Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

Critical Reviews in Clinical Laboratory Sciences

ISSN: 1040-8363 (Print) 1549-781X (Online) Journal homepage: https://www.tandfonline.com/loi/ilab20

Designing and optimizing new antimicrobial


peptides: all targets are not the same

Adriana Barreto-Santamaría, Manuel E. Patarroyo & Hernando Curtidor

To cite this article: Adriana Barreto-Santamaría, Manuel E. Patarroyo & Hernando Curtidor (2019):
Designing and optimizing new antimicrobial peptides: all targets are not the same, Critical Reviews
in Clinical Laboratory Sciences, DOI: 10.1080/10408363.2019.1631249

To link to this article: https://doi.org/10.1080/10408363.2019.1631249

Published online: 09 Aug 2019.

Submit your article to this journal

Article views: 35

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ilab20
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES
https://doi.org/10.1080/10408363.2019.1631249

REVIEW ARTICLE

Designing and optimizing new antimicrobial peptides: all targets are not
the same
Adriana Barreto-Santamarıaa,b , Manuel E. Patarroyoa,c and Hernando Curtidora,b
a
Fundacion Instituto de Inmunologıa de Colombia - FIDIC, Receptor-Ligand Department, Bogota D.C, Colombia; bUniversidad del
Rosario, School of Medicine and Health Sciences, Bogota D.C., Colombia; cUniversidad Nacional de Colombia - Bogota, Faculty of
Medicine, Bogota D.C., Colombia

ABSTRACT ARTICLE HISTORY


Because the resistance of microorganisms to the available antibiotics is a growing healthcare Received 10 December 2018
problem worldwide, the search for new antimicrobial peptides (AMPs) that provide useful thera- Revised 5 May 2019
peutic options has been increasing in importance. Many initial candidates have had to be dis- Accepted 10 June 2019
carded after having advanced to the preclinical and clinical stages. This has led to substantial Published online 9 August
2019
losses in terms of time and money. For that reason, the essential characteristics of AMPs (i.e.
their activity, selectivity, stability in physiological conditions and low production cost) must be KEYWORDS
considered in their design. In addition, peptides could be active against several kinds of cells Antimicrobial peptides;
with activity and selectivity resulting from interaction with multiple target cell components, minimum inhibitory
which sometimes are present in mammalian cells as well. Thus, the cellular composition is concentration (MIC);
important in the AMP-target cell interaction and must be considered in the design of AMPs, too. antimicrobial activity;
This review describes general aspects of AMP design, limitations concerning their therapeutic hemolytic activity;
application, and optimization strategies for overcoming such limitations. selectivity; SAR study

Abbreviations: Aa: amino acid; AMP: antimicrobial peptide; CAMPs: cationic antimicrobial pepti-
des; CL: cardiolipin; LPC: lysophosphatidylcholine; LPS: lipopolysaccharide; LTA: lipoteichoic acid;
MIC: minimal inhibitory concentration; MOA: mechanism of action; PA: phosphatidic acid; PC:
phosphatidylcholine; PE: phosphatidylethanolamine; PG: phosphatidylglycerol; PI: phosphatidyli-
nositol; PIP2: phosphatidylinositol 4,5-bisphosphate; PS: phosphatidylserine; RBC: red blood cell;
SAR: structure-activity relationship; SPPS: solid-phase peptide synthesis; STAMPs: specifically tar-
geted antimicrobial peptides; TA: teichoic acid; TCS: two-component regulatory systems; GlcCer:
glucosylceramide.

Introduction therapeutic agents with different mechanisms of action


(MOA) that act effectively against the pathogens
The resistance of microorganisms to most currently
responsible for these infections [4].
available antimicrobials is a worldwide public health
Peptides that have antimicrobial activity have been
problem [1]. This resistance represents a large expend-
iture in terms of cost-effectiveness and health because isolated from plant and animal species since the 1980s;
hospital-acquired infections sometimes induce other they are considered natural antibiotics and are called
injuries or diseases such as sepsis that require add- antimicrobial peptides (AMPs) [5]. Many natural AMPs
itional treatment or that can lead to death [2]. have been used as models to make synthetic AMPs,
Around 31.5 million cases of sepsis, 19.4 million thereby showing these molecules’ versatility in terms of
cases of severe sepsis and 5.3 million deaths occur being improved through design. Although resistance to
annually worldwide as a result of infections caused by some AMPs has been reported (i.e. staphylococci’s pro-
antibiotic-resistant pathogens [2]. It has been estimated teolytic defense mechanisms induced by the anionic
that if effective strategies are not introduced to coun- AMP, dermicidin) [6], AMPs are of great interest in clin-
teract such phenomena, such infections will cause ical research because of their broad spectrum of activ-
around ten million deaths annually by 2050 [3]. Hence ity, the ability to combine multiple MOAs, and their
there is urgent need to develop regulatory strategies rapid activity that minimizes resistance developing to
concerning antibiotic use and to search for new these molecules [7,8].

