encapsulated [46,47,51,54,57,59,61,62]
display interaction genotype leading to the presentation of molecular libraries on
the phage surface, which is utilized in studying protein‑ligand
interaction, receptor binding sites and in improving or modifying the
affinity of protein for their binding partners
ROS=Reactive oxygen species

initial population of phage particles through several rounds


of selection gives rise to a subpopulation with increased
fitness.[60] In vitro, selection can be performed either on a solid
phase or on an antigen in solution.[61,62] Alternatively, antibody
displaying phage particles can be directly selected against
markers on cell surfaces. Such cell panning selects antibodies
that are more likely to recognize epitopes accessible in vivo
and is important for the selection of therapeutic antibodies.
Figure 3: The recognition of target‑specific peptides through
In vivo panning, where phage repertoires are directly injected
bioscreening or panning process. Screening of combinatorial peptide
into animals, allows the selection of peptides that home to
libraries has been carried out to identify ligands for peptide receptors.
The phage particles are selected against an appropriate antigen, and
target organs [Figure 3].
the specific phages are eluted and amplified repeatedly allowing the
selection of phage clones, identified as an appropriate diagnostic and APPLICATIONS OF BACTERIOPHAGE AS AN IN VITRO AND
therapeutic agent IN VIVO IMAGING AND TARGETING AGENT (USING PHAGE
DISPLAY LIBRARY‑BASED SCREENING METHODS)
Construction of antibody libraries and their selection
The advent of the phage display technology provides an The use of surface display bacteriophage technology
excellent means for the construction and selection of antibody for displaying proteins or polypeptides on the surface,
libraries. Antibody genes can be inserted into a phagemid, a in combination with the in vitro and in vivo selection
plasmid containing a phage‑derived origin of replication.[56] techniques, has paved the way to scientists in generating and
The phagemid is then introduced into competent E. coli cells, manipulating ligands, viz., enzymes, antibodies, and peptides.
which will express the antibody in the form of a fusion The technology is based on the ability to express proteins
protein to the bacteriophage coat, thus providing a direct and peptides on the phage coat of bacteriophage. Libraries
physical linkage between phenotype (antibody specificity) of phage‑displayed polypeptides and proteins in this way are
and genotype (sequence of the variable regions).[57] Selection physically liked to their encoding nucleic acids allowing the
of specific antibodies can be achieved by a panning procedure selection of binding partners to the target site.
where phage particles expressing the antibody fragment are
selected against an appropriate antigen and the phage particles The phage display can further be translated for various
carrying nonspecific antibodies are culled, thereby enriching bacteriophage applications such as in studying protein–ligand
only for those expressing specific antibodies.[58,59] Culling the interaction, in identifying receptor binding sites, and also in

S4 Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019


[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

modifying the affinity of proteins for their binding partners. R a d i o l a b e l i n g o f M 1 3 p h a g e w i t h 9 9 m Tc v i a


