You are on page 1of 18

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/331577874

Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches to


Overcome AMR

Article  in  Current Drug Discovery Technologies · March 2019


DOI: 10.2174/1570163816666190304122219

CITATIONS READS
24 1,175

8 authors, including:

Shun Kai Yang Khatijah Yusoff


Universiti Putra Malaysia Universiti Putra Malaysia
19 PUBLICATIONS   378 CITATIONS    278 PUBLICATIONS   4,092 CITATIONS   

SEE PROFILE SEE PROFILE

Mokrish Ajat Erin Lim


Universiti Putra Malaysia Universiti Putra Malaysia
59 PUBLICATIONS   304 CITATIONS    64 PUBLICATIONS   1,380 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Pregnancy Ketosis in Goats View project

Development of novel vaccines against viral infections View project

All content following this page was uploaded by Lai Koksong on 04 May 2021.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.net
Current Drug Discovery Technologies, 2019, 16, 00-00 1

REVIEW ARTICLE

Mechanisms of Antimicrobial Resistance (AMR) and Alternative


Approaches to Overcome AMR

Chew-Li Mooa, Shun-Kai Yanga, Khatijah Yusoffb, Mokrish Ajatc, Warren Thomasd, Aisha Abushelaibie,
Swee-Hua-Erin Limd, e and Kok-Song Laia,*

a
Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Ma-
laysia, 43400 Serdang, Selangor, Malaysia, bDepartment of Microbiology, Faculty of Biotechnology and Biomolecular
Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia, cDepartment of Veterinary Pre Clinical Sci-
ences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia, dPerdana
University-Royal College of Surgeons in Ireland School of Medicine, Perdana University, MAEPS Building, Serdang,
Selangor, Malaysia, eHealth Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, 41012
Abu Dhabi, United Arab Emirates.

  Abstract: Antimicrobials are useful compounds intended to eradicate or stop the growth of harm-
ful microorganisms. The sustained increase in the rates of antimicrobial resistance (AMR) world-
wide is worrying and poses a major public health threat. The development of new antimicrobial
agents is one of the critical approaches to overcome AMR. However, in the race towards develop-
ing alternative approaches to combat AMR, it appears that the scientific community is falling be-
A R T I C L E H I S T O R Y  
hind when pitched against the evolutionary capacity of multi-drug resistant (MDR) bacteria. Alt-
Received: August 09, 2018
hough the “pioneering strategy” of discovering completely new drugs is a rational approach, the time
Revised: January 30, 2019 and effort taken are considerable, the process of drug development could instead be expedited if ef-
Accepted: January 30, 2019
forts were concentrated on enhancing the efficacy of existing antimicrobials through: combination
DOI: therapies; bacteriophage therapy; antimicrobial adjuvants therapy or the application of
10.2174/1570163816666190304122219  
nanotechnology. This review will briefly detail the causes and mechanisms of AMR as background,
and then provide insights into a novel, future emerging or evolving strategies that are currently being
evaluated and which may be developed in the future to tackle the progression of AMR.

Keywords: Antimicrobial resistance (AMR), current approach, combination therapy, nanotechnology.

1. INTRODUCTION resistance (AMR) rapidly accelerated and spread among dif-


ferent pathogenic species as a result of persistent exposure
Antimicrobials represent a broad spectrum of compounds
and the non-targetted use of antimicrobials in the clinical as
that may act against a wide repertoire of disease-causing well as in agricultural settings. The advancement of AMR
microorganisms such as bacteria, viruses, parasites, fungi
has rendered many current antimicrobials ineffective and it is
and protozoa. These compounds have been used since the
believed that the non-judicious use and overdosing of antibi-
early 20th century to treat infected patients and this has
otics are the primary factors causing the increase in the de-
helped significantly in lowering the morbidity and mortality
tection of drug-resistant bacteria [2].
rates of most infectious diseases. In 1928, Alexander Flem-
ing discovered penicillin and in the 1940s, it came into clini- Over the last few decades, research on AMR has been fo-
cal use just in time for World War II [1]. After only four cused on understanding the resistance mechanisms and also
years of use, however, the first penicillin-resistant strains of on the development of new alternative drugs. Nevertheless,
bacteria emerged; this phenomenon of evolved antimicrobial pharmaceutical companies are investing less than ever in
antibiotic development because of the projected amount of
time and the scale of resources required to demonstrate safe-
ty and to gain approval for the use of novel drugs including
antibiotics [3]. It is a daunting task to identify new potential
drugs, determine their efficacy in appropriate models and
*Address correspondence to this author at the Department of Cell and Mo- then to demonstrate their safety and efficacy in humans. The
lecular Biology, Faculty of Biotechnology and Biomolecular Sciences, scale of investment required and large lead-in time mean that
Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia, only a limited number of novel antimicrobials are at an ad-
Tel: +603-8946 8021; E-mail: laikoksong@upm.edu.my

1570-1638/19 $58.00+.00 © 2019 Bentham Science Publishers


2 Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 Moo et al.

vanced stage of development at the current time [2]. Howev- cies operating without a license may seem more accessible as
er, the development of AMR is a very pressing clinical prob- the waiting time is shorter, and also provide services at a lower
lem that is expected to significantly worsen in the coming cost without consultation fees [8]. Some pharmacies may be
years. In order to mitigate this effectively, clinical diagnostic willing to provide a spectrum of treatment options to accom-
processes and treatment practices that are both rapid and yet modate the financial resources of the patient, and which may
highly specific are required This should minimize unneces- not be the most appropriate for treating the presenting illness.
sary prescriptions and lead to a general improvement in the
quality of prescribing practice. Greater control needs to be 2.2. Extensive Non-Human Use of Antimicrobials
exercised in the use of existing effective antibiotics and pa-
tients need to be educated in the need to adhere to specific In agriculture, antibiotics are regularly administered to
prescription regimes. livestock as growth supplements. It has been estimated that
in the United States alone, 80% of antibiotics sold are used
Since the development of novel antimicrobials is going to in animal husbandry, mainly to boost growth and to prophy-
take a considerable amount of time, a stop-gap measure is lactically prevent infection [9]. It is believed that antimicro-
required. Research resources should be invested in strategies bial treated livestock has better overall health, produces larg-
that focus on lowering the resistance of microorganisms to- er yields and higher quality products. Nevertheless, contact
wards existing, approved antimicrobial agents, or at least, de- with treated animals or consumption of food from animals
celerating the emergence and sharing of AMR- related genes treated with antimicrobials promotes the transfer of resistant
between pathogenic organisms. Here we briefly detail the bacteria to humans. There is a particular risk when food is
causes and mechanisms of AMR, and aim to provide insights improperly prepared. This issue was first noted in 1980
into the strategies presently being applied to mitigate the where high rates of antibiotic resistance were found in or-
spread of AMR. ganisms from the microbiome of farmers and this was linked
to the detection or resistance in the intestinal flora of their
2. CAUSES OF ANTIMICROBIAL RESISTANCE farm animals [10]. Molecular detection methods have re-
2.1. Inappropriate Use and Prescribing vealed that consumers come into contact with resistant bacte-
ria from the farms through meat consumption [9]. It is be-
The inappropriate use of antibiotics is one of the lieved that such a transfer may lead to significant human
principal factors that will continue to contribute to the emer- health implications. Antibiotics used in agriculture can also
gence of AMR in bacterial pathogens. Several studies have affect the environmental microbial ecosystem since as much
shown instances where either antibiotics have been inappro- as 90% of antibiotics given to livestock are excreted in the
priately administered, an incorrect agent was chosen or an stool and urine. These excretions enter the environment, then
inappropriate course duration was defined in 30% to 50% of spread through groundwater, fertilizer and surface runoff
all prescriptions [4]. It was also found that approximately into rivers exposing environmental microorganisms to low
30% to 60% of the antibiotics specifically prescribed for doses of clinically important antibiotics.
patients in hospital intensive care units are unnecessary, in-
appropriate or are not at the optimal in dosage form [4]. In- 2.3. Lack of Surveillance on The Susceptibility of Antimi-
deed antibiotic prescriptions are sometimes dispensed by the crobials
primary healthcare providers just to placate a persistent pa-
tient who has a viral infection, with no therapeutic benefit Epidemiological information allows the distribution of
whatsoever for the patient. Over time, the unnecessary use of AMR to be monitored among the members of a population
antibiotics will increase exposure and the frequency with and geographically. The collection of community-based an-
which AMR emerges in exposed bacterial populations. Anti- timicrobial susceptibility data will help health care centers in
biotic concentrations that are sub-inhibitory and sub- specific communities to correctly treat infections in a timely
therapeutic will encourage bacteria to become resistant to the manner. Susceptibility testing of antimicrobials has to be done
antibiotic by driving the natural selection of bacteria that have routinely for surveillance and to better understand the local
only a slightly better resistance to the antibiotic. In time, muta- epidemiology of resistance, these data are vital for effective
tions result in altered resistance gene sequences or changes in clinical practice in order to develop appropriate responses to
gene expression levels that allow resistance to higher, therapeu- AMR. However, due to the lack of human resources, cost and
tic doses of antibiotics to develop. Horizontal gene transfer available infrastructure, locality-based susceptibility testing of
(HGT) allows the exchange of resistance gene cassettes be- antimicrobials is rarely performed in remote rural settings due
tween strains, species and even different genera of bacteria [5]. to resourcing issues. Since resistance rates in a particular re-
Bacteria with enhanced gene expression or gain of gene func- gion of a country may change dramatically over time, ideally
tion mutations that are driven by antibiotic exposure can in- the collection of surveillance data should be performed regu-
crease the virulence of pathogens, whereas HGT and increased larly in order to have an accurate epidemiological model [7].
mutagenesis may cause antibiotic resistance genes to spread
through a mixed bacterial population, for example in the gut [3]. 3. ANTIBIOTICS CLASSIFICATION AND MODE
OF ACTIONS
A lack of strict legislation and its ineffective implementa-
tion on the sale of antimicrobials also lead to the misuse of Antibiotics can be classified according to their mecha-
antimicrobials by the general public [6, 7]. In some countries, nism(s) of action and the effects that they exert on target
antimicrobials can be obtained over-the-counter without prop- microorganisms, causing either bacteriostatic or bacteri-
er medical prescription or advice, and even from an cidal-type activity. Nowadays, there are many different
unqualified vendor. For some members of the public pharma- types of antibiotics available, but the most widely accept-
Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 3