CONTACT Hernando Curtidor hernando.curtidor@urosario.edu.co Fundacion Instituto de Inmunologıa de Colombia - FIDIC, Carrera 50 No. 26-20,
Bogota D.C., Colombia.
ß 2019 Informa UK Limited, trading as Taylor & Francis Group
2 A. BARRETO-SANTAMARIA ET AL.

However, despite the large number of studies on the mechanisms of microorganisms [13]. They consist of
development of AMPs, few AMPs have advanced to the amino acid (aa) sequences having activity against a var-
stage of clinical trial and/or therapeutic use, partly due iety of microorganisms; many AMPs are broad spectrum,
to the pharmaceutical industry’s interests and the sup- while some have narrow spectrum activity, meaning that
posedly disadvantageous characteristics of these types they have been classified as antibacterial, antifungal,
of molecules [9,10], e.g. high production costs, suscepti- antiparasitic or antiviral peptides (Figure 1) [16].
bility to modification by enzymes, or lack of superiority Although AMPs are found naturally in almost all life
regarding currently available agents [9,11,12]. For a forms, from bacteria (e.g. bacteriocins produced by
peptide to be considered as a therapeutic option, it Lactobacillus [17]) to mammals, most have been isolated
must have powerful antimicrobial properties and low or from different animal species; e.g. cecropins come from
zero hemolytic and cytotoxic activities (high selectivity), insects [18,19] and magainins and dermaseptins have
which is why obtaining AMPs that have high selectivity been isolated from frog skin [20–22]. A considerable
has gained great importance [13,14,15]. It has been number of AMPs, such as the puroindolines and some
argued that a major difficulty concerning AMP develop- defensins, have been obtained from plants [23,24].
ment lies in the fact that candidates have been pro- Unfortunately, many naturally occurring AMPs have
moted to preclinical and clinical trials too quickly, certain limitations, e.g. low selectivity, which results in
without having been fully optimized, thereby leading toxic effects on host cells, or the requirement for a high
to them being discarded [9]. concentration for their antimicrobial activity. Thus, sev-
This paper reviews relevant aspects of AMP design, eral approaches to AMP design have been gaining
limitations regarding their use as therapeutic agents, increasing relevance as a strategy for enhancing AMP
and some optimization strategies that have been sug- development [25,26].
gested for overcoming such limitations early on, high-
lighting the importance of optimization during the
Synthetic AMPs
early stages of AMP development, i.e. in the laboratory.
Finally, the diverse composition of the potential target Limitations of naturally occurring AMPs have hampered
cells of AMPs and their relevance in the design process their therapeutic use. The search for a new generation
are reviewed. of AMPs has focused on computational design of pepti-
des or on the selection of new candidates from large
peptide libraries [27–31]. Such designed or selected
Natural AMPs
sequences can be produced by chemical synthesis or
Natural AMPs have been widely studied because they recombinant expression [32,33] for testing in antimicro-
represent an alternative to the antibiotic resistance bial, hemolytic and cytotoxic activity assays, thereby

Figure 1. Biological activities described to date for AMPs. These peptides are active against different types of microorganism and
against cancer cells. Such peptides are sometimes characterized by having an immunomodulatory or anti-inflammatory effect due
to binding to certain pathogen components such as LPS from Gram-negative bacteria.
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 3