The corresponding section deals with the advancement in the mercaptoacetyltriglycine (MAG3) was carried out both with a
therapeutic applications of bacteriophage and phage display target bacterial strain and with a nontarget control bacterial
concept through the work by different scientist groups globally, strain 25922 (E. coli) and strain 29213 of Staphylococcus aureus.
demonstrating the potential of bacteriophage as a futuristic The results obtained from the study revealed that in
drug/gene delivery platform and also as an ideal screening tool. the early hours of dose delivery, the organs related to
reticuloendothelial system (RES), i.e. liver, lungs, and spleen,
Imaging using bacteriophages (phages) show higher percentage of phage accumulation, with about
The phage library‑related screening methods can be 30% accumulation in liver, 8%–10% in lungs, and 2% in spleen
implemented into the field of in vivo screening of various after 30 min. The radioactivity in almost all organs gradually
targets. The designing of peptides and proteins can bind decreases over time, and after 6 h of delivery, only 1% content
various targeting and imaging agents and has opened the of the injected dose was reported to be present in the kidneys,
way to in vivo screening and in monitoring of specific targets spleen, and lungs. It was finally concluded from the study that
using the whole phage body as a targeting as well as an the M13 phage when labeled with Tc‑99m using MAG3 shows
imaging agent.[63] no such sign of in vitro instability both in saline and serum and
was reported that the radiolabeled phage shows equal binding
A bi‑functional peptide ligand system involving the use affinity both with live and heat‑killed bacteria and that once
of bacteriophage has been developed for targeting of bound, the phage is surprisingly stable to dissociation.
receptors.[64] The peptide or protein ligands selectively home
onto tissue‑specific cell receptors can effectively be used as The application of targeted phages as a new tool for magnetic
targeted therapeutic or imaging agents to the selective cells resonance imaging (MRI) imaging has been demonstrated.[72]
both in vitro and in vivo. The authors in their reported study The term “magnetophage” has been coined to describe the
explained that despite extensive advances, such constructs particles they developed. They had isolated peptide displaying
have been limited to use only for in vitro applications.[65] phages that specifically bind phosphatidylserine (PS), a marker
Taking a step forward into an in vivo application, the concept of apoptosis. Magnetophages were prepared by reacting the
has been expanded by constructing and validating a more phages with ultra‑small particles of iron oxide (USPIO), in a
precise bifunctional ligand phage display system with a way that the USPIO reacted with amine groups on the phage
dual binding affinity. Finally, they developed a targeting coat. The magnetophages injected to BALB/c mice were
application in vivo and observed a preferential homing of the mostly sequestered by the liver. To overcome the nonspecific
targeted phages to the tumor site, with four times higher liver uptake, “stealthy magnetophages” were prepared using
accumulation of targeted phage in tumors in comparison to polyethylene glycol (PEG)‑modified USPIO that were linked
control, nontargeted phages. The main achievement of this to the PS‑specific phages. These stealthy magnetophages
study supports the proof of in vivo application of targeted no longer accumulate in the liver and other organs of
phages as imaging agents.[66] RES.[73,74] The fact that phages are taken up by RES cells is well
documented and considered as an obstacle for their potential
A study supporting the use of bacteriophage as a potential as therapeutics. It was shown that the biodistribution of
targeting and imaging agent has been reported by developing a phages in mice can be modulated by conjugating sugar groups
comparative in vivo screening tool of higher value over in vitro to the phage coat.[75] Preparation of stealthy magnetophages
identified clones.[67] Direct labeling of phage clones with far‑red with PEG by Laumonier et al. presented a useful insight to the
fluorochromes was reported, imaging them by a multichannel unfavorable biodistribution of phages.[76]
fluorescence imaging system in mouse tumor models. As a
model targeting moiety, secreted protein acidic rich in cysteine, Bacteriophage (as internalizing phages) for in vivo gene
and as a target, vascular cell adhesion molecule‑1 were used. delivery
It has been demonstrated that how novel targeted peptide The application of phage display technology has turned out
sequences presented on phages can rapidly be selected against to be an exploring approach in identifying and selecting
a particular target and how a fluorescently labeled phage internalizing antibodies from phage library and which in fact
retained target specificity on labeling.[68,69] Finally, it was laid the foundation of their application as a gene delivery
concluded that the fluorescently labeled phage can itself act vehicle. Antibodies that can bind itself on the surface of
as a rechargeable imaging agent. The ease of preparation of the cell receptor are useful molecules for delivering drug or
phage‑imaging agent compared favorably with peptide‑modified DNA into the cytosol of mammalian cell. The resulting work
magneto‑fluorescent nanoparticles and was evaluated using the in this regard was reported by Poul and Marks at UCSE,[77]
same model system.[70] involving the use of an anti‑ErbB2 (anti‑human epidermal
growth factor [EGF] receptor‑2) antibody for determining the
The use of bacteriophage for in vivo imaging of bacterial possibility of a direct selection of an internalizing antibodies
infection has been reported involving the use of radiolabeled and also in identifying the most efficient display format from
technetium (99mTc).[71] the phage libraries. They used ErbB2 antibody C6.5, in a ScFv

Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019 S5


[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

format, displayed monovalently on phage as a model and their therapeutic efficacy using phage‑assisted drug delivery
demonstrated that anti ErbB2‑phage antibodies can undergo approach, by making a chemical modification in their groups
receptor‑mediated endocytosis. The same group went on and including amino (‑NH2), carboxylic (‑COOH), and phenolic
isolated internalizing phages by selecting antibody phage group, which can act as a linker for drug fabrication for its
library on cultured tumor cells.[78] successful delivery.[13]