ed classification based on chemical structure identifies the reducing N-acetylglucosamine of the new saccharide-
following groups: beta-lactams; tetracyclines; macrolides; peptide. Peptidoglycan is linked to the cell wall and the
aminoglycosides; peptides antibiotics; lincosamides and new peptidoglycan attaches to the existing peptidoglycan
streptogramins. Antibiotics can be further grouped ac- of the cell wall. The attachment is catalyzed by a trans-
cording to their mode of action, which may be inhibition peptidase reaction, involving the peptide chains of both
of bacterial cell wall synthesis; inhibition of ribosomal molecules [21]. One of the polymers must contain a D-
function; inhibition of nucleic synthesis or inhibition of alanyl-D-alanine terminus. The peptide bond connecting
other metabolic processes. two D-alanyl residues in the pentapeptide is then cleaved
by transpeptidase. This is followed by acylation through
3.1. Inhibitors of Cell Wall Synthesis the carbonyl group of the second to the last D-alanine
residue. Antibiotics that are responsible for inhibiting the
The biosynthesis of the bacterial cell wall is separated cell wall phase synthesis are the β-lactams like penicillin,
into several stages. The first stage is the cytosolic phase, which contain a four-membered ring that undergoes
which is followed by the membrane synthesis phase and, acylation reaction with the transpeptidases [22].
subsequently, by the cell wall synthesis phase.
3.1.1. The Cytosolic Phase of Synthesis 3.2. Inhibitors of Cell Membrane Function
The cytosolic phase of cell wall generation coincides The small antimicrobial peptides (AMPs), are de-
with the synthesis of the nucleotide precursors UDP-N- scribed as cell membrane acting antibiotics and the bacte-
acetylmuramyl (UDP-MurNAc)-pentapeptide and UDP- rial membrane is usually their target. The action of AMPs
N-acetylglucosamine (UDP-NAG [11]. Fosfomycin is a is initiated by the binding of the peptide to the cell mem-
broad-spectrum antibiotic that acts on common Gram- brane. The binding causes perturbation of the structure of
positive (Staphylococcus aureus (S. aureus), Enterococ- the membrane and hence disrupts the membrane function-
cus), and Gram-negative (Klebsiella pneumoniae and ality. Disruption of membrane structure and function can
Pseudomonas aeruginosa) bacteria by inhibition of the disturb events that are associated with the membrane,
enzyme, pyruvyl transferase, which is also known as UDP- such as the biosynthesis of the cell wall. AMPs can also
N-acetylglucosamine enolpyruvyl transferase, or MurA be transported across the membrane, enabling the peptides
[12, 13]. This enzyme catalyzes the first step in e cell wall to interact with targets in the cytoplasm [23]. The interac-
biosynthesis, by transferring an enolpyruvyl group from tion between AMPs and the intracellular target structure
PEP to UDP-NAG [14]. Fosfomycin acting as an analogue causes the inhibition of the microorganism's normal biol-
of PEP is able to form a covalent bond due to the presence ogy. The precise mechanisms by which AMPs cause cel-
of the cysteine amino acid at the active site of the MurA lular dysfunction and mortality are now being studied by
enzyme [15]. This is similar to the molecular interaction many researchers. One of the ways in which the AMPs
between UDP-NAG and PEP. However, the formation of a work is through the disruption of the biochemical struc-
covalent bond occurs when the epoxide ring of fosfomycin ture and composition of the membrane [24]. AMPs can
reacts with the thiol group of the cysteine resulting in per- also form pores in some membranes causing the leakage
manent inactivation of the enzyme [16]. of metabolites, membrane depolarization and ultimately
cell death [24]. In addition, disturbance in the normal cell
3.1.2. The Membrane Phase of Synthesis
membrane structure affects peptidoglycan precursor syn-
At this stage, the processes of cell wall synthesis are thesis and translocation. Examples of natural antimicrobi-
localized to the cytoplasmic membrane and are catalyzed al peptides include defensins and protegrins, which exhib-
by membrane-bound enzymes [17]. The previously it antibiotic properties against Gram-positive and Gram-
formed non-nucleotide portion of the precursor molecules negative bacteria such as Streptococcus spp., S. aureus, K.
is transferred to a carrier in the cytoplasmic membrane. A pneumoniae and Escherichia coli [25]
UDP-MurNAc-pentapeptide precursor is attached to un-
decaprenyl pyrophosphate, a transport lipid. Lipid I 3.3. Inhibitors of Ribosome Function
[MurNAc-(pentapeptide)-pyrophosphoryl-undecaprenol]
is then formed. Subsequently, the formation of lipid II The ribosomes of bacteria consist of the 50S and 30S
[GlcNAc-β-(1,4)-MurNAc-(pentapeptide)- subunits [26]. Antibiotics are able to target one or both
pyrophosphoryl-undecaprenol] occurs by adding glu- subunits simultaneously. Aminoglycoside antibiotics are
cosamine from UDP-NAG [18]. The peptide cross-bridge complex sugars with glycosidic linkages that allow them
is added at the third amino acid residue where peptidogly- to bind to particular ribosomal subunits [27]. Aminogly-
can is indirectly cross-linked. The flipping of lipid II to cosides are differentiated by the structure of their molecu-
the external surface of the cell membrane occurs and pen- lar nucleus, with either of the aminohexoses , 2-
icillin-binding proteins (PBPs) incorporate it into the nas- deoxystreptamine or streptamine linked to the nucleus.
cent peptidoglycan layer [19]. One of the critical antibiot- NH4 and OH groups are essential for the interaction of
ics that inhibit the cell wall synthesis during this phase is aminoglycosides with particular ribosomal proteins [28].
bacitracin, which acts most effectively on S. aureus [20]. The first aminoglycoside studied was streptomycin. Strep-
tomycin causes the misreading of DNA genetic code by
3.1.3. The Cell Wall Phase of Synthesis binding to a specific S12 protein in the 30S ribosomal
At the cell wall phase, polymerization of the wall sub- subunit (Fig. 1). The tetracycline family antibiotics also
units occurs by transferring new peptidoglycan to the non- work by binding to the 30S ribosomal subunit (Fig. 1).
4 Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 Moo et al.

Fig. (1) Inhibition of sites of protein biosynthesis sites by various antibiotics that bind to the 30S and 50S ribosome.

This binding obstructs the interaction of the aminoacyl- pyrimidine and purine bases or with the incorporation of
tRNA with site A of the bacterial ribosome [29]. The the nucleotides into polymers. Some agents that act as
bacteriostatic action of tetracyclines inhibits a wide range analogues are able to incorporate themselves into polynu-
of bacteria including Mycoplasma sp. and Chlamydia sp. cleotides to interfere with accurate nucleic acid transcrip-
[30]. Drugs such as chloramphenicol and the macrolides tion or replication. For example, flucytosine (5-
inhibit the 50S ribosomal subunit. The formation of pep- fluorocytosine) is an antifungal agent. Flucytosine is con-
tide bonds is inhibited by chloramphenicol through at- verted into 5-fluorouracil in the target cell. Fluorouracil
tachment to a peptidyltransferase enzyme. This enzyme is will then be integrated into RNA resulting in premature
located on the 50S ribosomal subunit of both Gram- chain termination. Premature chain termination inhibits
positive and Gram-negative bacteria [31]. The structure of the action of thymidylate synthetase, halting the synthesis
macrolides comprises a large lactone ring that binds to 50S of thymine nucleotides [32]. This, in turn, result in im-
subunits and impairs the activity or the translocation (or paired DNA synthesis. Arabinoside analogues are phos-
both) of peptidyltransferase. Erythromycin is considered phorylated by kinases in the same manner as deoxynucle-
to be the most important macrolide and inhibits some osides and inhibit DNA synthesis through DNA polymer-
Gram-negative species belonging to the genera Chlamyd- ase antagonism. Arabinosides can be considered as
ia, Haemophilus, Legionella and Mycoplasma as well as deoxyucleoside analogues [33], and adenosine arabino-
Gram-positive bacteria [13]. side can be integrated into DNA in competition with, the
integration of dATP. Clinically the nucleoside analog,
3.4. Inhibitors of Nucleic Acid Synthesis acyclovir, inhibits DNA polymerase and also the thymi-
dine kinase of herpes viruses once the drug is converted
Antimicrobial compounds interfering with the synthe- into a triphosphate [34].
sis of nucleic acids can act at several different stages of
the process, either by inhibiting: nucleotide synthesis; 3.4.2. Inhibition of DNA-Directed DNA Polymerase
DNA-directed DNA polymerase activity or the replication Rifamycins are in a group of antibiotics that inhibit the
of the DNA. DNA-directed RNA polymerase [35]. Promoter sites that
3.4.1. Interference With Nucleotide Synthesis initiate DNA transcription will be bound by the RNA pol-
ymerase. The RNA polymerase attaches to a factor that
Most antimicrobial compounds, that interfere with nu- confers specificity for recognition of specific promoter
cleic acid synthesis, either interfere with the synthesis of sites. Although rifampin binds to a subunit of RNA non-
Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 5

covalently, the binding is still strong enough to interfere 4. MECHANISMS OF ANTIBIOTIC RESISTANCE
with the transcription initiation process (Fig. 1), and there
Concurrent with the emergence of new resistance
will be no effect of the antibiotic on RNA synthesis if it is
mechanisms and novel resistance-associated genes, new
administered after a cycle of polymerization has com-
transmission vectors are also being discovered. Therefore,
menced [13]. Rifamycins bind to RNA polymerase tightly
and, hence, only small amounts of these antibiotics are we will now focus on current discoveries relating to the
mechanisms by which AMR is conferred to bacteria via
needed in order to inhibit RNA synthesis. In addition, the
intrinsic, naturally acquired or externally acquired mecha-
binding does not affect DNA polymerase activity, as it is
nisms. The mechanisms discussed will include how the
specific for the RNA polymerase.
pathogen prevents access to drug targets, changes in the
3.4.3. Inhibition of DNA Replication structure of antibiotic targets, as well as the modification
DNA gyrase and topoisomerase I act together to sus- or inactivation of antibiotics that can be affected.
tain the DNA double helix in the supercoiled state [36]. Resistance in bacteria to some antibiotics can either be
During chromosomal replication in bacteria, DNA gyrase, intrinsic or acquired through gene mutations or via HGT.
a tetrameric protein comprising two A and two B subunits Bacterial species with intrinsic resistance have the capa-
is essential in relieving the molecular torsion. The catalyt- bility to survive the actions of certain antibiotics due to
ic action of gyrase gives rise to a temporary covalent bond innate structural or functional features [40]. Intrinsic re-
between the DNA and the A subunit in what is termed as sistance is achieved via inherent structural or functional
“the double strand passage reaction”. Quinolones can an- characteristics of the molecular target. The antibiotic dap-
tagonize DNA replication. During strand passage, quin- tomycin is active against Gram-positive bacteria but inef-
olones bind to the cleavage intermediate; and this causes a fective against Gram-negative bacteria; this is the result of
deleterious effect on the normal process of DNA replica- innate differences in the cytoplasmic membrane composi-
tion. Quinolones bind to the cut ends of the DNA in the tion between the Gram-positives and Gram-negatives. The
enzyme-DNA complex to prevent the rotation and religa- Gram-negative cell membrane is comprised of less
tion of the cut ends of DNA [36]. Quinolones can either be anionic phospholipids than the cytoplasmic membrane of
bacteriostatic or bactericidal depending on the concentra- Gram-positive bacteria [40]. This reduction in the fraction
tion. At low concentrations, the antibiotic remains bound to of anionic phospholipids affects the Ca2+ -dependent in-
the cut ends of DNA. Replication and transcription are in- sertion efficiency of daptomycin into the cytoplasmic
hibited as DNA remains supercoiled which stops the repli- membrane, which is needed for its antibacterial activity.
cation fork or the transcription complex from further pro-
Recent studies have determined that several genes are
gression (Fig. 1). The attachment of quinolone to the gyrase
responsible for the innate resistance that exists towards
enzyme complexed with the cut ends of the DNA is re-
certain classes of antibiotics, such as fluoroquinolones, β-
versible, and so explains how quinolones have bacteriostat-
lactams and aminoglycosides. The key resistance genes are
ic properties. At high concentrations of a quinolone, the
thioredoxin reductase (trxB), thioredoxin A (Trx A), SapC,
DNA ends will be freed from the complex, chromosomal DacA, FabI and D-Ala-D-Ala carboxypeptidase. So, com-
DNA is cleaved and this eventually leads to cell death [37].
bined antagonism of these gene products could help to
boost the activity of existing drugs such as rifampin, ami-
3.5. Inhibitors of Other Metabolic Processes noglycosides as well as some β-lactams in the treatment of
One mechanism by which drugs can inhibit the critical infection [41]. The genes responsible for innate resistance
metabolism of bacterial cells is by blocking the biosyn- were identified from high-density genome mutant libraries
thesis of tetrahydrofolate. Tetrahydrofolate is a co-factor and high-throughput screening studies. To create the librar-
in the process of de novo synthesis of thymidine mono- ies, either mutagenesis by random transposon insertion or
phosphate, which is needed for the biosynthesis of bacte- targeted insertion was carried out in bacteria such as Staph-
rial DNA and RNA [38]. When tetrahydrofolate is defi- ylococcus aureus, Escherichia coli and Pseudomonas ae-
cient, there is no synthesis of thymidine and hence the ruginosa [41, 42]. This library screening approach is par-
bacteria is unable to produce new DNA or RNA. Tetrahy- ticularly effective in discovering possible novel drug com-
drofolate is synthesised from dihydropteroate diphosphate binations that will enable inhibition of intrinsic resistance
and p-aminobenzoic acid (PABA). Bacteria produce mechanisms [40]. Consequently the spectrum of activity of
dihydropteroic acid from these two precursor compounds, other antibiotics can be extended to other pathogens be-
a reaction catalyzed by dihydropteroate synthetase. Sul- yond their common target species.
fonamides block the synthesis of dihydropteroate by com- In addition to intrinsic resistance, there are several
peting with PABA for binding to the enzyme as a struc- other resistant mechanisms that can be acquired by bacte-
tural mimic [39]. Consequently, sulfonamides act as com- ria, all of which fall into three primary groups: prevention of
petitive inhibitors of dihydropteroate synthetase to block access to target; mutational changes in antibiotic targets and
the synthesis of dihydropteroate. This leads to tetrahydro- finally, modification of targets [40]. These modes of action
folate deficiency. Deficiency in tetrahydrofolate results in have been reviewed extensively over the past decades. Thus,
the inability to synthesize thymidine, which will eventual- this review will provide an update on the most recent studies
ly cause cell death. for each type of antibiotic resistance mechanism.
6 Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 Moo et al.