enabling their selectivity for microorganisms and their within the therapeutic industry [47]. Such results can be
therapeutic potential to be ascertained [34,35]. presented as MIC (concentration inhibiting 100% of the
Although AMP design is complex due to the consid- growth of a particular strain), or MIC100, MIC90 or
erable number of natural aa and possible combinations MIC50 values (concentrations inhibiting 100% of the
depending on peptide length (e.g. 4  1015 possible growth of 100%, 90% or 50% of a considerable number
combinations for a 12 aa sequence), this could be used of evaluated strains) (Table 1).
for overcoming natural limitations of AMPs [10,34,36]. The MIC values for AMPs being developed range
Table 1 shows how candidates having potent activity from <0.03 lg/mL to 32 lg/mL (Table 1), supporting
can be obtained through effective design. These mole- their potential as a therapeutic alternative to currently
cules’ scope goes beyond their use in clinical therapy as available antibiotics whose MIC varies between 0.5 lg/
some AMPs have been studied for alternative applica- mL and 4 lg/mL [48]. For example, the lipoglycopep-
tions, e.g. the amidated antifungal peptide H-Orn-Orn- tide, dalbavancin, has a  0.03 mg/mL MIC against
Trp-Trp-NH2 has been evaluated regarding its use as a Staphylococcus aureus (Gram-positive strain) whilst the
preservative against food contamination [37]. glycopeptide antibiotic, vancomycin, has a 0.5–2 lg/
mL MIC against the same type of bacteria [49]. It is
important to note that as AMPs are generally molecu-
Antimicrobial activity of AMPs larly larger than antibiotics, their molecular weights,
Evaluating a peptide’s antimicrobial activity during the and consequently, their MIC values in mg/mL, are usu-
experimental stage consists of determining its activity ally higher too. MIC values in lmol/L should thus be
on the microorganism of interest (bacteria, fungi, para- used for a more apt comparison between AMPs and
sites and/or viruses), the minimal inhibitory concentra- antibiotics.
tion (MIC) assay being the standard method [38]. Although a  16 lg/mL or  16 lmol/L MIC has been
Different culture conditions may be required to ensure posed as a requirement for a candidate to be evaluated
optimum microorganism growth and to obtain reliable in clinical studies [10], this may vary depending on the
results [39–42]. Certain methodological conditions pathogen against which it acts and the advantages it
are also required, because of the implications of work- may offer compared to the available agents against the
ing with cationic peptides; e.g. the need to use same pathogen. For example, pexiganan, a synthetic
Mueller–Hinton broth without cation adjustment for 22 aa-long cationic peptide derived from magainin
the MIC assay against bacteria, since these ions (Caþþ (currently in Phase 3 clinical trials), has emerged as a
or Mgþþ) can cause substantial inhibition of cationic topical therapeutic option for mild infections associated
AMP activity [38]. with diabetic foot ulcers. Pexiganan has MIC values
The MIC value obtained is a crucial factor in the ini- of 8–16 mg/mL against Escherichia coli, Klebsiella pneumo-
tial selection of candidate peptides, because the US niae, Citrobacter koseri, Enterobacter cloacae, Acinetobacter
Food and Drug Administration (FDA) approves a new species and Pseudomonas aeruginosa (Table 1).
antimicrobial for use in humans only if it has thera-
peutic advantages over already available agents. AMPs Designing AMPs with improved antimicrobial activ-
must thus be effective at concentrations equal to or ity and selectivity
less than that of available antibiotics to justifying the AMPs can theoretically be designed de novo or from
enormous investment in time and money required to one or several preexisting sequences that can be used
fully develop a new therapeutic agent [43]. as a template, with the aim of obtaining a sequence
Higher concentrations of AMPs are usually required with powerful activity and high selectivity [11,50–56].
to inhibit pathogen growth than those for already avail- Structure-activity relationship (SAR) studies form
able antibiotics. Their design thus requires an approach an integral part of AMP research, where charge, second-
which involves constantly seeking to improve these ary structure, and amphipathicity are the physicochemi-
molecules’ antimicrobial activity and to understand the cal properties most closely related to AMP activity
impact of certain modifications on such activity in order [25,54,57–60]. Some studies have focused on establish-
to optimize new candidates in the early stages of ing the MOA of natural and designed AMPs, highlight-
laboratory development [9,34,44–46]. ing interactions with the phospholipids forming the
An overview of MIC values for peptides that have membrane over binding to receptors [44,45,61].
reached advanced stages of development is useful for Membrane-AMP contact may induce changes, such
establishing MIC ranges, to decide whether a peptide as conformational transition, self-association or oligo-
enters the clinical study phase and becomes profiled merization, in the peptide, thereby increasing specific
4

Table 1. MIC Values in vitro for some AMPs which are currently being developed.
Administration MIC against Gram- MIC against Gram-
AMP Description Indication route Activity negative bacteria positive bacteria MIC against fungi Reference
Pexiganan A 22 aa-long Diabetic foot infection Topical Pathogens associated 8–16 mg/mLc 8–32 lg/mLc – [62–64]
magainin analog with diabetic foot (3.23–6.46 mmol/L) (3.23–12.9 mmol/L)
infection
(broad spectrum)
Novarifyn Synthetic antimicrobial MRSA, P. aeruginosa, C. Topical Mainly against 31.25 lg/mLb 62.5 lg/mLb – [65]
(NP432) difficile, A. Acinetobacter
baumannii and E. baumannii and
coli. infections Staphylococcus
aureus
Dalbavancin Lipoglycopeptide Acute bacterial Intravenous Gram-positive bacteria, – 0.03 mg/mLc – [62,66–68]
A. BARRETO-SANTAMARIA ET AL.

(approved by antibiotic infections of the mainly (0.016 mmol/L)