The possible use of bacteriophage as an internalizing gene The chemical conjugation of biotin (Vitamin H) with phage
delivery vehicle has been explored by researchers at selective nanoparticle to form a biotinylated phage was demonstrated.[81]
genetics.[79] An example for this was set by them where an The extent of labeling was detected by a confocal fluorescence
M13 bacteriophage when linked with targeted EGF receptor microscopy using an avidin‑biotin System. The avidin‑biotin
is capable of delivering a green fluorescent protein gene to system has been advocated for the successful delivery of
EGF receptor cells. large number of radioimmune drugs. On account of having
high affinity between avidin and biotin (highest among the
Similar approach could then also be applied to select biological system), the approach has been successfully explored
genetically modified phages that can be selectively designed by us (Hazra et al.) both for direct and for indirect targeting
and emerge out as gene therapy vectors. Selective genetics of radionuclides to the tumor cells following the prereported
patented the technology of methods using genetic package pretargeting strategies.[82,83]
display for selecting internalizing ligands for gene delivery
(US patent‑US‑6451527B1).Two years later, the same group The approach of using bacteriophage as a drug delivery
reviewed the technology for efficient selection of internalizing platform was successfully evaluated by Benhar for retardation
phages and compared them to viral and synthetic vectors used of tumor cell growth. Antibodies such as anti‑ERGR and
for gene therapy. A similar related study at selective genetics anti‑ErbB2 used as targeting agents were chemically
reported the additional advantage of using a phagemid attached to a drug hygromycin by means of a covalently
system over phage vector and was demonstrated successfully linked amide bond. Successful release of a nanoconjugate
in terms of high vector stability and efficient cell binding drug with significant inhibition in tumor cell growth with
and internalizing capacity. The use of multivalent phagemid over 1000 times more than the corresponding free drug release
system was suggested to work more efficiently for a targeted was reported, revealing an important feature of filamentous
ligand, selectively designed for targeted mammalian cell phage as a drug delivery platform and also recommends the
transduction.[80] application of drug‑carrying phage nanoparticle as an ideal
antibody–drug nanoconjugate.[84]
Gene therapy is a technique in which gene is delivered to the
target cell to restore, over‑express, or inhibit desired gene Polymeric‑encapsulated drugs can be another alternative for
product that can further be used to treat various types of conjugation with phage nanoparticles. Nanoassemblies of
cancers. It is reported that the potential of bacteriophage can a polymer conjugated with phage particle were developed
successfully be harnessed in delivery of gene to eukaryotic with a name FA‑M13‑Poly caprolactone-b-2-vinyl pyridine
cells, through toxic genes transaction to cancer cells. Such coated with folate conjugated M13 bacteriophage (PCL-PLVP)
type of delivery of gene will have a great prospective in the composed of two functional units. The results from the
near future. Similarly, identification of tumor‑homing peptides study showed that polymeric‑loaded drug with phage
can be helpful for targeting cancer cells and antigen detection nanoparticle has significantly higher tumor/nontumor
and in drug and gene delivery.[19] ratio uptake compared to free doxorubicin drug.[85] Similar
therapeutic agents such as photosensitizer and radionuclides
Bacteriophage as a targeted drug delivery agent through fabrication with phage nanoparticles via
The bacteriophages are reported to be used as a successful 1‑ethyl‑3‑(3‑dimethylaminocarbodiimide) chemistry can be
delivery agent for a large number of cyto toxic drugs to used for selective binding of SKBR3 cancer cells.[86] The phage
the target cells. The drugs can be linked to a genetically nanoparticle proved to be useful for targeting cancer cells,
modified bacteriophage by means of a chemical conjugation. antigen detection, and drug delivery through the identification
The phages are modified genetically to exhibit a targeting of tumor and organ homing peptides. Such approach facilitates
species on their phage coat, demonstrating the potential the selective targeting of cancerous cells, through the use of
of using a multiengineered phage as a universal nanodrug bacteriophage, and can further be optimized for selective
carrier. The main achievement of this approach is the targeted treatment and as drug delivery vehicle.[87]
transformation of drug selectivity itself to target selectivity.
Apart from this, a large number of therapeutic drugs including Antibody, peptide engineering has proven its importance in
antibiotics, anticancer drugs (doxorubicin), radionuclides, developing tumor‑targeted therapies and diagnostics. However,
and cytokines, which on account of having higher toxicity the efficacy of this treatment is limited due to nonspecific
and less sufficient quantity earlier have been excluded from uptake of drugs by the RES uptake. Peptides are much smaller
the list of therapeutics, can now successfully be evaluated for molecule reported to be best suited for specific targeting of

S6 Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019


[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

cancer cells. Combinatorial bacteriophage‑displayed peptide earlier has been considered as an obstacle, either to their high
libraries do offer a good source of potential ligands in toxicity effect or uptake by RES cells or immunogenicity, can
developing various cancer‑related molecular targets. Several now be an active participant in the phage‑mediated cancer
peptides are derived from phage‑displayed screening methods therapy. To conclude, we can say that although phages are
and can be translated into effective therapeutic agent. being considered as an ideal for therapeutic applications in
Therefore, the current clinical trials validate the potential phage therapy with their unique structural features, high
of peptide targeting and also the importance of phage drug‑carrying capacity, and diversification in displaying
display technique to identify cancer screening and as a drug targeting moieties, with a potential of becoming an additional
delivery tool, which has been further explored by scientist advantage to the fast‑evolving modalities of nanomedicine;
through their relevant scientific studies reported.[22,88,89] The however, still, a more intensive and thorough research is needed
tabular summarization of all the mentioned applications so that many remaining questions relating to the possibility
of bacteria has been collectively summarized in a separate of altering the pharmacokinetic and pharmacodynamic
self‑explanatory [Table 2]. properties of phages by chemical modification and also about
the safety of using phages in vivo limiting the immune system
CONCLUSION reactivity can evidently be explored out.