Fig. (2). Mechanisms of antibiotic resistance. (a) β-lactamase breaks β-lactam antibiotics via hydrolysis and hence deactivating the
antibiotics. (b) Prevention of access to target due to increased efflux. The efflux pump of bacteria transport antibiotics actively out of
the cell. (c) Downregulation in the expression of porin proteins or the substitution of porins with more-selective channels. (d) Changes
in the structure of the target such as mutation makes the binding of antibiotics less efficient, without affecting the functionality of the
target itself, conferring antimicrobial resistance. (e) Post-translational modification of targets.

family (MATE), the major facilitator superfamily (MFS),


4.1. Prevention of Access to Target
the small multidrug resistance family (SMR) and the re-
Denial of access of an antibiotic to its target can be sistance-nodulation-cell-division family (RND) [45].
exemplified by the resistance of Enterobacteriaceae to Multidrug-resistant (MDR) efflux pumps transport a vast
carbapenems, which is due to reduced permeability of the range of structurally different substrates. There are several
bacterial membrane (Fig. 2). Hydrophilic antibiotics dif- genes encoding the MDR efflux pumps located on bacte-
fuse into the bacterial cell via the porin proteins in the rial chromosomes, however, some of those genes can be
outer membrane. OmpC and OmpF of E. coli are typical transferred between bacteria, due to mobilization on the
examples of the major porins found in most Enterobacte- genes onto plasmids [40]. Recently, it has been found out
riaceae. Several studies have suggested that resistance that the IncH1 plasmid, isolated from Citrobacter freun-
strategies employed by bacteria include the downregula- dii, has a gene cassette encoding a novel resistance modu-
tion in expression of porin proteins, or the substitution of lation division (RND) pump together with the gene for
major porins with more-selective membrane channels New Delhi metallo-β-lactamase 1 (NDM1) [46]. This de-
(Fig. 2). So the resistance to carbapenems in the Entero- velopment is a major concern since this indicates that the-
bacteriaceae in the clinical setting persists even without se specific resistances can be transmitted between bacteria
carbapenemase production by the bacteria; instead critical on plasmids, which may facilitate the spread of novel re-
mutations reduce porin production or result in the expres- sistances to other bacterial pathogens that are clinically
sion of mutant porin alleles [43, 44]. relevant.
The prevention of access to the antibiotic's target can The most well-characterized RND family pump that
be achieved through increased efflux of the antibiotic. The has clinical relevance is found in Gram-negative bacteria.
efflux pump of bacteria transports antibiotics actively out A broad range of substrates can be exported at an acceler-
of the cell in bulk. This contributes significantly to the ated rate when the RND is overexpressed [47]. RND
intrinsic resistance in Gram-negative bacteria to a variety pumps are located in the inner membrane and a tripartite
of drugs that are used in the treatment of bacterial infec- complex is formed with an outer-membrane channel TolC
tions. The five major types of efflux pumps that have been or OprM and a periplasmic adaptor protein AcrA or
identified include the ATP-binding cassette family MexA [40]. One of the identified RND pumps is AcrB,
(ABC), the multidrug and toxic compound extrusion which is a homotrimeric protein. Computational studies
Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 7

and co-crystallization of E. coli AcrB in complex with other example recently identified is the chloramphenicol-
transported substrates have identified two binding pockets florfenicol resistance gene (cfr) product which methylates
in AcrB. The binding pockets are able to accommodate the A2503 residue in the 23S rRNA. This causes the tar-
substrates of different sizes and chemical properties, get to become resistant to an extensive range of antibiotics
which establishes how the pumps confer resistance to a that interacting with 23S rRNA near this methylation site.
wide range of different antibiotics [48, 49]. Enterobacteri- These antibiotics include lincosamides, oxazolidinones,
aceae, S. aureus and P. aeruginosa overexpress the RND pleuromutilins, phenicols and streptogramins [56]. Both
efflux pump. Overexpression of the efflux pumps is regu- of the genes, erm and cfr are often found on plasmids
lated by local regulators, such as EmrR from E. coli, which drive the dissemination of AMR among bacteria
QacR from S. aureus and also by global regulators. Tran- [57]. Another example of such a modification strategy is
scription factors of the AraC-XyIS family are important the methylation of the ribosomal proteins which prevents
global expression regulators. MtrA enhances transcription the binding of aminoglycosides, so inhibiting protein syn-
of mtrCDE in N. gonorrhoeae [50]. The regulator family, thesis. The armA and rmt genes, which encode different
Arac-XyIS is encoded together with a repressor of the methyltransferases, have been discovered in clinical iso-
several antibiotic resistance protein MarR family [51]. lates of Enterobacteriaceae [58]. In addition, the re-
sistance genes for quinolone, the qnr gene families, have
4.2. Changes in Antibiotic Targets by Mutation also been found on plasmids. Pentapeptide repeat proteins
Antibiotics have evolved or are designed specifically (PRPs) are encoded by the qnr genes. PRPs bind to topoi-
so that they are able to bind to their specific targets with somerase IV and DNA gyrase to protect them from quino-
high affinity and consequently disrupt the normal activity lone action. A recent discovery suggested that after drug
of the target. Alterations in the structure of the target can binding, the interaction of PRPs and the topoisomerase-
make the binding of antibiotics less efficient, without af- quinolone complex liberates the quinolone [59]. The lib-
fecting the functionality of the target itself, conferring eration of quinolone enables the topoisomerase to main-
antimicrobial resistance (Fig. 2). A single point mutation tain its activity and re-ligate the DNA, preventing the
in the gene encoding an antibiotic target can result in re- breakage of the double-stranded DNA [59].
sistance towards the given antibiotic [40]. One of the clas-
sic examples is the development of resistance towards 4.4. Direct Modification of Antibiotics
rifampin. This occurs due to single point mutations, caus- In addition to preventing antibiotics from entering the
ing an amino acid substitution in the rpoB gene. Conse- cell, bacteria can also modify the structure of antibiotics,
quently, these mutations decreased the affinity of rifampin rendering them inactive. One of the mechanisms by which
for its target, allowing the transcription to continue [45]. bacteria inactivate antibiotics is through a hydrolysis reac-
Some organisms have multiple copies of genes that tion (Fig. 2). To date, there are a collection of enzymes,
encode the targets of some antibiotics. For example, the such as carbapenemases, chloramphenicol acetyltrans-
23S rRNA ribosomal unit gene of Gram-positive bacteria ferases, which can degrade or alter different classes of
is encoded by multiple identical copies of the gene. Re- antibiotics, such as macrolides, β-lactams, aminoglyco-
sistance to the antibiotic linezolid is conferred to S. sides and phenicols. Over the years, the classes of antibi-
pneumoiniae and S. aureus through multiple copies of the otics have been expanded in order to overcome the prob-
23S rRNA gene. The gene copies have a high recombina- lems posed by hydrolytic enzymes. Both the early β-
tion rate that produces bacterial populations with resistant lactamases and the extended-spectrum β-lactamases
mutant alleles. Eventually, the whole population becomes (ESBLs) were active against β-lactams, while ESBLs also
resistant towards linezolid [52, 53]. Another example of a have activity against oxyimino-cephalosporins. Imipen-
mutation in a target that confers resistance is possession emase (IMP), New Delhi Metallo-β-lactamase and Vero-
of a gene that is homologous to the original target. This na integron encoded Metallo β-lactamase (VIM) are car-
can be seen in methicillin-resistant S. aureus (MRSA) bapenemases and also ESBL carriers which give rise to
whereby the resistance is conferred by having the genetic the emergence of β-lactam antibiotic-resistant isolates.
element known as staphylococcal cassette chromosome mec These have serious consequences for the treatment of se-
(SCCmec). This element bears the mecA gene, which is re- vere bacterial infections [60]. Due to the increase in
sponsible for the β-lactam insensitive protein penicillin- ESBL gene carrying bacteria, carbapenem antibiotic use
binding protein PBP2a, the mutated penicillin-binding pro- is increasing in clinic. Subsequently, this will cause an in-
tein in this case, supports cell wall biosynthesis, despite the crease in the number of β-lactamase expressing clinical
fact that the native PBP is inhibited by antibiotics [54]. isolates that carry carbapenemases with carbapenem
hydrolyzing activity [61, 62]. Another mechanism by
4.3. Modification (and Protection) of Targets which bacteria inactivate antibiotics is by addion of a
chemical group. Addition of a chemical group to the anti-
Post-translational modification of targets is another biotic molecules prevents its binding to the target mole-
way of achieving antibiotic resistance without a mutation- cule due to steric hindrance. Chemical groups that are
al event occurring in the genes that encode target mole- transferable onto antibiotics include acyl, nucleotidyl,
cules (Fig. 2). For instance, methylation of 16S rRNA and phosphate and ribitoyl groups. There is a broad range of
the resulting alteration in the drug-binding sites is enzymes responsible for these transferase reactions result-
achieved by the erythromycin ribosome methylase (erm) ing in a substantial and diverse group of antibiotic-
family. This activity prevents the binding of lincosamides, resistance gene families [63].
macrolides and streptogramin to the 16S rRNA [55]. An-
8 Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 Moo et al.

The aminoglycosidic antibiotics are large molecules (pyrazinamide and rifampicin [66]. The principle behind
carrying exposed amide and hydroxyl groups that are this therapy anticipates that the bacterium will modify the
prone to modification. The enzymes that modify amino- individual antibiotic targets in order to become resistant. If all
glycosides confer a high level of resistance to the antibi- strategies of resistance are targetted simultaneously this ena-
otics [40]. Acetyltransferases, nucleotidyltransferases and bles the drug combination to remain effective, thus, keeping
phosphotransferases modify the aminoglycoside family, the likelihood of bacterial propagation at the lowest level.
that include the commonly prescribed antibiotics gen-
Combination therapy has also proved effective for the
tamicin, streptomycin and amikacin [64]. Recently, there
treatment of Plasmodium falciparum, malaria pathogen.
has been a worrying development, whereby a novel ge-
The efficacy of chloroquine has reduced due to the spread
nomic island in Campylobacter coli has been discovered;
of the resistant P. Falciparum strains. Hence, artemisinin
this genomic island was originally isolated from broiler is used to treat malaria by inhibiting SERCA-family Ca2+-
chickens in China. The genomic island encoded all three
ATPase PfATP6. Artesunate or artemether, derived from
classes of the six aminoglycoside-modifying enzymes. This
artemisinin is usually administered in combination with
cassette confers resistance to all the aminoglycoside antibi-
lumefantrine, amodiaquine or mefloquine in order to ex-
otics, which are currently in use to treat Campylobacter
tend the efficacy of artemisinin.
infections. Of particular concern is resistance to gentami-
cin, which is considered an antibiotic of last resort in severe A study reported that tunicamycin interacts synergisti-
bacterial infections with highly resistant strains [65]. cally with β-lactam antibiotics. Tunicamycin is an inhibi-
tor of an enzyme involved in the biosynthesis pathway
5. STRATEGIES TO COMBAT AMR for cell wall teichoic acid, the N-acetylglucosamine-1-
phosphate transferase TarO, in S. aureus. Synergistic in-
AMR is a global threat that is escalating and is a cause teraction between tunicamycin and β-lactam antibiotics
of increased morbidity and mortality in the community greatly decreased the minimum inhibitory concentration
from previously controllable infections. The clinical set- (MIC) of oxacillin against one MRSA clinical isolate
ting is increasingly associated with the development of from 50 µg/mL to 0.4 µg/mL at a tunicamycin concentra-
AMR due to the high level of antibiotic use. The spread of tion of only 0.08 µg/mL [67]. This is an example of the
antibiotic resistance to other environmental reservoirs fur- inhibition of different targets within the same pathway.
ther complicates effectual strategies in combating AMR. Furthermore, β-lactam antibiotics also showed significant
The process of developing new antimicrobials needs to be synergy with early cell wall synthesis inhibitors, such as
expedited as the speed of AMR development has exceeded β-chloro-D-alanine, bacitracin, D-cycloserine, fosfomy-
the developmental rate of antimicrobials. Unfortunately, cin, vancomycin and teicoplanin at sub-inhibitory concen-
identification of new and effective pharmaceuticals is not a trations. The synergistic interaction is able to lower the
straightforward task. Thus, researchers have embraced alter- resistance of S. aureus strain towards methicillin by two-
native strategies in order to combat AMR. In the subsequent fold for β-chloro-D-alanine, and up to 128-fold for
sections, of this review, we will discuss current and future fosfomycin, as measured by the reduction in MIC [68]. A
approaches to combating AMR. combination of drugs acting on different targets in the
same pathway has less breadth of efficacy in comparison
5.1. Combination Drug Therapy to a combination of drugs acting on different targets in
Combination drug therapy is defined as the use of two different pathways. For instance, inhibiting two enzymes
or more medications or therapies to address the same con- that are involved in the biosynthesis pathway of folate will
dition. Combination drug therapy is used rather than indi- be effectual as the target cell requires folate to synthesize
vidual therapy (monotherapy) on microorganisms because dTMP for DNA synthesis. However, if a subpopulation of
microorganisms are more prone to resistance development target cells can avoid the need of folate, then this combina-
in monotherapy. Although drug-drug interactions may be tion of drugs would become less effective. An example of a
a stumbling block in this strategy and needs to be taken combination of drugs acting on different targets in unrelat-
into consideration during the development of drugs, evi- ed pathways, is the combination of amoxicillin, the β-
dence points to this therapeutic approach greatly lessening lactam antibiotic with clavulanic acid [69]. Amoxicillin
the probability of primary resistance to the components of inhibits cell-wall synthesis of bacteria but is often de-
the combination course (Kaur, 2016). The combination stroyed by β-lactamase, on the other hand, clavulanate acid
drug regimens are divided into 3 mechanistic strategies: is an inhibitor of β-lactamase. So both of the compounds
1) Combination of drugs acting on different targets in dif- work synergistically, wherein clavulanate prevents β-
ferent pathways. 2) Combination drugs acting on different lactamase from degrading amoxicillin at bacterial cell wall,
targets in the same pathway. 3) Combination drugs acting so maintaining the level of amoxicillin, thereby enhancing
on a single target, but in different dimensions [66]. its antibacterial activity. Streptogramins are another group
of agents used in combination therapy. They act on a single
One classical example of the combination of drugs act- target via different mechanisms as they are made up of two
ing on different targets in different pathways, is the treat- active molecules: a non-ribosomal peptide and a polypep-
ment used for Mycobacterium tuberculosis infections. The tide non-ribosomal peptide hybrid. Streptogramins attach at
four combined drugs are: an inhibitor of arabinosyltrans- two adjacent sites on the 50S subunit, near to the peptidyl
ferases involved in cell wall biosynthesis (ethambutol); an transferase center [70]. The potency is 10 to 100 fold high-
inhibitor of the enoyl reductase subunit of the fatty acid er when combined together as compared to either one mol-
synthase (isoniazid) and twoRNA polymerase inhibitors ecule acting as a single agent.
Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 9