the FDA) skin and acute Staphylococcus
hematogenous aureus,
osteomyelitis including MRSA
Brilacidin Defensin mimetic Oral mucositis in Topical Multiple pathogens, 0.25–0.5 lg/mLc – [69]
patients undergoing Gram-negative and (0.27–0.53 mmol/L)
Chemo-radiation for Gram-positive
treating head and bacteria, including
neck cancer MRDS. Mainly
against
Staphylococcus
aureus
HB1345 Synthetic Acne, broad- Topical Mainly against 0.5–1 lg/mLa – [70]
lipohexapeptide spectrum antibiotic Propionibacterium
acnes
Surotomycin Cyclic lipopeptide Diarrhoea Oral Mainly against – <1 mg/mLc – [71,72]
Clostridium difficile (<0.6 mmol/L)
Lytixar (LTX-109) Synthetic antimicrobial Uncomplicated Gram- Topical Staphylococcus aureus 2–4 lg /mLa 4 lg/mLb – [73]
peptidomimetic positive skin (2.5–5.1 mmol/L)
infections, MRSA/
MSSA nasal colonies
AP138 Plectasin derivative MRSA Not specified Mainly against 2–4 lg/mLa [74]
implant infections Staphylococcus (0.45–0.9 mmpl/L)
aureus
Omiganan Indolicidin-derived 12 Papulopustular Topical Staphylococcus aureus – 16 lg/mLc – [75]
(CLS001) aa-long synthetic rosacea, acne (8.99 mmol/L)
cationic peptide vulgaris, vulvar
intraepithelial
neoplasia (VIN)
MU1140 Lantibiotic naturally Gram-positive bacteria Not specified Broad spectrum 0.5–32 lg/mLa – [76]
produced by the (MRSA, C. difficile) against Gram-
SMaRT strain parent positive bacteria
C16G2 35 aa-long designed Mouthwash having Topical Streptococcus mutans 4.2–5.2 mmol/La [77]
peptide derived selective elimination (17–21 lg/mL)
from the S. mutans of the oral
competence- cariogenic pathogen
stimulating peptide Streptococcus
(CSP) pheromone mutans
and novispirin bound
by a flexible tri-
glycine linker region
(continued)
Table 1. Continued.
Administration MIC against Gram- MIC against Gram-
AMP Description Indication route Activity negative bacteria positive bacteria MIC against fungi Reference
PAC-113 12 aa-long Oral candidiasis in HIV Topical Candida albicans – – 1.3–3.1 lg/mL [78]
antimicrobial seropositive patients (0.83–1.98 mmol/L)
peptide derived
from a naturally
occurring histatin
protein found
in saliva
Novamycin Poly-arginine based Yeast and Topical Mainly against 8–32 lg/mLb [79]
(NP339) cationic peptide mold infections Staphylococcus
aureus and
Aspergillus spp.
NovexatinR Cyclic, highly cationic Fungal nail infection Topical Saccharomyces – – 3 mmol/La (3.28 mg/mL) [80]
(NP213) peptide cerevisiae
(arginine/rich)
SGX942 Synthetic 5-aa peptide, Oral mucositis in head Intravenous Effective for tackling – – – [81]
innate defense and neck cancer. Gram-negative (i.e.
regulator (IDR), Complement for Burkholderia
lacking direct antibiotics. pseudomallei) and
antimicrobial Gram-positive (i.e.
activity Staphylococcus
aureus)
bacterial infections
p2TA (AB103) 10 aa-long designed Necrotizing soft Intravenous Attenuates T-cell CD28 – – – [82]
synthetic peptide tissue infections signaling in
Contains D-aa exacerbated
immune responses
during bacterial
infection. Effective
in mice challenged
with E. coli bacteria
The range of MIC values concerning several microorganisms is shown for broad-spectrum peptides.
a
MIC, bMIC100, cMIC90, dMIC50.
The values calculated from the reported MIC and the peptide sequence’s molecular weight are shown in brackets.
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES
5
6 A. BARRETO-SANTAMARIA ET AL.