The current review article has been framed with an aim of Future prospective
demonstrating the potential of filamentous bacteriophage The direct or indirect application of bacteriophage plays
as a targeted therapeutic agent, especially for cancer cells an important role in suppressing cancerous tumor growth.
delivery agent. The fact that phages can be used to identify Since phages can be used to display polypeptides, which
internalizing antibodies and peptides can successfully be can regulate different peptides, mRNAs, and micro‑RNA that
translated into using the phage as a gene delivery vehicle. are interfering in the cancer cell mechanism, modulating
Similarly, the chemical modification on the phage coat, to load the useful constituent of extracellular matrix is responsible
it with different imaging agents and chemotherapeutic drugs, for regulating the anticancerous activities in the cellular
exploited its role in the development of a new targeted drug mechanism. The another futuristic application of phage can
delivery system. A wealth of new targeting moieties such as be for the detection of biomarkers, which can be used to
imaging agents, radioisotopes, quantum dots, MRI contrast detect bacterial infection in living host and also for detecting
agents, and cytotoxic drugs of therapeutic importance which cancerous tissues and organs in humans. Thus, it is hoped that

Table 2: Tabular summarization of bacteriophage application as imaging, gene, and drug delivery agent
Role of bacteriophage Applications References
Bacteriophage as A precise protein/peptide bifunctional ligand system with dual affinity was developed which [67,68,71,72,74,76]
an imaging or in vivo shows a preferential homing of the target phage to the tumor site with four time’s higher
screening tool accumulation as compared to nontargeted phage
Direct labeling of bacteriophage with far‑red fluorochromes was reported as an in vivo screening
tool. A fluorescently labeled bacteriophage itself acts as a rechargeable imaging agent
Radiolabeling of filamentous bacteriophage with Tc‑99 m as an ideal imaging tool and also
subsequent reduction in the accumulation of radioactive dose to the nontargeted site (RES
uptake) was reported therefore increasing the efficacy of targeting tumor cells
The application of targeted bacteriophage as a new tool for MRI imaging was the developed
using healthy magnetophages. The phage was reported to be react with USPIO with significant
reduction in the accumulation of dose to the nonspecific organs

Bacteriophage as The application of bacteriophage as a gene delivery vehicle involving the use of anti‑ErbBr [77‑80]
in vivo gene delivery antibody has been reported for determining the possibility of direct selection of internalization
agent antibodies that can undergo receptor‑mediated endocytosis
The possible use of genetically modified bacteriophages linking it with target receptor for gene
delivery applications by screening of phage libraries capable of delivering DNA to the receptor cells
The use of multivalent phagemid system which seems to be of high stability with efficient cell
binding properties and internalizing capacity can work efficiently for a target ligand system

Bacteriophage as The potential of using a multiengineered filamentous bacteriophage as a universal nanodrug [19,82‑87]
targeted drug delivery carrier in chemical conjugation of drugs by making a chemical modification in the functional
agent groups that can act as a linker for drug fabrication on the phage coat for its successful delivery
The use of biotinylated phage in an avidin‑biotin system for the successful delivery of large
number of radioimmunotherapeutic drugs both for direct and indirect targeting of radionuclides
to the tumor cells
Polymeric encapsulation of drugs with phage nanoparticles
The result showed that these polymeric phage nanoassemblies are having higher tumor/
nontumor uptake compared to free unconjugated drug
Bacteriophage displayed peptide libraries reported to have a fair source of potential ligands for
developing cancer‑related molecular targets for increasing the efficacy of tumor targeting ratio
RES=Reticuloendothelial system, USPIO=Ultra small particles of iron oxide, MRI=Magnetic resonance imaging

Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019 S7


[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

bacteriophage can be used as an efficient cancer research tool, 18. Diamandis M, White NM, Yousef GM. Personalized medicine:
which can provide us with new futuristic, innovative options Marking a new epoch in cancer patient management. Mol Cancer
Res 2010;8:1175‑87.
for the treatment of cancer.[90]
19. Edelstein ML, Abedi MR, Wixon J. Gene therapy clinical trials
worldwide to 2007 – An update. J Gene Med 2007;9:833‑42.
Acknowledgment 20. Gorski A, Dabrowska K, Switala‑Jeleń K, Nowaczyk M,
Ww would like to acknowledge the support of our esteemed Weber‑Dabrowska B, Boratynski J, et al. New insights into the possible
teacher and guide Prof. D. K. Hazra, Emeritus Scientist and role of bacteriophages in host defense and disease. Med Immunol
Professor, S. N. Medical College, Agra, for his technical help 2003;2:2.
21. Górski A, Weber‑Dabrowska B. The potential role of endogenous
and support in designing and reviewing this manuscript.
bacteriophages in controlling invading pathogens. Cell Mol Life Sci
2005;62:511‑9.
Financial support and sponsorship 22. Benhar I. Biotechnological applications of phage and cell display.
Nil. Biotechnol Adv 2001;19:1‑33.
23. Gross AL, Gillespie JW, Petrenko VA. Promiscuous tumor targeting
Conflicts of interest phage proteins. Protein Eng Des Sel 2016;29:93‑103.
24. Hoogenboom HR, de Bruïne AP, Hufton SE, Hoet RM, Arends JW,
There are no conflicts of interest. Roovers RC, et al. Antibody phage display technology and its
applications. Immunotechnology 1998;4:1‑20.
REFERENCES 25. Petrenko V. Evolution of phage display: From bioactive peptides to
bioselective nanomaterials. Expert Opin Drug Deliv 2008;5:825‑36.
1. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R, et al. 26. Vaks L, Benhar I. In vivo characteristics of targeted drug‑carrying
Nanocarriers as an emerging platform for cancer therapy. Nat filamentous bacteriophage nanomedicines. J Nanobiotechnology
Nanotechnol 2007;2:751‑60. 2011;9:58.
2. Wang AZ, Langer R, Farokhzad OC. Nanoparticle delivery of cancer 27. Dabrowska K, Opolski A, Wietrzyk J, Switala‑Jelen K, Godlewska J,
drugs. Annu Rev Med 2012;63:185‑98. Boratynski J, et al. Anticancer activity of bacteriophage T4 and its
3. Bar H, Yacoby I, Benhar I. Killing cancer cells by targeted drug‑carrying mutant HAP1 in mouse experimental tumour models. Anticancer
phage nanomedicines. BMC Biotechnol 2008;8:37. Res 2004;24:3991‑5.
4. Kaur T, Nafissi N, Wasfi O, Sheldon K, Wettig S, Slaveev R. 28. Dąbrowska K, Miernikiewicz P, Piotrowicz A, Hodyra K, Owczarek B,
Immunocompatibility of bacteriophages as nanomedicines. Lecion D, et al. Immunogenicity studies of proteins forming the T4
J Nanotechnol 2012;2012:1687. phage head surface. J Virol 2014;88:12551‑7.
5. Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer 29. DePorter SM, McNaughton BR. Engineered M13 bacteriophage
nanotechnology: The impact of passive and active targeting in the nanocarriers for intracellular delivery of exogenous proteins to
era of modern cancer biology. Adv Drug Deliv Rev 2014;66:2‑5. human prostate cancer cells. Bioconjug Chem 2014;25:1620‑5.
6. Blanco E, Hsiao A, Mann AP, Landry MG, Meric‑Bernstam F, Ferrari M, 30. Bloch H. Experimental investigation on the relationships between