Many debates have arisen regarding the prospective harming commensal bacterial flora is an advantage over
benefits of combination therapy versus monotherapy. One conventional antibiotics. Nevertheless, bacteria can de-
of the advantages of combination therapy is that it em- velop resistance to phages in a brief period of time inter-
ploys the synergistic interaction between two drugs result- rupting the antibacterial effect [79]. In order to reduce the
ing in enhanced bacterial killing. These synergies are of- development of resistance to phages, mixtures of phages
ten initially identified through in vitro studies. This can be are often used. However, obtaining a mixture of phages
seen for example in the case of the combination of colistin which are sufficiently effective against pathogens and
and the glycopeptide vancomycin or teicoplanin. In vitro their variants, is a difficult task [80]. One problem that
studies showed synergistic interactions between these can arise is that when a mixture of phages is used in an
antibiotics against multidrug-resistant isolates of Acineto- effort to increase the host range the effectiveness of spe-
bacter baumannii that may have clinical relevance [71]. cific phages in the mixture against certain strains of bacte-
Combination therapy may also give a greater overall spec- ria may decrease. To circumvent this, a cocktail of phages
trum of activity of specific antibiotics against different should be made specifically for each patient so that only
pathogens. Despite the benefits of this combination ap- phages that are effective against the bacterial strains re-
proach, some adverse effects, such as aminoglycoside sponsible for the infection will be used.
nephrotoxicity, have been documented. In a study involv-
Antibiotics are non-specific, promoting the destruction
ing 876 patients, ceftazidime was compared with gen- of pathogens and commensal bacteria with equivalent
tamicin and piperacillin, the renal toxicity was notably
potency, this is a significant problem, as loss of the nor-
higher in the combination therapy group (P < 0.001),
mal microflora can result in a novel pathogenic state. This
whereby 5 of the patients required hemodialysis and one
can be observed with the use of fluoroquinolones, which
died due to renal insufficiency [72]. Roughly 85% of
can lead on to superinfections with Clostrodium difficile,
aminoglycosides are found in the renal cortex [73], and
following the loss of the normal gut bacteria. Unlike anti-
when cytosolic concentrations become high, apoptosis is biotics, bacteriophages have high specificity in killing
triggered [74]. In addition, several meta-analyses of com-
bacteria without harming or altering gut microbiota. High-
bination therapies have shown that renal toxicity occurs
ly specific bacteriophages, however, only infect a single
more often in patients who receive aminoglycosides ther-
species of bacteria. Bacteriophages attach to highly spe-
apy than those who do not [75].
cific receptors available on the host cell surface, in order
to enter a bacterial cell. These receptors are absent from
5.2. Bacteriophage Therapy non-target species so avoiding disruption of the normal
The rise in the number of infections caused by drug- microflora [78]. Moreover, bacteriophages are only need-
resistant bacteria within the clinical setting has propelled ed in small doses for the treatment of bacterial infections
the research community to explore alternatives to conven- since they are able to replicate in vivo. Bacteriophages are
tional antibiotics. Bacteriophages are abundant [76], and consequently less immunogenic because a reduced dose
most likely every bacterium has their own specific viruses of a foreign substance is being administered into the body
that could be used as antibacterial agents [77]. Phages [67]. So a single bacteriophage has the capability to kill a
were first used experimentally as therapeutic agents in the single bacterial strain, unlike antimicrobials, which work
early 20th century. The main challenges regarding phage via the concept of sub-lethal dosing that can affect the
therapy are the strict regulatory guidelines that needed to whole gut microbiome.
be complied with, as well as developing effective thera- A study has demonstrated that the overall effect of the
peutic practices [78]. In spite of this, phage therapy could antibiotic and phage could be increased synergistically.
potentially be used as an alternative to conventional anti- Through manipulating the structures of the bacterial cell
biotics as long as the special requirements of phage-based by genetic engineering, the effect of the drug was en-
medicines are met. hanced increasing the susceptibility of E. coli towards
A bacteriophage is a virus that lyses and disrupts the antibiotic therapy. Phage-encoded genes that repress the
metabolism of bacteria following cellular invasion. Phage SOS response of E. coli resulted in reduced selection
therapy suppresses pathogenic bacterial infections during pressure. This eventually led to a decrease in antibiotic
the lytic cycle [40]. The lytic cycle begins with phage resistance. Following the treatment, there was no sign of
attachment to a host cell. The tail fibers of the virion then bacterial recovery after a period of either 12 or 24 hours
penetrate the bacterial cell wall and inject the viral ge- and killing efficacy increased tremendously by several
nome into the cytoplasm. Immediately after penetration, orders of magnitude in comparison with conventional
phage DNA is transcribed and begins early protein syn- therapy. Nonetheless, strains of bacteria were not com-
thesis. Some of the synthesized proteins break down the pletely eliminated by this treatment, so some bacteria
host (bacterial) DNA whereas some are enzymes required must have arisen that were resistant to the phage and anti-
for the replication of the phage DNA. The newly synthe- biotic [81]. In the year 2016, in vivo studies investigating
sized phage DNA produces late proteins, which are the how Enterococcus faecalis acquired resistance to bacteri-
subunits of phage capsid. Capsid proteins assemble to ophages were reported. In the experiments, it was found
form a head with condensed viral DNA packed inside. out that the pivotal role of a phage infection protein from
The tail then joins to head forming progeny virions. Late Enterococcus faecalis (PIPEF) was a factor in phage tro-
proteins lyse the host cell, which then releases viral prog- pism [82]. The in vivo studies on a gnotobiotic mouse
eny into the surrounding environment [70]. The fact that model showed that E. faecalis acquired phage resistance
phages infect bacterial hosts very selectively, without through mutations in PIPEF. Phage therapy may also have
10 Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 Moo et al.

a prophylactic application. Administering phages orally tion as well as maintaining the efficacy of the antibiotic
can eliminate pathogens such as Salmonella spp., E. coli against β-lactamase expressing Gram-positive bacteria.
and Clostridium difficile which can cause diarrhea. Bacte- One remarkable example is the addition of the adjuvant
riophages could be designed to regulate the gastrointestinal cilastatin to inhibit dehydropeptidase, the enzyme that
microbiota composition, ultimately benefitting the host in degrades the β-lactam antibiotic imipenem. Cilastatin pro-
different ways given the growing interest in understanding tects imipenem from degradation and prolongs its antimi-
how the microbiome affects overall metabolism [83]. crobial effects when given in combination [88]. Adjuvants
also enhance the uptake of antimicrobial agents by the
Bacteriophages are active against both Gram-positive,
targets organisms. Colistin (Polymyxin B) is a poly-
as well as Gram-negative bacteria [84]. Bacteriophages
cationic molecule containing both hydrophilic and lipo-
function by lysis, which is totally different from the
mechanism of how antibiotics work. As mentioned be- philic ends. Its use is limited due to the nephrotoxicity of
the drug. Despite this limitation, in low doses, colistin can
fore, bacteriophages are specific. This specificity enables
be used as an adjuvant because it acts as a membrane de-
phage therapy to narrow the antibacterial spectrum, limit-
tergent and so enhances the penetration of simultaneously
ing its effect to either single species or even single strain
administered hydrophilic antibiotics, such as carbapenems,
within a species, without damaging other non-targeted
glycopeptides, rifampicin and tetracyclines [66].
bacteria. Moreover, in terms of the economic aspects of
phage therapy, this approach appears to be promising. Some adjuvants also nullify the effect of the efflux
Although the duration of phage therapy is longer than pump in exporting antibiotics from the bacterial cells. For
conventional antibiotic treatment, the cost is lower. This example, the primary mechanism of tetracycline resistance
was shown in phage treatment study on 6 patients infected is through its expulsion from bacterial cells by efflux. An
with various Staphylococcus bacterial strains including adjuvant that is structurally similar to tetracycline and
methicillin-resistant S. aureus [85]. competes with tetracycline for binding to the efflux pumps
will reduce the efflux of active tetracycline [89].
Despite the fact that phage therapy has several ad-
vantages, studies still need to be done to determine the This could suppress the development of resistance by
capability of phages to treat infections in vivo. The admin- maintaining the effective intracellular concentration of the
istration route, the optimal dose, the duration and fre- antibiotic. Examples of derivatives that act as tetracycline
quency of treatment all have to be determined for specific efflux inhibitors include the 6-(alkythio)methyl-
pathogens and phages. The specificity of phage therapy doxycycline analogues. These derivatives are more potent
being specific to a pathogen is an advantage, but it is also inhibitors of Gram-negative bacterial growth compared to
a liability. A clinical sample has to be isolated, cultured Gram-positive bacteria. Therefore, the derivatives pro-
and the pathogen identified before a bacteriophage specif- duce synergistic effects with tetracyclines in Gram-
ic to that particular sample can be defined and given to the negative but have an additive effect in Gram-positive bac-
patient for administration. Innovations in rapid diagnostic teria where they support the activity of tetracycline by
methods may help. However, all of these are time- competing for access to the efflux pump. Another exam-
consuming, and most clinical microbiology laboratories ple of a compound which has the ability to inhibit efflux
and health care centers have limited resources. Besides pump is reserpine. Reserpine is a known mammalian
that, the possibility of phage transferring DNA from a MDR pump inhibitor. It has the ability to suppress the
bacterium to another is also a concern of phage therapy. growth of S. aureus and Streptococcus pneumoniae
The chances of developing a new microbe or resistant strains that have become resistant to ciprofloxacin [89].
bacteria are high if the genetic material is transferred [86]. Through a screening program for inhibitors of the efflux
Fortunately, the frequency of resistance in vivo during pump, Phe-Arg-β-naphthylamine (PAβN), MC-207,110 ,
phage therapy, thus far, is reportedly low, as compared to showed an ability to inhibit the 4 clinically relevant
in vitro resistance [87]. pumps in P. Aeruginosa: MexCD-OprJ; MexAB-OprM;
MexEF-OprN and MexXYOprM, and also similar pumps
5.3. Antimicrobial Adjuvants in other multidrug-resistant Gram-negative bacteria.
Antimicrobial adjuvants are compounds that generally PAβN lowered the resistance of wild-type strains of
do not possess any intrinsic antimicrobial activity. The P.aeruginosa towards levofloxacin by eight-fold, while in
primary aim of using antimicrobial adjuvants is to en- efflux pump overexpressing strains the resistance was
hance the efficacy of existing antibiotics, as well as to decreased by up to 6-fold [90].
suppress the emergence of resistant strains. Adjuvants Adjuvants may also disrupt biofilm stability. Bacteria
usually act by reversing the existing resistance mecha- form biofilms to protect themselves from antimicrobial
nisms of microbes. The functions of adjuvants are: 1) In- agents and to overcome mechanical stress. Bacillus sub-
hibition of antimicrobial resistance elements, 2) Enhance- tilis produces a mixture of D-amino acids, preventing bio-
ment the uptake of antimicrobial in target cells, 3) Nullifi- film formation as well as disassembling the existing bio-
cation of the effect of the efflux pumps, 4) Disruption of film of other organisms. The mixture of D-amino acids
biofilms and 5) Promotion of bacterial oxidative stress. consists of D-leucine, D-tyrosine, D-methionine and D-
Antimicrobial adjuvants may act to inhibit antimicro- tryptophan. Treatment of biofilms with D-amino acids
bial resistance elements. For example, adjuvants may act causes the detachment of amyloid fibers that link bacterial
as β-lactamase inhibitors and so can be added to a β- cells together in the biofilm. It is now known that D-
lactam antibiotic to protect theβ-lactam ring from distor- amino acids are released by many bacteria, and can be co-
Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 11