cell toxicity [83]. This is why circular dichroism has been antimicrobial and hemolytic activity of the modified
used for studying the structure of AMPs and their rela- peptides [97].
tionship with biological activity. These assays are car- Increasing the positive charge of a molecule can
ried out by simulating the microorganisms’ membrane reduce its hemolytic activity [14]; however, when such
conditions using media containing trifluoroethanol, charge is increased (particularly in a peptide’s hydro-
anionic surfactant sodium dodecyl sulfate, or more philic region), this can result in increased antimicrobial
complex components such as liposomes, lipopolysac- and hemolytic activity [98].
charides (LPS) or whole bacteria [36,84,85]. These stud-
ies are the basis for developing useful strategies to Structure. AMPs can have or assume differing second-
modify physicochemical properties of AMPs and ary structures, including a-helix, b-sheet, extended or
improve their antimicrobial potential, as shown below. mixed structures [99,100]. AMPs with an a-helical struc-
ture are usually more active on microbial membranes
Sequence. Amino acid composition and primary but can have activity on red blood cell (RBC) mem-
sequence are critical for the peptides’ antimicrobial activ- branes as well [13,97]. It has been reported that altering
ity since they determine the sequence’s structural and helix peptide structure by introducing D-aa, prolines or
physicochemical properties. Peptides having antimicro- glycines into their sequences can reduce hemolytic
bial activity have been reported to contain mainly leu- activity [36,45,101,102]. However, such modifications do
cine, glycine, and lysine in their sequences, and having not enhance selectivity for microorganisms in all cases,
arginine or lysine with tryptophan favors the peptides’ since selectivity may be modified only moderately by
insertion into the membrane of the pathogen [86–88]. In the structural changes, or antimicrobial activity may
addition, modifying aa side-chains can affect the anti- decrease [101,103]. Disulfide bond formation for struc-
microbial activity of AMPs, as occurs with arylation of tural alteration has been more effective in some cases,
tryptophan residues, which has been shown to increase because it leads to stronger structural change, as seen
the antimicrobial activity of a human lysozyme-derived in modified LK peptide analogs where hemolytic activ-
AMP [89]. N-methylation of peptide backbone amides ity has been reduced while antimicrobial activity has
can lead to derivatives having similar antimicrobial activ- not been greatly affected [103].
ity but less hemolytic activity [90], whilst partially replac-
ing L-aa with D-aa can reduce hemolytic activity and Hydrophobicity. A peptide’s hydrophobicity refers to
increase antimicrobial activity [36,91]. the percentage of hydrophobic aa in its sequence. This
characteristic enables water-soluble AMPs to bind (via
Charge. Although neutral [92] and anionic [93] AMPs hydrophobic aa) to the membrane and divide the lipid
have been reported, positively-charged peptides (þ1 to bilayer. Although a molecule’s optimum hydrophobicity
þ9) are usually more active because they are electro- value varies according to its other characteristics, the
statically attracted by microbial membranes that hydrophobicity of most natural AMPs is around 50%
are negatively-charged [94]; this gives rise to a subcat- [104]. Increasing peptide hydrophobicity (by aa substi-
egory of AMPs called cationic antimicrobial peptides tution or lipid conjugation) could induce greater
(CAMPs) [95,96]. antimicrobial activity because it favors interaction with
AMP charge can be increased by replacing phospholipid membranes [105–107] and it could
uncharged aa with positively-charged aa or replacing increase sequence selectivity and stability if non-natural
acid residues with positively-charged or amide-type res- hydrophobic aa are involved [102]. However, increasing
idues; the latter approach has even conferred anti- hydrophobicity too much could lead to the formation
microbial activity on originally inactive peptides [96]. of peptide aggregates that would reduce its capability
Amidating the C-terminal extreme is one strategy for antimicrobial action [98]. Increasing cationic and
for increasing peptide charge without modifying its amphipathic peptide hydrophobicity could increase
sequence; it consists of leaving a –CONH2 group affinity for zwitterionic phospholipids (neutral) on the
(neutral at physiological pH) instead of –COOH mammalian cell membrane, thereby increasing hemo-
(negatively-charged at physiological pH), thereby lytic activity [35].
increasing a peptide’s net charge by þ1 [87]. Such an
amidation strategy has also stabilized the secondary Amphipathicity. Amphipathicity is a feature whereby
structure of peptides having a significant helical struc- peptides have a spatial aa distribution conferring both a
ture element content, thus favoring interaction with the hydrophilic and a hydrophobic face. This property is
phospholipid bilayer, which may lead to increased measured as a peptide’s hydrophobic moment (lH);
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 7