et al. Nanomedicine in cancer therapy: Innovative trends and bacteriophages and malignant tumors. Arch Virol 1940;1:481‑96.
prospects. Cancer Sci 2011;102:1247‑52. 31. Kantoch M, Mordarski M. Binding of bacterial viruses by tumor cells
7. Bakhshinejad B, Karimi M, Sadeghizadeh M. Bacteriophages and in vitro. Postepy Hig Med Dosw 1958;12:191‑2.
medical oncology: Targeted gene therapy of cancer. Med Oncol 32. Dabrowska K, Opolski A, Wietrzyk J, Switala‑Jelen K, Boratynski J,
2014;31:110. Nasulewicz A, et al. Antitumor activity of bacteriophages in murine
8. Biju V. Chemical modifications and bioconjugate reactions of experimental cancer models caused possibly by inhibition of beta3
nanomaterials for sensing, imaging, drug delivery and therapy. Chem integrin signaling pathway. Acta Virol 2004;48:241‑8.
Soc Rev 2014;43:744‑64. 33. Dabrowska K, Opolski A, Wietrzyk J, Nevozhay D, Szczaurska K,
9. Clark JR, March JB. Bacteriophages and biotechnology: Vaccines, gene Switała‑Jeleń K, et al. Activity of bacteriophages in murine tumor
therapy and antibacterials. Trends Biotechnol 2006;24:212‑8. models depends on the route of phage administration. Oncol Res
10. Citorik RJ, Mimee M, Lu TK. Bacteriophage‑based synthetic biology for 2005;15:183‑7.
the study of infectious diseases. Curr Opin Microbiol 2014;19:59‑69. 34. Dabrowska K, Zembala M, Boratynski J, Switala‑Jelen K, Wietrzyk J,
11. Bárdy P, Pantůček R, Benešík M, Doškař J. Genetically modified Opolski A, et al. Hoc protein regulates the biological effects of T4
bacteriophages in applied microbiology. J Appl Microbiol phage in mammals. Arch Microbiol 2007;187:489‑98.
2016;121:618‑33. 35. Nishikawa M. Reactive oxygen species in tumor metastasis. Cancer
12. Webster R, Barbas FC, editors. Filamentous phage biology. In: Lett 2008;266:53‑9.
Phage Display: A Laboratory Manual. New York: Cold Spring Harbor 36. Nishikawa M, Hashida M, Takakura Y. Catalase delivery for inhibiting
Laboratory Press; 2001. p. 1‑37. ROS‑mediated tissue injury and tumor metastasis. Adv Drug Deliv
13. Li K, Chen Y, Li S, Nguyen HG, Niu Z, You S, et al. Chemical modification Rev 2009;61:319‑26.
of M13 bacteriophage and its application in cancer cell imaging. 37. Eriksson F, Tsagozis P, Lundberg K, Parsa R, Mangsbo SM, Persson MA,
Bioconjug Chem 2010;21:1369‑77. et al. Tumor‑specific bacteriophages induce tumor destruction
14. Longmire MR, Ogawa M, Choyke PL, Kobayashi H. Biologically through activation of tumor‑associated macrophages. J Immunol
optimized nanosized molecules and particles: More than just size. 2009;182:3105‑11.
Bioconjug Chem 2011;22:993‑1000. 38. Przerwa A, Zimecki M, Switała‑Jeleń K, Dabrowska K, Krawczyk E,
15. Merzlyak A, Indrakanti S, Lee SW. Genetically engineered Łuczak M, et al. Effects of bacteriophages on free radical production
nanofiber‑like viruses for tissue regenerating materials. Nano Lett and phagocytic functions. Med Microbiol Immunol 2006;195:143‑50.
2009;9:846‑52. 39. Miedzybrodzki R, Switala‑Jelen K, Fortuna W, Weber‑Dabrowska B,
16. Petrenko VA, Jayanna PK. Phage protein‑targeted cancer Przerwa A, Lusiak‑Szelachowska M, et al. Bacteriophage
nanomedicines. FEBS Lett 2014;588:341‑9. preparation inhibition of reactive oxygen species generation by
17. Clark JR, March JB. Bacterial viruses as human vaccines? Expert Rev endotoxin‑stimulated polymorphonuclear leukocytes. Virus Res
Vaccines 2004;3:463‑76. 2008;131:233‑42.