administrated therapeutically with broad range antibiotics efflux pump activity and decreased uptake in bacteria
to disperse biofilms and regain antibiotic sensitivity. such as P. aeruginosa and E. coli [94].
Some adjuvants promote oxidative stress. Tellurium oxy- Nanoparticle-based targeted drug delivery is divided
anion tellurite (tellurite) causes high levels of oxidative into passive or active targeting. Passive targeting enhanc-
stress in bacterial cells. When tellurite is used at sub- es the permeability and retention at the infection site,
lethal concentrations, together with antibiotics, it inter- whereas active targeting is directed by factors, such as
feres with cell wall and protein synthesis, enhancing the receptor interactions or and local activation temperatures.
bactericidal action of the antibiotics by many folds. In addi- Active targeting is achieved through modifying the sur-
tion to direct bactericidal activity, the generation of reactive face of nanoparticles which allow them to recognize spe-
oxygen species results in damage to metabolic enzymes, cific ligands on the cells at the infection site. Vancomycin
lipid peroxidation and glutathione depletion [66]. has high efficacy in inhibiting Gram-positive bacteria.
However, vancomycin has high toxicity towards the ear
5.4. Nanotechnology for Targeted Drug Delivery to and kidney. Vancomycin mesoporous silica nanoparticles
Combat Antibiotic Resistance make the specific killing of pathogenic Gram-positive
Nanotechnology is the design and application of nano- bacteria selectively over macrophage-like cells [95]. De-
sized materials (1-100 nm). Nano-sized structures are spite the fact that nanoparticles can be beneficial as a drug
small in size, have a high surface-to-volume ratio and are delivery system, they may, however, promote drug re-
amenable to surface modification. They may be exploited sistance. This was identified in one study, whereby plas-
as a means of delivering therapeutic or diagnostic com- mids were transferred at a higher frequency between bac-
pounds across physiological barriers, gaining access to terial cells with nanoparticle treatment. It was assumed
biological molecules and intended targets. Nanoparticles that aluminum nanoparticles promoted the conjugative
may be used to combat microbial resistance and multi- transfer of plasmids and that this led to the spread of mul-
drug-resistant mutants. Nanoparticle-based materials have tidrug resistance between bacteria [96]. The three factors
been investigated in combating microbial resistance re- that promoted this effect are probably: Cell membrane
cently, since they may facilitate the delivery and action of damage due to oxidative stress caused by aluminum na-
conventional antibiotics at the site of the membrane [91]. noparticles, temperature and pH of water which affects
It has been proposed that nanoparticles may have inherent the transfer and lastly, the selectively promoted expres-
bactericidal action based on their physiochemical proper- sion of specific genes that is crucial for replication and
ties and also that the nanoparticles are able to prevent bio- transfer of plasmids. The high surface-to-volume ratio
film formation [92]. Biofilm structure is one of the factors increases the possibility of nano-materials interacting with
that promote bacterial resistance by protecting the bacteria pathogens and membranes [97]. Therefore, nano-
from high antibiotic concentrations while, at the same technology has great potential uses for medical applica-
time, exposing the bacterial population to sufficient anti- tions such as specific drug targeting, cell labeling and
biotic to drive mutation of potential resistance genes, gene therapy. Some examples are discussed below.
among different bacterial cells. Studies have identified
gold-based, silver-based and magnesium-based nanoparti- 5.5. Chitosan
cles that prevent biofilm formation [93]. This activity is
achieved by modifying the shape and in size of the parti- Chitosan is a linear polysaccharide, derived from chi-
cles in order to maximize the surface-area-to-mass-ratio. tin. It interacts well with negatively charged microbial cell
walls and cytoplasmic membranes due to its polycationic
Nanoparticles may also be used as an antibiotic carrier properties. This membrane interaction causes decreased
system. There are several types of nanoparticles that are osmotic stability, subsequently leading to disruption of
currently used for drug delivery, such as liposomal and the cellular membrane and eventually leakage of intracel-
dendrimer nanoparticles and also carbon nanomaterials, lular contents. Nano-sized chitosan has a high surface area
just to name a few. The advantages of nanoparticles for to volume ratio. It is able to enter the bacterial cells and
drug delivery, in comparison with the conventional the nuclei of fungal cells to inhibit mRNA and protein
approaches, are in terms of size, protection of the cargo synthesis when microbial DNA is bound by it. In addition,
and precision in the site of delivery [93]. In particular, the high surface area to volume ratio of chitosan
nanoparticles are suitable to be used in the treatment of nanoparticles results in higher surface charge density, en-
intracellular bacterial infections. Antibiotics have poor hancing the affinity of chitosan for bacterial and fungal
trans-membrane transport properties due to their large cells and so conferring greater antimicrobial activity [97].
molecular size, whereas nanoparticles can be loaded with Wound infections are caused acutely by microorganisms
a drug and used to transfer the drug across a membrane. and, most chronic infection is accompanied by infections
The drug enters host cells through endocytosis and is re- with by multiple AMR bacteria. Nanoparticles have a
leased intracellularly to achieve a high intracellular con- broad-spectrum antimicrobial activity that is able to inhib-
centration [93]. In addition, drugs incorporated into a na- it the growth of different pathogenic bacteria. Nanosilver,
noparticle carrier are protected from chemical reactions polyvinyl alcohol and chitosan combinations have been
that may be detrimental to their activity, hence, the drugs’ studied for antiseptic activity, and an impregnated fiber
potency can be maintained for a longer period of time. mat with a combination of these agents has been evaluat-
Researchers have shown that nanoparticles can overcome ed in wound healing studies [98].
the resistance mechanisms of bacteria such as increased
12 Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 Moo et al.

5.6. Carbon Nano-tubes of the body. It was concluded that liposome-entrapped


phages had longer bio-retentivity rates in the lungs and in
Carbon nanotubes (CNTs) are new nano-vectors used
the kidney, making this approach an appropriate treatment
for the delivery of a variety of therapeutic molecules.
for respiratory tract and urinary tract infections caused by
CNTs can be loaded with macromolecules, such as pro-
K. pneumoniae [102].
teins, oligosaccharides, proteins and antibiotics. Organi-
cally functional end groups and side walls of f-CNTs can
be covalently modified. Further modification can render f- 5.9. Metal Ions
CNTs more soluble in a range of solvents, including wa- The properties of nanoparticles can be altered by
ter, and the interaction between water-soluble carbon changing their size. This was shown in E. coli and S. au-
nanotubes and mammalian cells leads to cytoplasmic reus which are highly susceptible to CuO and ZnO nano-
translocation of the nanotubes into mammalian cells [97]. particles of a specific size range. Since the size of bacteria
is usually in the micrometer range, the nanoparticles have
5.7. Dendrimers to have the ability to enter the bacterial cells. The activity
of nanoparticles also depends on their stability and con-
Dendrimers are highly branched polymers with low
centration in the growth medium [103]
polydispersity and defined nano-architecture. Their size
can be precisely controlled, and drug conjugation capabil- Synthetic CuO, ZnO and iron (III) oxide (Fe2O3) na-
ities are considerable due to the multiple branching points noparticles were tested for bactericidal activity against E.
in the structure. Dendrimers have a high surface function- coli, S. aureus, B. subtilis,and P.aeruginosa in a well-
ality as their synthetic process takes place in a layer-by- diffusion based assay. CuO and ZnO were more potent
layer manner, around a central unit allowing for sequen- than Fe2O3 nanoparticles, with ZnO having the greatest
tial modifications [99]. The highly branched characteris- bactericidal activity. In addition, the studies showed that
tics of dendrimers provide a large surface area to the ratio with a larger surface-area-to-volume ratio, the antibacteri-
for microorganisms to interact in vivo. The functional al activity of the nanoparticles increased [104]. J.S. Möh-
groups achieve a high surface density and allow the den- ler et al., found that silver exhibited a synergistic interac-
drimers to be synthesized with high binding affinity for a tion with beta-lactam antibiotics. The structure and inter-
wide variety of viral and bacterial receptors [100]. Hydro- action between Zn(II), Ag(I), or Cu(II), and β-lactam an-
philic or hydrophobic drugs can be inserted into the empty tibiotics were examined using isothermal titration calo-
hollow in the central unit and on the multivalent surfaces rimetry (ITR) and nuclear magnetic resonance (NMR).
of the dendrimers. Increased efflux and decreased uptake ITC and NMR revealed that the antibiotic activity was
of antibiotics in bacterial cells are the most common increased in the presence of metal ions which could be
mechanism for AMR to develop. Studies showed that na- due to the inhibition of β-lactamase activity. Moreover,
noparticles are able to overcome the resistance mecha- kinetic assays conducted demonstrated that IMP-1enzyme
nisms and, hence, suppress drug resistance. For instance, activity was inhibited in the presence of an Ag(I)-
nanoparticle dendrimers inhibit the P-glycoprotein medi- cefuroxime complex. These findings indicated that the
ated efflux in the gastrointestinal tract microflora [101] synergistic interaction between Ag(I) and β-lactam antibi-
otics inhibited the β-lactamase activity, enabling the anti-
5.8. Liposomes biotics to act on bacteria with greater efficacy [105]. Ex-
posing bacterial cells to silver causes puncturing of the
Liposomes are spherical vesicles made up of one or cell wall and consequently leakage of the cytoplasmic
more phospholipid bilayers surrounding a water-filled contents. The bacteria become more susceptible to the
cavity. In order to change the physiochemical properties influx of silver ions, disrupting the production of cellular
of liposomes, the proportions and composition of different protein and interfering with cellular metabolism. Eventu-
lipids in the liposome formulation can be adjusted to mod- ally, cell growth is affected because silver ions cause ge-
ify the size of the liposome; surface charge; pH sensitivi- netic disturbance and enzymatic inhibition [104].
ty; temperature sensitivity and liposomal membrane fluid-
ity [97]. The liposome vesicles can act as carriers with Gold nanoparticles also exhibit antibacterial activity
hydrophilic or hydrophobic properties. Drug distribution by interfering with protein synthesis in prokaryotes. Col-
is highly influenced by the size of the liposomal vesicle, loidal gold reduced the affinity of the ribosome for tRNA
and encapsulation of antibiotics in precisely designed li- andgold nanoparticles also affected bacterial metabolism
pid vesicles can achieve the optimal pharmacokinetic and by lowering the ATP synthase activity required for ATP
pharmacodynamic properties for delivery and activity production. In the bacterial respiratory chain, gold nano-
[101]. By encapsulating antibiotics in lipid vesicles, bio- particles suppressed nicotinamide adenine dinucleotide
distribution and pharmacokinetics can be improved within (NADH) dehydrogenase activity by binding irreversibly
the host, toxicity is decreased, and activity against intra- onto the thiol groups on NADH dehydrogenase. This
cellular and extracellular pathogens is enhanced, particu- affects the reduction-oxidation balance in the cell which
larly in helping to overcome antibiotic resistance. The results in the generation of oxidative stress [106]
biodistribution of Klebsiella pneumoniae phage particles
incorporated into liposomes in the different organs of 5.10. Molecular Docking
BALB/c mice was assessed. From this study, it was Molecular docking is a computational tool that can be
shown that liposome-entrapped phages were more stable used to discover drugs. This method assesses the affinity of
than free phages in the bloodstream and also in the organs binding between two molecules, such as small molecules
Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 13

Table 1. Examples of studies done using a few alternatives for combat of AMR.