values range from 0 to 3.26, and it is calculated according obtained, i.e. charge of 5 to þ10, hydrophobicity of
to a standard scale of hydrophobicity for every aa [108]. 10–80%, and length of 5–174 aa (preferably less than
Amphipathicity has been shown to be very import- 60 aa) [86].
ant for AMP activity and selectivity for peptides having In general, predictive methods developed from
an a-helix structure but also for peptides having a these databases have been based on parameter
b-sheet structure [11,13,109,110]. The increased amphi- space strategy, database filtering technology, align-
pathicity of helical peptides (i.e. greater definition of ment, and machine learning (i.e. artificial neural net-
hydrophilic and hydrophobic faces) has been associated works, genetic algorithms, support vector machine, and
with activity on membranes and reducing it could lead hidden Markov models), among others (Figure 2)
to peptides having reduced hemolytic activity [14]. [113–120]. Methods based on machine learning and
However, there are some discrepancies regarding the combined methods have played important roles in
effect of perfect or imperfect amphipathicity on AMP such approaches, thereby providing greater scope for
function and selectivity; designed peptides having making predictions [121,122].
imperfect amphipathicity could be more active and The predictors used for peptide computational
selective than those having perfect amphipathicity [25]. design are usually based on information regarding
hydrophobicity, charge distribution, and size [86]; how-
Computational AMP design ever, some new approaches have brought greater pre-
cision to designing AMPs. For example, an innovative
AMP activity and selectivity result from many intercon-
membrane-pore-forming peptide prediction tool/
nected properties, thereby hampering any prediction
descriptor, based on the peptides’ topological hydro-
regarding the effect on the physical-chemical properties
phobic distribution, gives more in-depth information
caused by modifying one aa and also on their anti-
about aa spatial distribution features and patterns, as
microbial and hemolytic activity [106]. To overcome
well as about hydrophobicity, charge, and length [123].
such complexity, designing AMPs has been supported
A guideline for designing a computational model for
by AMP sequences and their characteristics being
designing effective nontoxic antibacterial peptides has
deposited in databases, as well as by using computa-
been suggested recently; it includes molecular docking
tional tools for predicting properties of peptide sequen-
and molecular dynamics simulations for improving AMP
ces and their possible activity on the membranes of
activity predictions [124].
microorganisms [86,111,112].
However, it should be noted that, in spite of their
Several databases have been created to date for
great usefulness for obtaining AMPs, such methods for
managing the available information on AMPs; they
rational peptide design in silico are strongly influenced
sometimes include tools for analyzing and predicting
by the information encoded in the sequences depos-
sequences having antimicrobial activity. For example,
ited in the databases, thereby limiting their capability
the Yet Another Database of Antimicrobial Peptides
for identifying novel sequences beyond those described
(YADAMP), the Database of Anuran Defense Peptides
for AMPs in the pertinent literature [119]. Even after
(DADP) and the Linking Antimicrobial Peptides (LAMP)
overcoming the stage of optimizing peptide activity
database specialize mainly in peptides isolated from
and selectivity, there is still a long way to go before
specific sources [86].
they can be used as therapeutic agents [43].
The Collection of Anti-microbial Peptide 3 (CAMP3)
database (created in 2010), with 10,247 AMP sequences
Tailor-made?
[113], together with the Antimicrobial Peptide Database
3 (APD3) (created in 2003), with 2747 antimicrobial Although the physicochemical characteristics of candi-
sequences from the six kingdoms (mainly animal and date sequences are fundamental when designing new
vegetal), have been the most used to date. APD3 has AMPs, their activity and selectivity also depend on tar-
rigorous parameters for introducing new sequences, get cell composition, particularly the outer membrane
which can be only from natural sources, thereby confer- surface which forms the initial pathogen-AMP contact
ring great reliability on the data deposited there [114]. point. Such targets must thus be considered when
APD3 includes a secondary structure and antimicro- designing and optimizing AMPs.
bial activity predictor; it can also calculate some pepti- AMPs can act against bacteria, fungi, parasites or
des’ physical-chemical characteristics, such as their viruses via different MOA, including anionic internal tar-
composition, charge, and hydrophobicity. It has set gets such as DNA [125], RNA [126] or regulatory
ranges in which such properties must be maintained enzymes [127]. However, the main mechanism that
for increasing the probability of an AMP being AMPs use is disruption of the membrane, not by
8 A. BARRETO-SANTAMARIA ET AL.

Figure 2. Databases and prediction methods regarding the computational design of peptides. The main AMP databases are shown
on the left-hand side of the Figure. The main methods for predicting AMP activity are shown on the right-hand side; they are based
on the information stored in the databases. ANN: Artificial Neural Networks; GA: Genetic Algorithms, SVM: Support Vector Machine;
HMM: Hidden Markov Models.

binding to specific receptors but by interacting with Eukaryotic membranes also contain glycolipids and
the phospholipids forming it [44,45,128], and they act sterol; the sterol in mammalian cells is cholesterol,
in a similar way against RBC and other animal cells which is found symmetrically on both sides of the
[15,61,129]. AMPs have a clear preference for nega- bilayer, while the glycolipids are preferentially located
tively-charged lipids; they have the greatest affinity for on its outer face [134,136]. The loss of fragmentation
phosphatidylglycerol (PG), followed by phosphatidic activity caused by antibacterial peptides (including pex-
acid (PA), phosphatidylserine (PS) and cardiolipin (CL) iganan) in assays involving phospholipid vesicles has
[130,131]. However, other non-phospholipid compo- been related to an increase in the vesicles’ cholesterol
nents such as wall components or LPS should not be content, so sterol seems to be crucial for discrimination
ruled out as they may be crucial for AMP activity [8]. between bacterial and animal cells [137]. The PS con-
tent of mammalian cells varies from 8–15% of the total
Mammalian cells. A good peptide candidate must molar content of phospholipids; this becomes translo-
have minimal or no activity against mammalian cells so cated to the membrane’s outer face because of aging,
that it can be used therapeutically without any restric- injury or disease, thereby increasing its distribution and
tions [14,54]. However, because microorganisms share producing a negative charge on its surface. This PS
certain membrane characteristics and components with translocation has deep implications regarding cell func-
mammalian cells, many AMPs are poorly selective, i.e. tion [134,138]. Such negative surface charges enable
they are active against microorganisms and mammalian cationic AMPs to affect the cells of microorganisms
cells (toxicity) [14,25,132]. and/or cells altered by injury or disease, while AMPs
Mammalian cell membranes are rich in zwitterionic have low toxicity for healthy mammalian cells.
phospholipids phosphatidylcholine (PC) and phosphati-
dylethanolamine (PE), negatively-charged phospholi- Bacteria. The bacterial membrane is formed by nega-
pids PS and phosphatidylinositol (PI), and sphingolipids tively-charged phospholipids such as PG, CL (lacking in
mainly containing choline (sphingomyelin) [133]. These mammalian cells) and PS (found in mammalian cells),
lipids are segregated on opposite faces of the bilayer, thereby favoring initial electrostatic interaction with
producing asymmetry [134]; PE, PI, and PS are preferen- positively-charged aa of cationic AMPs [136,139].
tially located on the inner face of erythrocyte mem- Hydrophobic interactions subsequently occur between
branes whilst the external face is enriched by PC and AMP apolar aa and the hydrophobic tails of the phos-
sphingolipids [134,135]; this means that the cell surface pholipids making up the membrane; a peptide thus
tends to have a net neutral charge [136]. becomes inserted into the lipid bilayer and disrupts it,
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 9