S8 Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019


[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

40. Victor VM, Rocha M, Esplugues JV, De la Fuente M. Role of free radicals heterogeneity: Implications for targeted medicine. Curr Opin Chem
in sepsis: Antioxidant therapy. Curr Pharm Des 2005;11:3141‑58. Biol 2002;6:399‑404.
41. Diacovich L, Gorvel JP. Bacterial manipulation of innate immunity to 66. Hajitou A, Baramova EN, Bajou K, Noë V, Bruyneel E, Mareel M,
promote infection. Nat Rev Microbiol 2010;8:117‑28. et al. FGF‑3 and FGF‑4 elicit distinct oncogenic properties in mouse
42. Pajtasz‑Piasecka E, Rossowska J, Duś D, Weber‑Dabrowska B, mammary myoepithelial cells. Oncogene 1998;17:2059‑71.
Zabłocka A, Górski A, et al. Bacteriophages support anti‑tumor 67. Kelly KA, Waterman P, Weissleder R. In vivo imaging of molecularly
response initiated by DC‑based vaccine against murine transplantable targeted phage. Neoplasia 2006;8:1011‑8.
colon carcinoma. Immunol Lett 2008;116:24‑32. 68. De S, Chen J, Narizhneva NV, Heston W, Brainard J, Sage EH, et al.
43. Górski A, Wazna E, Dabrowska BW, Dabrowska K, Switała‑Jeleń K, Molecular pathway for cancer metastasis to bone. J Biol Chem
Miedzybrodzki R, et al. Bacteriophage translocation. FEMS Immunol 2003;278:39044‑50.
Med Microbiol 2006;46:313‑9. 69. Koukourakis MI, Giatromanolaki A, Brekken RA, Sivridis E, Gatter KC,
44. Sidhu SS. Engineering M13 for phage display. Biomol Eng Harris AL, et al. Enhanced expression of SPARC/osteonectin in the
2001;18:57‑63. tumor‑associated stroma of non‑small cell lung cancer is correlated
45. Ju Z, Sun W. Drug delivery vectors based on filamentous with markers of hypoxia/acidity and with poor prognosis of patients.
bacteriophages and phage‑mimetic nanoparticles. Drug Deliv Cancer Res 2003;63:5376‑80.
2017;24:1898‑908. 70. Cybulsky MI, Gimbrone MA Jr. Endothelial expression of a
46. Griffiths AD, Duncan AR. Strategies for selection of antibodies by mononuclear leukocyte adhesion molecule during atherogenesis.
phage display. Curr Opin Biotechnol 1998;9:102‑8. Science 1991;251:788‑91.
47. Kehoe JW, Kay BK. Filamentous phage display in the new millennium. 71. Rusckowski M, Gupta S, Liu G, Dou S, Hnatowich DJ. Investigations
Chem Rev 2005;105:4056‑72. of a (99m) Tc‑labeled bacteriophage as a potential infection‑specific
48. Bedi D, Musacchio T, Fagbohun OA, Gillespie JW, Deinnocentes P, imaging agent. J Nucl Med 2004;45:1201‑8.
Bird RC, et al. Delivery of siRNA into breast cancer cells via phage 72. Segers J, Laumonier C, Burtea C, Laurent S, Elst LV, Muller RN, et al.
fusion protein‑targeted liposomes. Nanomedicine 2011;7:315‑23. From phage display to magnetophage, a new tool for magnetic
49. Wang T, D’Souza GG, Bedi D, Fagbohun OA, Potturi LP, resonance molecular imaging. Bioconjug Chem 2007;18:1251‑8.
Papahadjopoulos‑Sternberg B, et al. Enhanced binding and killing 73. Merril CR, Biswas B, Carlton R, Jensen NC, Creed GJ, Zullo S, et al.
of target tumor cells by drug‑loaded liposomes modified with Long‑circulating bacteriophage as antibacterial agents. Proc Natl
tumor‑specific phage fusion coat protein. Nanomedicine (Lond) Acad Sci U S A 1996;93:3188‑92.
2010;5:563‑74. 74. Zou J, Dickerson MT, Owen NK, Landon LA, Deutscher SL.
50. Mori T. Cancer‑specific ligands identified from screening of Biodistribution of filamentous phage peptide libraries in mice. Mol
peptide‑display libraries. Curr Pharm Des 2004;10:2335‑43. Biol Rep 2004;31:121‑9.
51. Krumpe LR, Mori T. The use of phage‑displayed peptide libraries to 75. Molenaar TJ, Michon I, de Haas SA, van Berkel TJ, Kuiper J, Biessen EA,
develop tumor‑targeting drugs. Int J Pept Res Ther 2006;12:79‑91. et al. Uptake and processing of modified bacteriophage M13 in mice:
52. Smith GP. Filamentous fusion phage: Novel expression vectors Implications for phage display. Virology 2002;293:182‑91.
that display cloned antigens on the virion surface. Science 76. Laumonier C, Segers J, Laurent S, Michel A, Coppée F, Belayew A, et al.
1985;228:1315‑7. A new peptidic vector for molecular imaging of apoptosis, identified
53. Jespers LS, Roberts A, Mahler SM, Winter G, Hoogenboom HR. Guiding by phage display technology. J Biomol Screen 2006;11:537‑45.
the selection of human antibodies from phage display repertoires to 77. Poul MA, Marks JD. Targeted gene delivery to mammalian cells by