Types of Therapy Strategy Outcome Reference

Tunicamycin synergized with oxacillin, lowering the mini-


Tunicamycin and oxacillin [66]
mal inhibitory concentration against S. aureus

Combinatory Therapy Plasmodium falciparum is resistant to chloroquine alone.


Artesunate/ artemether (artemisinin
However, the combination of an artemisinin derivative and
derivative) and amodiaquine/ meflo- [109]
amodiaquine/ mefloquine successfully inhibited the SER-
quine/ lumefantrine
CA-family Ca2+-ATPase PfATP6

The combination of cinnamon bark oil and meropenem had


successfully reduced the effective dosage of cinnamon bark
- Cinnamon bark oil and meropenem [110]
essential oil from 0.16% to 0.08% and meropenem from 32
µg/ml to 16 µg/ml when applied on K. pneumoniae.

Minimum inhibitory concentration (MIC) of peppermint


essential oil (PEO) was 8% v/v and meropenem was 4
µg/mL when used singly on KPC-3 producing E. coli
- Peppermint essential oil and meropenem pMG309. In combinatorial treatment, PEO and meropenem [111]
reacted synergistically against KPC-3 producing E. coli
pMG309, significantly lowered the MIC of PEO to 1% v/v;
and meropenem to 0.5 µg/mL.

Combination of staphylococcal phage and antibiotics cured


Staphylococcal phage and antibiotics [112]
78 % of patients with sepsis
Bacteriophage Therapy Phage application decreased the bacterial titre in all patients'
Phage preparations against secondary
sputum specimens and the general health of patients im- [112]
infections in cystic fibrosis patients
proved.

Celecoxib is a non-steroidal drug used to treat arthritis by


inhibiting cyclooxygenase-2 (COX-2). Suppression of drug
resistance has been shown in cancers because of the inhibi-
tion of the MDR1 efflux pump. It has also been determined
Celecoxib [113]
to increase the sensitivity of S. aureus to several antibiotics,
such as ciproflaxin, chloramphenicol, ampicillin ciproflaxin
Antimicrobial Adjuvants and kannamycin by causing the bacteria to accumulate
drugs inside their cells.

Chitosan enhanced the antimicrobial efficacy of silver-


loaded membranes up to 70% which resulted in larger zones
Chitosan of inhibition on E. coli and S. Aureus cultures. Antimicrobi- [114]
al activity was enhanced by 70 % by increasing the chitosan
concentrations,

Liposomes were used as antibiotic carrier systems to coun-


ter extracellular pathogens. Studies found that with proper
lipid formulations, drug distribution, and vesicle-bacterium
Liposome [115]
interactions, antimicrobial activity against bacteria such as
Nano-particles E. coli, P. aeruginosa, Acinetobacter sp., K. pneumoniae,
`and S. aureus can be enhanced.

The uptake of silver ions and the resulting cascade of intra-


Silver cellular reactions in teicoplanin resistant S. Pneumoniae [116]
overcame resistance.

Colloidal gold caused damage to DNA and the bacterial cell


Gold [117]
wall in cefotaxime resistant E. coli and K. pneumoniae

Predicted the binding mechanism of S-1360, one of the


Molecular Docking Docking studies first beta-diketoacid integrase inhibitors to enter clinical [107]
studies.
14 Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 Moo et al.

and protein targets to identify potential inhibitory com- period of time, disturbance of the human microbiome due to
pounds based on the binding affinity [107]. This method the use of broad-spectrum antibacterial agents may result in a
can maximize the chances of success in producing new drive for the use of phage therapy as an alternative narrow-
antibiotics which reduces the cost of developing new an- spectrum antibacterial treatment. In such a situation, it will
tibiotics empirically. Ligands can be developed with dif- be important to design a cocktail of phages that can be made
ferent degrees of selectivity. For instance, if the binding specifically for each bacterial species or even strain, so that
sites of the bacterial target and human protein are availa- only phages that are effective against the bacterial strains
ble, they can be superimposed in order to identify features responsible for the infection will be used.
that are distinctive to the bacterial binding site. This can
In addition to combination therapy and phage therapy,
be used to favour the ligand toward the bacterial target
there is also increasing interest in nano-technology for medi-
and minimize the side effects of the antibiotic. Further- cal applications such as specific drug targeting, cell labeling
more, resistance in bacteria sometimes arises from the
and gene therapy. However, the full spectrum of the antibac-
mutation of the bacterial target that results in weakened
terial mechanisms elicited by nanoparticles (NPs) is still un-
affinity for the antibiotic. In such a situation, ligands that
certain and requires further research. For example, antibacte-
target both the mutant and wild type binding sites can be
rial activity is attributed to oxidative stress, this may not be
developed by focusing on conserved structures in the
the case for all nanoparticle formulations and some of the
binding sites of the two targets [108]. antibacterial mechanism may not be related to oxidative
Studies undertaken using each of the alternatives de- stress. It is challenging to study the antibacterial actions of
scribed with examples are summarized in Table 1. nanoparticles as there is no single analytical method which
fulfills the criterias for obtaining all the necessary infor-
CONCLUSION mation. Experience with chemotherapeutic drugs to treat
cancer has taught us that, in vitro studies seldom mimic in
Focused efforts and sound research are needed to over- vivo conditions. Thus, it is impossible to anticipate the spe-
come the challenges that will be posed by the continuous cific antibacterial action of NPs by depending on in vitro
advancement of AMR. Since the development of new anti- bacterial cell culture alone. The mechanism and safety of
microbials takes time, in the immediate future, it is more membrane crossing NPs have yet to be fully studied. Moving
practical to focus on improving and augmenting existing forward, researchers have proposed that endocytosis by bac-
antimicrobials. Important strategies include combination teria, which resembles what is observed in eukaryotic cells,
therapies which help to circumvent the problems that cause could be a mechanical movement of the nanoparticles into
existing antimicrobials to fail, in effect, by supplementing bacterial cells. Nevertheless, no findings have yet been re-
them with the missing link in their effective lytic action. Re- ported regarding this topic. The current most reasonable
search also needs to be streamlined on understanding the mechanism is that bacteria cells exposed to the lower con-
toxicity of potential adjuvant therapies in animal and human centration of nanoparticles experience some damage to their
cells. The mechanisms by which resistance to common anti- structure including the removal of the LPS layer. With the
biotics is developed needs to be fully elucidated, so that ap- LPS removed, the inner cell membrane protrudes in the form
proaches to overcoming resistance can be designed. The of vesicular structures from the cell surface. The vesicles
primary challenge of combination therapy is the ability to bind to nanoparticles that later enter the cell by electrostatic
develop customized techniques that are optimized for the attraction [93]. Furthermore, it is also worth addressing the
isolation and purification of safer and newer natural antimi- intracellular inhibitory mechanisms. Some studies have also
crobials against MDR, as well as having a better understand- considered the action of nanoparticles on gene expression,
ing of the structure, mechanism and function of AMPs, metabolism of the bacterial cells and protein synthesis.
which will lead to better design to mitigate the rise in MDR However, studies addressing this issue remain limited.
pathogens [118]. Moreover, a number of antibiotics have
been put to one side due to their high intrinsic resistance Although there are still many unanswered questions and
rates such as ADEPs. By incorporating these antibiotics in hurdles to be overcome in tackling AMR, the future pro-
combination therapy strategies, new therapies with novel spects for these approaches (combination therapy, phage
resistance profiles that do not overlap, could be introduced therapy nanotechnology and docking studies) for targeted
into clinical use. The main challenges that will be encoun- drug delivery seem promising. Combination therapy presents
tered are the specific toxicity, pharmacokinetic and pharma- several advantages compared to the development of new
codynamic properties that arise out of combination therapies antibiotics, such as decreasing the chance of developing re-
that are not seen when the same drugs are used in monother- sistance that may assist us in the development of novel and
apy [105]. Another key question relevant to combination effective biomarkers [119], although there are issues with
therapy is whether there is a way to identify molecules that drug-drug interactions. In phage therapy, the manipulation of
can act as specific adjuvants for individual antimicrobial phages to overcome pathogens is not as simple as it seems
agents. Furthermore, can adjuvants which do not possess and it is unlikely that there is a definitive approach that is
any intrinsic antimicrobial activity not only enhance the able to treat all bacterial infections. Nanoparticle develop-
effect of co-administered antibiotic but also prevent the ment is another viable alternative to antibiotics which seems
emergence of resistance? There may be any such adjuvants to have high potential to solve the problems encountered in
which have yet to be identified; some phages have greater using conventional antibiotics such as the emergence of
diversity in their mechanisms of action in comparison with multidrug-resistant bacteria. In-depth studies of how nano-
other antibacterial agents, and. phage therapy shows increas- particles exhibit their antibacterial actions could contribute to
ing promise in the treatment of infections. Over an extended developing effectual antibacterial nanoparticles and prevent
Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 15