Figure 3. The components of target cells involved in AMP activity. AMPs are active against different microorganism and animal
cells, where various phospholipid and non-phospholipid components are crucial for defining their activity and selectivity. Anionic
cell components are important for the initial electrostatic interactions between an AMP and a target cell. The arrow next to the
phospholipid membrane composition indicates a reduction in phospholipid component abundance. Three membranes can be dis-
tinguished in parasitic cells: (1) the parasite’s membrane (P), (2) the parasitophorous vacuole (PV), and (3) the host cell (H). Viral
genetic information is covered by the nucleocapsid (nucleocap); this nucleocapsid, in turn, is surrounded by a phospholipid mem-
brane in covered viruses that is not present in naked ones.

with or without pore formation [44,129]. However, and LPS (a complex glycolipid acting as the first contact
AMPs must first cross certain barriers that protect the site of AMPs) are on the external face whilst lipopro-
bacteria to reach the bacterial cytoplasmic membrane teins are present on the inner face. There is a high zwit-
(which differs between Gram-negative and Gram-posi- terionic phospholipid PE content and somewhat lower
tive bacteria) (Figure 3). Gram-negative bacteria have a levels of negatively-charged phospholipid PG [141].
permeable external membrane and a more robust The LPS of the external face involve three compo-
internal membrane that gives rise to a thin peptidogly- nents: a hydrophobic lipid portion called lipid A, a central
can cell wall in the middle; this is a polysaccharide polysaccharide, and a variable terminal polysaccharide,
formed by a peptide and N-acetyl muramic acid and N- which includes sugar acids that provide the molecule
acetylglucosamine linked by b-1,4 bonds [140]. with a negative charge. Initial interaction is thus facili-
The outer membrane of Gram-negative bacteria has tated through the positively-charged aa of cationic AMPs
an asymmetric composition; proteins such as porins [141,142], thereby enabling their contact with the
10 A. BARRETO-SANTAMARIA ET AL.

external membrane and subsequent passage through antimicrobial efficacy since AMPs can become retained
permeabilization of the lipid bilayer [143,144]. Increasing in the wall. This would inhibit their accumulation on
the positive charge of AMPs seems to favor interaction the membrane that is needed for disruption in the lipid
with LPS; e.g. MBI-27 (26 aa) and MBI-28 (28 aa), peptides bilayer [150].
active against Gram-negative bacteria (Pseudomonas aer- A fundamental difference between bacterial and
uginosa), interact with LPS, but MBI-28 is more powerful mammalian cells is the transmembrane potential, which
due to two additional lysine residues in the C-terminal refers to the electrochemical gradient produced by the
region [145]. velocity of ion exchange through the membrane. The
The internal membrane also has high PE content transmembrane potential in normal mammalian cells is
(but less than that of the outer membrane) and has a 90 to 110 mV compared to 130 to 150 mV in
relatively low amount of PG; Escherichia coli contains pathogenic bacteria; this difference could be important
60–80% PE and 15–20% PG, along with CL in some in the recognition by and selectivity of AMPs [104].
cases [141,146]. The activity of some AMPs may be Bacteria are able to induce resistance to AMPs due to
related to the bacterial inner membrane PE content changes in their membrane composition; such changes
that could, in turn, be related to these peptides’ low could be due to flippase expression which can modify
selectivity, because they also act on human cells such cytoplasmic membrane composition, making it more sta-
as RBC, which contain PE [132,134]. ble against AMPs [151]. For example, when an AMP inter-
Gram-positive bacteria have a single membrane cov- acts with PG-containing membranes, bacteria can increase
ered by a thick wall of peptidoglycan and an outermost CL or aminoacylated lipid levels. CL is a negatively-
protein layer called the S layer [140,142] (Figure 3). Some charged phospholipid that may protect lipid membranes
AMPs (particularly lantibiotics, a family of small polycyclic from permeabilization [152], and the aminoacylated lipid
antimicrobial peptides such as nisin, that are too short to can introduce positive charges (lysines) that reduce the
pass through the membrane) act as complexes with lipid affinity of the AMPs for the membrane [151]. Changes in
II (bacterial wall precursor) and thus inhibit synthesis of membrane rigidity or a reduction of its negative charge
the wall in Gram-positive bacteria [147]. by translocation of positively-charged phospholipids from
The PE content is low in the plasmatic membrane of the internal face have been found to be related to bacter-
Gram-positive bacteria and there is a large amount of ial resistance against AMPs [152]. The two-component
PG so they have a higher negative charge on their sur- regulatory systems (TCS) are important in the develop-
face; e.g. the PE content of Staphylococcus aureus is less ment of bacterial resistance. They perceive a stimulus in
than 10%, compared to 80% PG and 10% CL [146]. The the environment and activate signal transduction to
higher negative charge on the membrane of Gram-posi- induce modifications in the membrane, making it more
tive bacteria could be related to the greater activity of resistant to the action of AMPs [153]. Strategies must thus
AMPs on this type of bacteria. In fact, one limitation of be sought, taking into account the TCS, to reduce the
most AMPs is their lack of activity or poor activity appearance of resistance to AMPs.
against Gram-negative pathogens that cause many hos-
pital infections [9]. This is reflected in the activity of Fungi. Fungi are eukaryotic microorganisms; their cells
AMPs found in clinical studies, which are mostly against are thus very similar to human cells, making it more dif-
Gram-positive bacteria (Table 1). ficult to develop drugs having selective toxicity against
The outer surface of Gram-positive bacteria contain them. The fungal lipid bilayer mainly consists of sphin-
teichoic (TA) and lipoteichoic (LTA) acids; LTA, in par- golipids, PE, and PI; such components are also present
ticular, cross the wall and become exposed on the bac- in mammalian cells. However, fungal sphingolipids
terial surface. They play an important role in bacterial mainly contain inositol as monosaccharide and are dis-
growth and physiology and contribute towards mem- tinct from those in mammalian cells [154]; the sphingo-
brane homeostasis (cell osmoprotection) and bacterial lipid glucosylceramide has particularly been proposed
virulence [148]. It has been suggested that LTA may be as an important target for AMP selective activity against
important for the activity of AMPs because modifica- fungi (Figure 3) [154].
tions like D-alanylation can confer resistance on the Fungal phospholipid composition is also asymmetric;
bacteria by increasing the density of its wall [149]. It e.g. the internal face of the plasma membrane of
has also been proposed that AMPs interact with the Saccharomyces cerevisiae is enriched with PE, PI and PS,
wall’s negatively charged components, even with TA as in erythrocytes [135]. However, ergosterol is these
and LTA; however, interaction with LTA may not be microorganisms’ main neutral lipid; this is a vital lipid
favorable for AMP activity as this may reduce that could be involved in antifungal AMP activity and
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 11