a single epitope of an antigen. Biotechnology (N Y) 1994;12:899‑903. filamentous bacteriophage. J Mol Biol 1999;288:203‑11.
54. Becerril B, Poul MA, Marks JD. Toward selection of internalizing 78. Poul MA, Becerril B, Nielsen UB, Morisson P, Marks JD. Selection of
antibodies from phage libraries. Biochem Biophys Res Commun tumor‑specific internalizing human antibodies from phage libraries.
1999;255:386‑93. J Mol Biol 2000;301:1149‑61.
55. Giordano RJ, Cardó‑Vila M, Lahdenranta J, Pasqualini R, Arap W. 79. Kassner PD, Burg MA, Baird A, Larocca D. Genetic selection of phage
Biopanning and rapid analysis of selective interactive ligands. Nat engineered for receptor‑mediated gene transfer to mammalian cells.
Med 2001;7:1249‑53. Biochem Biophys Res Commun 1999;264:921‑8.
56. O’Connell D, Becerril B, Roy‑Burman A, Daws M, Marks JD. Phage 80. Larocca D, Baird A. Receptor‑mediated gene transfer by phage‑display
versus phagemid libraries for generation of human monoclonal vectors: Applications in functional genomics and gene therapy. Drug
antibodies. J Mol Biol 2002;321:49‑56. Discov Today 2001;6:793‑801.
57. Nissim A, Hoogenboom HR, Tomlinson IM, Flynn G, Midgley C, Lane D, 81. Nakamura M, Tsumoto K, Ishimura K, Kumagai I. A visualization
et al. Antibody fragments from a ‘single pot’ phage display library method of filamentous phage infection and phage‑derived proteins
as immunochemical reagents. EMBO J 1994;13:692‑8. in Escherichia coli using biotinylated phages. Biochem Biophys Res
58. Rader C, Barbas CF 3rd. Phage display of combinatorial antibody Commun 2001;289:252‑6.
libraries. Curr Opin Biotechnol 1997;8:503‑8. 82. Garg P, Hazra DK. Conjugation of antibodies with radiogold
59. Brian K, Winter J, McCafferty J. Phage Display of Peptides and Proteins: nanoparticles, as an effector targeting agents in radiobioconjugate
A Laboratory Manual. 1996. p. 1-384. Doi: 10.1016/8978-012-402380. cancer therapy: Optimized labeling and biodistribution results. Indian
60. Smith GP, Petrenko VA. Phage display. Chem Rev 1997;97:391‑410. J Nucl Med 2017;32:296‑303.
61. Clackson T, Hoogenboom HR, Griffiths AD, Winter G. Making antibody 83. Hazra DK, Garg P. Pretargeting in radio‑bioconjugate therapy: With
fragments using phage display libraries. Nature 1991;352:624‑8. reference to Rhenium Re‑186, Gold Au‑199 and Lutetium Lu‑177 as
62. Azzazy HM, Highsmith WE Jr. Phage display technology: Clinical candidate therapy isotopes. Indian J Nucl Med 2007;22:1‑8.
applications and recent innovations. Clin Biochem 2002;35:425‑45. 84. Yacoby I, Benhar I. Targeted filamentous bacteriophages as
63. Rami A, Behdani M, Yerdehnavi N, Anbouhi MH, Lomedasht FM. An therapeutic agents. Expert Opin Drug Deliv 2008;5:321‑9.
overview on application of phage display technique in immunological 85. Suthiwangcharoen N, Li T, Li K, Thompson P, You S, Wang Q, et al.
studies. Asian Pac J Trop Biomed 2017;7:599‑602. M13bacteriophage polymer nano assemblies as drug delivery
64. Arap W, Pasqualini R, Ruoslahti E. Cancer treatment by vehicles. Nano Res 2011;4:483‑93.
targeted drug delivery to tumor vasculature in a mouse model. 86. Mao C, Wang F, Cao B. Controlling nanostructures of mesoporous
Science 1998;279:377‑80. silica fibers by supramolecular assembly of genetically modifiable
65. Trepel M, Arap W, Pasqualini R. In vivo phage display and vascular bacteriophages. Angew Chem Int Ed Engl 2012;51:6411‑5.

Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019 S9


[Downloaded free from http://www.cancerjournal.net on Tuesday, March 26, 2019, IP: 143.0.115.80]

Garg: Filamentous bacteriophage: A novel nano-drug carrier platform

87. Kucharewicz‑Krukowska A, Slopek S. Immunogenic effect of 89. Hufton SE, Moerkerk PT, Meulemans EV, de Bruïne A, Arends JW,
bacteriophage in patients subjected to phage therapy. Arch Immunol Hoogenboom HR, et al. Phage display of cDNA repertoires: The pVI
Ther Exp (Warsz) 1987;35:553‑61. display system and its applications for the selection of immunogenic
88. Lunder M, Bratkovic T, Doljak B, Kreft S, Urleb U, Strukelj B, et al. ligands. J Immunol Methods 1999;231:39‑51.
Comparison of bacterial and phage display peptide libraries in search 90. Henry M, Debarbieux L. Tools from viruses: Bacteriophage successes
of target‑binding motif. Appl Biochem Biotechnol 2005;127:125‑31. and beyond. Virology 2012;434:151‑61.

S10 Journal of Cancer Research and Therapeutics - Volume 15 - Supplement Issue 1 - 2019

You might also like