nanoparticle cytotoxicity. Computational methods have the [12] Michalopoulos AS, Livaditis IG, Gougoutas V. The revival of
capacity to enable the design of new antibiotics to combat fosfomycin. Vol. 15, International Journal of Infectious Diseases.
Elsevier: 2011.
resistant organisms. In order to maximize its potential, this [13] Neu HC, Gootz TD. Antimicrobial Chemotherapy. In: Baron S
approach needs to be integrated into systems biology Medical Microbiology. 4th edition. Galveston (TX): University of
modelling where potential off-target effects on non- Texas Medical Branch at Galveston; 1996; Chapter 11
microbial processes can also be identified at an early stage. [14] Shahab M, Verma M, Pathak M, Mitra K, Misra-Bhattacharya S.
Cloning, expression and characterization of UDP-N-
Modelling the potential for resistance to lead compounds acetylglucosamine enolpyruvyl transferase (MurA) from Wolbach-
allows for compounds with estimated high resistance poten- ia endosymbiont of human lymphatic filarial parasite Brugia mala-
tial to be excluded from investigation at earlier stages in or- yi. PLoS One. 2014; 9: e99884.
der to focus resources more efficiently[108]. To effectively [15] McLuskey K, Cameron S, Hammerschmidt F, Hunter WN. Struc-
overcome the challenges of AMR, antimicrobial compounds ture and reactivity of hydroxypropylphosphonic acid epoxidase in
fosfomycin biosynthesis by a cation- and flavin-dependent mecha-
with new mechanisms of action need to be discovered by the nism. Proc Natl Acad Sci 2005; 102: 14221-6.
scientific community and only through methodical experi- [16] Bhattacharjee MK. Antibiotics That Inhibit Cell Wall Synthesis. In
mental design and perseverance can we fully exploit the po- Chemistry of Antibiotics and Related Drugs, Springer International
tential of the existing approaches to AMR described in this Publishing 2016; 49-94.
[17] Pinho MG, Kjos M, Veening JW. How to get (a) round: Mecha-
review. nisms controlling growth and division of coccoid bacteria. Nat Rev
Microbiol 2013; 11: 601-14.
CONSENT FOR PUBLICATION [18] Liu Y, Breukink E. The membrane steps of bacterial cell wall syn-
thesis as antibiotic targets. Antibiotics 2016; 5: 28.
Not applicable. [19] Kåhrström CT. Bacterial physiology: Flipping out over Mur. J Nat
Rev Microbiol 2014; 12: 595.
[20] Smith JL, Weinberg ED. Mechanisms of Antibacterial Action of
CONFLICT OF INTEREST Bacitracin. J Gen Microbiol 1962; 28(3): 559-69.
The authors declare no conflict of interest, financial or [21] Jha RK, de Sousa SM. Microplate assay for inhibitors of the trans-
peptidase activity of PBP1b of Escherichia coli. J Biomol Screen
otherwise. 2006; 11: 1005-14.
[22] Rioseras B, Yagüe P, López-García MT, Gonzalez-Quiñonez N,
Acknowledgements Binda E, Marinelli F, Manteca A. Characterization of SCO4439, a
D-alanyl-D-alanine carboxypeptidase involved in spore cell wall
This work was funded by the Fundamental Research maturation, resistance, and germination in Streptomyces coelicolor.
Grant Scheme (FRGS/1/2018/SKK11/PERDANA/02/1) by Sci Rep 2016; 6: 21659.
[23] Alberts B, Johnson A, Lewis J. General principles of cell commu-
the Ministry of Education Malaysia and Malaysia Medical nication. Mol Biol Cell 2002; 4.
Association (MMA). The authors would like to thank all the [24] Li J, Koh JJ, Liu S, Lakshminarayanan R, Verma CS, Beuerman
members of Floral Biotechnology Laboratory, UPM. C.L.M RW. Membrane active antimicrobial peptides: Translating mecha-
wrote the review with assistance from S.K.Y. K.S.L, nistic insights to design. Frontiers in Neuroscience. Frontiers Media
S.H.E.L, K. Y, M.A and A. A. W. T. and S.H.E.L signifi- SA 2017; 73.
[25] Bellm L, Lehrer RI, Ganz T. Protegrins: New antibiotics of mam-
cantly refined the manuscript. All authors read and ap- malian origin. Expert Opin Investig Drugs 2000; 9(8): 1731-42.
proved the final manuscript. [26] Hong W, Zeng J, Xie J. Antibiotic drugs targeting bacterial RNAs.
Acta Pharm Sin B 2014; 4: 258-65.
REFERENCES [27] Lambert T. Antibiotics that affect the ribosome. Rev Sci Tech Off
Int Epiz 2012; 31: 57-64.
[1] Saga T, Yamaguchi K. History of antimicrobial agents and resistant [28] Kotra LP, Haddad J, Mobashery S. Aminoglycosides: Perspectives
bacteria. Japan Med Assoc J 2009; 52: 103-8. on mechanisms of action and resistance and strategies to counter
[2] Ellis A. Overcoming resistance. Overcoming Resist 2002; 5: 5-7. resistance. Antimicrobial Agents and Chemotherapy. American So-
[3] Ventola CL. The antibiotic resistance crisis: part 1: Causes and ciety for Microbiology (ASM) 2000; 3249-3256.
threats. J Formul Manag 2015; 40: 227-83. [29] Chopra S, Reader J. tRNAs as antibiotic targets. Int J Mol Sci
[4] Luyt CE, Bréchot N, Trouillet JL, Chastre J. Antibiotic stewardship 2014; 16: 321-49.
in the intensive care unit. Crit Care 2014; 18: 480. [30] Chopra I, Roberts M. Tetracycline antibiotics: Mode of action,
[5] Viswanathan VK. Off-label abuse of antibiotics by bacteria. Gut applications, molecular biology, and epidemiology of bacterial re-
Microbes 2014; 5: 3-4. sistance. Microbiol Mol Biol Rev 2001; 65: 232-60.
[6] Gebretekle GB, Serbessa MK. Exploration of over the counter sales [31] Constable PD, Hinchcliff KW, Kenneth W, Done SH, Grünberg W,
of antibiotics in community pharmacies of Addis Ababa, Ethiopia: Radostits OM. 2016. Veterinary medicine  : a textbook of the dis-
pharmacy professionals’ perspective. Antimicrob Resist Infect eases of cattle, horses, sheep, pigs and goats. 10th ed. Elsevier
Control 2016; 5: 2. Saunders: 2007.
[7] Ayukekbong JA, Ntemgwa M, Atabe AN. The threat of antimicro- [32] Odds FC, Brown AJP, Gow NAR. Antifungal agents: Mechanisms
bial resistance in developing countries: causes and control strate- of action. Trends Microbiol 2003; 11: 272-9.
gies. Antimicrob Resist Infect Control 2017; 6: 47. [33] Vodnala M, Ranjbarian F, Pavlova A, de Koning HP, Hofer A.
[8] Goel PK, Ross-Degnan D, McLaughlin T.J, Soumerai SB. Influ- Trypanosoma brucei methylthioadenosine phosphorylase protects
ence of location and staff knowledge on quality of retail pharmacy the parasite from the antitrypanosomal effect of deoxyadenosine:
prescribing for childhood diarrhea in Kenya. Int J Qual Health Care Implications for the pharmacology of adenosine antimetabolites. J
1996; 8: 519-26. Biol Chem 2016; 291: 11717-26.
[9] Bartlett JG, Gilbert DN, Spellberg B. Seven ways to preserve the [34] Kimberlin DW, Whitley RJ. Antiviral therapy of HSV-1 and -2. In:
Miracle of antibiotics. Clin Infect Dis 2013; 56: 1445-50. Arvin A, Campadelli-Fiume G, Mocarski E, et al. Human Herpes-
[10] Spellberg B, Gilbert DN. The future of antibiotics and resistance: A viruses: Biology, Therapy, and Immunoprophylaxis. Cambridge:
tribute to a career of leadership by John Bartlett. Clin Infect Dis Cambridge University Press 2007; Chapter 64.
2014; 59: S71-S75. [35] Parenti F, Lancini G. Antibiotic and Chemotherapy. Rifamycins. 9th
[11] Vollmer W, Blanot D, De Pedro MA, et al. Structural and mecha- ed. Elsevier 2011; pp. 861-900.
nistic basis of penicillin-binding protein inhibition by lactivicins. [36] Collin F, Karkare S, Maxwell A. Exploiting bacterial DNA gyrase
FEMS Microbiol Rev 2005; 30: 565-9. as a drug target: current state and perspectives. Appl Microbiol
Biotechnol 2011; 92: 479-97.
16 Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 Moo et al.

[37] Bhattacharjee MK. 2016. Chemistry of Antibiotics and Related [58] Fritsche TR, Castanheira M, Miller GH, Jones RN, Armstrong ES.
Drugs. Springer International Publishing 2016; pp. 60-94. Detection of methyltransferases conferring high-level resistance to
[38] Hitchings GH. Mechanism of action of trimethoprim- aminoglycosides in Enterobacteriaceae from Europe, North Ameri-
sulfamethoxazole-I. J Infect Dis 1973; 128: S433-S436. ca, and Latin America. Antimicrob. Agents Chemother 2008; 52:
[39] Qi J, Virga KG, Das S, Zhao Y, Yun MK, White SW, Lee RE. 1843-5.
Synthesis of bi-substrate state mimics of dihydropteroate synthase [59] Vetting MW, Hegde SS, Wang M, Jacoby GA, Hooper DC,
as potential inhibitors and molecular probes. Bioorganic Med Blanchard JS. 2011. Structure of QnrB1, a plasmid-mediated fluo-
Chem 2011; 19: 1298-305. roquinolone resistance factor. J Biol Chem 2011; 286: 25265-73.
[40] Blair JMA, Webber MA, Baylay AJ, Ogbolu DO, Piddock LJV. [60] Lynch JP, Clark NM, Zhanel GG. 2013. Evolution of antimicrobial
Molecular mechanisms of antibiotic resistance. Nat Rev Microbiol resistance among Enterobacteriaceae (focus on extended spectrum
2014; 13: 42-51. β-lactamases and carbapenemases). Expert Opin Pharma-
[41] Liu A, Tran L, Becket E, et al. Antibiotic sensitivity profiles de- cother 2013; 14: 199-210.
termined with an Escherichia coli gene knockout collection: gener- [61] Queenan AM, Shang W, Flamm R, Bush K. Hydrolysis and inhibi-
ating an antibiotic bar code. Antimicrob. Agents Chemother 2010; tion profiles of beta-lactamases from molecular classes A to D with
54: 1393-403. doripenem, imipenem, and meropenem. Antimicrob. Agents
[42] Blake KL, O’Neill AJ. Transposon library screening for identifica- Chemother 2010; 54: 565-9.
tion of genetic loci participating in intrinsic susceptibility and ac- [62] Tzouvelekis LS, Markogiannakis A, Psichogiou M, Tassios PT,
quired resistance to antistaphylococcal agents. J Antimicrob Daikos GL. Carbapenemases in Klebsiella pneumoniae and Other
Chemother 2013; 68: 12-6. Enterobacteriaceae: an evolving crisis of global dimensions. Clin
[43] Wozniak RAF, Waldor MK. Integrative and conjugative elements: Microbiol Rev 2012; 25: 682-707.
mosaic mobile genetic elements enabling dynamic lateral gene [63] Wright G. Bacterial resistance to antibiotics: Enzymatic degrada-
flow. Nat. Rev. Microbiol 2010; 8: 552-63. tion and modification. Adv Drug Deliv Rev 2005; 57: 1451-70.
[44] Baroud M, Dandache I, Araj GF, et al. Underlying mechanisms of [64] Yap PSX, Yang SK, Lai KS, Lim SHE. Essential Oils: The Ulti-
carbapenem resistance in extended-spectrum β- lactama- mate Solution to Antimicrobial Resistance in Escherichia
se-producing Klebsiella pneumoniae and Escherichia coli isolates coli? In Escherichia coli - Recent Advances on Physiology, Patho-
at a tertiary care centre in Lebanon: role of OXA-48 and genesis and Biotechnological Applications. Croatia: InTech 2017;
NDM-1 carbapenemases. Int J Antimicrob Agents 2013; 41: 75-9. pp. 299-313.
[45] Munita JM, Arias CA, Unit AR, Santiago A De. Mechanisms of [65] Qin S, Wang Y, Zhang Q, et al. Identification of a novel genomic
Antibiotic Resistance. Microbiol Spectr 2016; 4(2): 1-37. island conferring resistance to multiple aminoglycoside antibiotics
[46] Dolejska M, Villa L, Poirel L, Nordmann P, Carattoli A. Complete in Campylobacter coli. Antimicrob Agents Chemother 2012; 5:
sequencing of an IncHI1 plasmid encoding the carbapenemase 5332-9.
NDM-1, the ArmA 16S RNA methylase and a resistance- [66] Kaur I. Novel Strategies to Combat Antimicrobial Resistance. J
nodulation-cell division/multidrug efflux pump. J Antimicrob Infect Dis Ther 2016; 4: 292.
Chemother 2013; 68: 34-9. [67] Campbell J, Singh AK, Santa Maria JP, et al. Synthetic lethal com-
[47] Piddock LJV. Clinically relevant chromosomally encoded multi- pound combinations reveal a fundamental connection between wall
drug resistance efflux pumps in bacteria. Clin Microbiol Rev 2006; teichoic acid and peptidoglycan biosyntheses in Staphylococcus
1: 382-402. aureus. ACS Chem Biol 2011; 6: 106-16.
[48] Su CC, Long F, Zimmermann MT, Rajashankar KR, Jernigan RL, [68] Sieradzki K, Tomasz A. Suppression of beta-lactam antibiotic
Yu EW. Clinically relevant chromosomally encoded multidrug re- resistance in a methicillin-resistant Staphylococcus aureus through
sistance efflux pumps in bacteria. Nature 2011; 470: 558-62. synergic action of early cell wall inhibitors and some other antibiot-
[49] Eicher T, Cha H, Seeger MA, et al. Transport of drugs by the mul- ics. J Antimicrob Chemother 1997; 39: 47-51.
tidrug transporter AcrB involves an access and a deep binding [69] Lee N, Yuen KY, Kumana CR. Clinical role of beta-lactam/beta-
pocket that are separated by a switch-loop. Proc Natl Acad Sci U S lactamase inhibitor combinations. Drugs 2003; 63: 1511-24.
A 2012; 109: 5687-92. [70] Vannuffel P, Cocito C. Mechanism of action of streptogramins and
[50] Zalucki YM, Dhulipala V, Shafer WM. Dueling regulatory proper- macrolides. Drugs 1996; 51 Suppl 1: 20-30.
ties of a transcriptional activator (MtrA) and repressor (MtrR) that [71] Gordon NC, Png K, Wareham DW. Potent synergy and sustained
control efflux pump gene expression in Neisseria gonorrhoeae. bactericidal activity of a vancomycin-colistin combination versus
MBio 2012; 3: e00446-12. multidrug-resistant strains of Acinetobacter baumannii. Antimicrob
[51] Abouzeed YM, Baucheron S, Cloeckaert A. ramR mutations in- Agents Chemother 2010; 54: 5316-22.
volved in efflux-mediated multidrug resistance in Salmonella en- [72] De Pauw BE, Deresinski SC, Feld R, Lane-Allman EF, Donnelly
terica serovar Typhimurium Antimicrob. Agents Chemother 2008; JP. Ceftazidime compared with piperacillin and tobramycin for the
52: 2428-34. empiric treatment of fever in neutropenic patients with cancer: A
[52] Gao W, Chua K, Davies JK, et al. Two novel point mutations in multicenter randomized trial. Ann Intern Med 1994; 120: 834-44.
clinical Staphylococcus aureus reduce linezolid susceptibility and [73] Schentag JJ, Jusko WJ. Gentamicin Persistence In The Body; 1977;
switch on the stringent response to promote persistent infection. 486.
PLoS Pathog 2010; 6: 1000944. [74] Servais H, Jossin Y, Van Bambeke F, Tulkens PM, Mingeot-
[53] Billal DS, Feng J, Leprohon P, Légaré D, Ouellette M. Whole Leclercq MP. Gentamicin causes apoptosis at low concentrations in
genome analysis of linezolid resistance in Streptococcus renal LLC-PK 1 cells subjected to electroporation. Antimicrob
pneumoniae reveals resistance and compensatory mutations. BMC Agents Chemother 2006; 50: 1213-21.
Genomics 2011; 12: 512. [75] Paul M, Dickstein Y, Schlesinger A, Grozinsky-Glasberg S, Soa-
[54] Katayama Y, Ito T, Hiramatsu K. A new class of genetic element, res-Weiser K, Leibovici L. Beta-lactam versus beta-lactam-
staphylococcus cassette chromosome mec, encodes methicillin re- aminoglycoside combination therapy in cancer patients with neu-
sistance in Staphylococcus aureus. Antimicrob Agents Chemother tropenia. Cochrane Database of Systematic Reviews 2013; Issue 6.
2000; 44: 1549-55. [76] Hendrix RW, Smith MCM, Burns RN, Ford ME, Hatfull GF. Evo-
[55] Kumar N, Radhakrishnan A, Wright CC, et al. Crystal structure of lutionary relationships among diverse bacteriophages and pro-
the transcriptional regulator Rv1219c of Mycobacterium tubercu- phages: All the world’s a phage. Proc Natl Acad Sci 1999; 96:
losis. Protein Sci 2014; 23: 423-32. 2192-7.
[56] Long KS, Poehlsgaard J, Kehrenberg C, Schwarz S, Vester B. The [77] Ormälä AM, Jalasvuori M. Phage therapy: Should bacterial re-
Cfr rRNA methyltransferase confers resistance to phenicols, lin- sistance to phages be a concern, even in the long run? Bacte-
cosamides, oxazolidinones, pleuromutilins, and streptogramin A riophage 2013; 3: e24219.
antibiotics. Antimicrob Agents Chemother 2006; 50: 2500-5. [78] Keen EC. Phage therapy: Concept to cure. Front. Microbiol 2012;
[57] Zhang WJ, Xu XR, Schwarz S, et al. Characterization of the In- 3: 238.
cA/C plasmid pSCEC2 from Escherichia coli of swine origin that [79] Hyman P, Abedon ST. Bacteriophage host range and bacterial
harbours the multiresistance gene cfr. J Antimicrob Chemother resistance. Advances in applied microbiology 2010; 217-48.
2014; 69: 385-9. [80] Chan BK, Abedon ST, Loc-Carrillo C. Phage cocktails and the
future of phage therapy. Future Microbiol 2013; 8: 769-83.
Mechanisms of Antimicrobial Resistance (AMR) and Alternative Approaches Current Drug Discovery Technologies, 2019, Vol. 16, No. 0 17

[81] Lu TK, Collins JJ. Engineered bacteriophage targeting gene net- [101] Allen TM. Liposomal drug formulations. Rationale for develop-
works as adjuvants for antibiotic therapy. Proc Natl Acad Sci 2009; ment and what we can expect for the future. Drugs 1998; 56: 747–
106: 4629-34. 756.
[82] Duerkop BA, Huo W, Bhardwaj P, Palmer KL, Hooper LV. Mo- [102] Singla S, Harjai K, Raza K, Wadhwa S, Katare OP, Chhibber S.
lecular basis for lytic bacteriophage resistance in enterococci. Phospholipid vesicles encapsulated bacteriophage: A novel ap-
MBio 2016; 7: e01304-16. proach to enhance phage biodistribution. J. Virol. Methods 2016;
[83] Abedon ST, Kuhl SJ, Blasdel BG, Kutter EM. Phage treatment of 236: 68– 76.
human infections. Bacteriophage 2011; 1: 66-85. [103] Azam A, Ahmed AS, Oves M, Khan MS, Habib SS, Memic A.
[84] Wittebole X, De Roock S, Opal SM. A historical overview of bac- Antimicrobial activity of metal oxide nanoparticles against Gram-
teriophage therapy as an alternative to antibiotics for the treatment positive and Gram-negative bacteria: a comparative study. Int. J.
of bacterial pathogens. Virulence 2016; 5: 226-35. Nanomedicine 2012; 7: 6003–6009.
[85] Miedzybrodzki R, Fortuna W, Weber-Dabrowska B, Górski A. [104] Möhler JS, Sim W, Blaskovich MAT, Cooper MA, Ziora ZM.
Phage therapy of staphylococcal infections (including MRSA) may Silver bullets: A new lustre on an old antimicrobial agent. Biotech-
be less expensive than antibiotic treatment. Postepy Hig Med Dosw nol Adv 2018; doi:10.1016/j.biotechadv.2018.05.004
2007; 61: 461-5. [105] Möhler JS, Kolmar T, Synnatschke K, Hergert M, Wilson LA,
[86] Maiques E, Úbeda C, Tormo MÁ, Ferrer MD, Lasa Í, Novick RP, Ramu S, et al. Enhancement of antibiotic-activity through com-
Penadés JR. Role of staphylococcal phage and SaPI integrase in in- plexation with metal ions - Combined ITC, NMR, enzymatic and
tra- and interspecies SaPI transfer. J. Bacteriol 2007; 189: 5608-16. biological studies. J Inorg Biochem. 2017;167:134–41.
[87] Drulis-Kawa Z, Majkowska-Skrobek G, Maciejewska B, Delattre [106] Kumar M, Curtis A, Hoskins C. Application of nanoparticle tech-
AS, Lavigne R. Learning from bacteriophages - advantages and nologies in the combat against anti-microbial resistance. Pharma-
limitations of phage and phage-encoded protein applications. Curr ceutics 2018; 10(1): 1–17.
Protein Pept Sci 2012; 13: 699-722. [107] Phillips MA, Stewart MA, Woodling DL, Xie ZR. Has Molecular
[88] Van Bambeke F, Pages JM, Lee VJ. Inhibitors of bacterial efflux Docking Ever Brought us a Medicine? In: Molecular Docking.
pumps as adjuvants in antibiotic treatments and diagnostic tools for 2018; pp. 141-178.
detection of resistance by efflux. Recent Pat. Antiinfect. Drug Dis- [108] Young MJ, Mahfouz TM. New Bacterial Targets and Computa-
cov 2006; 1: 157-75. tional Methods Against Bacterial Resistance. Medical Research
[89] Lomovskaya O, Warren MS, Lee A, et al. Identification and char- Archives 2017;5(4): 1-24.
acterization of inhibitors of multidrug resistance efflux pumps in [109] Fischbach MA. Combination therapies for combating antimicrobial
Pseudomonas aeruginosa: Novel agents for combination thera- resistance. Current Opinion in Microbiology 2011; 519–523.
py. Antimicrob Agents Chemother 2001; 45: 105-16. [110] Yang SK, Yusoff K, Mai CW, Lim WM, Yap WS, Lim SH, Lai
[90] Nelson ML, Levy, SB. Reversal of tetracycline resistance mediated KS. Additivity vs synergism: Investigation of the additive interac-
by different bacterial tetracycline resistance determinants by an tion of cinnamon bark oil and meropenem in combinatory therapy.
inhibitor of the Tet(B) antiport protein. Antimicrob. Agents Molecules 2017; 22: 1733.
Chemother 1999; 43: 1719-24. [111] Yang SK, Yap PSX, Krishnan T, Yusoff K, Chan KG, Yap WS,
[91] Beyth, N, Houri-Haddad,Y, Domb A, Khan W, Hazan R. Alterna- Lai KS and Lim SHE. Mode of action: Synergistic interaction of
tive antimicrobial approach: Nano- antimicrobial materials. Evi- peppermint (Mentha x piperita L. Carl) essential oil and mero-
dence-based Complementary and Alternative Medicine 2015; 1-16. penem against plasmid-mediated resistant E. coli. Rec Nat Prod
[92] Pelgrift RY, Friedman AJ. Nanotechnology as a therapeutic tool to 2018;12(6):582–94.
combat microbial resistance. Advanced Drug Delivery Re- [112] Kutateladze M, Adamia R. Bacteriophages as potential new thera-
views 2013; 1803-15. peutics to replace or supplement antibiotics. Trends Biotechnol
[93] Wang L, Hu C, Shao L. The antimicrobial activity of nanoparticles: 2010; 28: 591–595.
present situation and prospects for the future. Int J Nanomedicine [113] Kalle AM, Rizvi A. Inhibition of bacterial multidrug resistance by
2017; 12:1227-49. celecoxib, a cyclooxygenase-2 inhibitor. Antimicrob. Agents
[94] Mühling M, Bradford A, Readman JW, Somerfield PJ, Handy RD. Chemother 2011; 55: 439–442.
An investigation into the effects of silver nanoparticles on antibi- [114] Ma Y, Zhou T, Zhao C. Preparation of chitosan–nylon-6 blended
otic resistance of naturally occurring bacteria in an estuarine membranes containing silver ions as antibacterial materials. Carbo-
sediment. Mar. Environ. Res 2009; 68: 27-283. hydr Res 2008, 343(2):230–7.
[95] Qi G, Li L, Yu F, Wang H. Vancomycin-modified mesoporous [115] Drulis-Kawa Z, Dorotkiewicz-Jach A. Liposomes as delivery sys-
silica nanoparticles for selective recognition and killing of patho- tems for antibiotics. Int. J. Pharm 2010; 387: 187–198.
genic gram-positive bacteria over macrophage-like cells. ACS [116] Thapa R, Bhagat C, Shrestha P, Awal S, Dudhagara P. Enzyme-
Appl. Mater. Interfaces 2013; 5: 10874–10881. mediated formulation of stable elliptical silver nanoparticles tested
[96] Qiu Z, Yu Y, Chen Z, Jin M, Yang D, Zhao Z, Wang J, Shen Z, against clinical pathogens and MDR bacteria and development of
Wang X, Qian D, Huang A, Zhang B, Li JW. Nanoalumina pro- antimicrobial surgical thread. Ann Clin Microbiol Antimicrob
motes the horizontal transfer of multiresistance genes mediated by 2017; 16(1): 39.
plasmids across genera. Proc. Natl. Acad. Sci 2012; 109: 4944– [117] Shaikh S, Rizvi SMD, Shakil S, Hussain T, Alshammari TM, Ah-
4949. mad W, et al. Synthesis and characterization of cefotaxime conju-
[97] Sharma A, Kumar Arya D, Dua M, Chhatwal GS, Johri AK. Nano- gated gold nanoparticles and their use to target drug-resistant CTX-
technology for targeted drug delivery to combat antibiotic re- M-producing bacterial pathogens. J Cell Biochem 2017; 118(9):
sistance. Expert Opin. Drug Deliv 2012; 9: 1325–1332. 2802–8.
[98] Azuma K, Izumi R, Osaki T, Ifuku S, Morimoto M, Saimoto H, [118] Mandal SM, Roy A, Ghosh AK, Hazra TK, Basak A, Franco OL.
Minami S, Okamoto Y. Chitin, chitosan, and its derivatives for Challenges and future prospects of antibiotic therapy: From pep-
wound healing: old and new materials. J. Funct. Biomater 2015; 6: tides to phages utilization. Frontiers in Pharmacology. Frontiers
104–142. Media SA 2014; 105.
[99] Huh AJ, Kwon YJ. ‘Nanoantibiotics’: A new paradigm for treating [119] Yang SK, Low LY, Yap PSX, Yusoff K, Mai CW, Lai KS, et al.
infectious diseases using nanomaterials in the antibiotics resistant Plant-derived antimicrobials: Insights into mitigation of antimicro-
era. J. Control. Release 2011; 156: 128–145. bial resistance. Rec Nat Prod. 2018;12(4):295–316.
[100] Gillies E, Frechet J. Dendrimers and dendritic polymers in drug
delivery. Drug Discov. Today 2005; 10: 35–43.

View publication stats

You might also like