selectivity since it is not present in mammalian cells phospholipids; an intermediate layer called the nucleocap-
[114,155,156]. Some AMPs having antifungal activity do sid is made up of proteins and contains a virus’ genetic
have toxicity against mammal cells and this has been information. Naked viruses do not have this membranous
associated with their preference for cholesterol [157]. envelope, so the nucleocapsid forms its surface. It has
This suggests the high specificity of some AMPs that been proposed that AMPs having antiviral activity exert
may be selective for a particular sterol (cholesterol or such action through three main mechanisms: (1) inhibiting
ergosterol) despite their great structural similarity [158]. viral binding and viral cell membrane fusion by viral aggre-
Other characteristics unique to fungi could be gation; (2) altering the viral envelope by lipid perturbation
important for AMP activity. For example, one compo- in the membrane; and (3) inhibiting viral replication
nent of yeast cell walls is chitin, a polysaccharide con- [166,167]. Some AMPs act as important immunomodula-
sisting of N-acetylglucosamine units linked by b-1,4 tors for counteracting viral infections [168]. Substituting a
bonds [159]. The interaction of antibiotics with chitin single aa in some peptides having antiviral and immuno-
has been related to a loss of membrane integrity in modulatory activity, e.g. replacing an arginine with a lysine
fungi so that the interaction with chitin could enhance in retrocyclin-1 [169] or leucine with a glycine in LL-37
AMP activity [160]. For example, the NP-1 peptide binds derivative GI20, has led to increased antiviral activity [170].
to chitin and causes leaks through the membrane, Hydrophobicity and charge have been highlighted as
resulting in fungal death [161]. important properties, particularly in hapivirins and diprovir-
The yeast cell wall consists mainly of intertwined ins, where their increase has given rise to peptides having
b-1,3-glucan fibrils that form a network of glucans cov- greater antiviral activity [168].
ering the cell surface; there is also a-galactomannan, AMPs are more effective against a covered than a
progressively filling the intrafibrillar spaces, and the naked virus [167] because membrane disruption is a
polysaccharide, laminarin, which is in the wall’s external viable neutralization mechanism [166].
layer. b-1,3-glucan fibrils have b-1,6-glucan branching Interestingly, the composition of mammalian mem-
which appears to be involved in the interconnection brane cells infected by viral particles does not vary sig-
with other wall components (including chitin) [162]. nificantly from that of uninfected cells [171]; although
Glucan is more abundant than mannan; e.g. glucan viral membranes are derived from host cells, they can
makes up 60–65% of total polysaccharides in Candida differ significantly by lipid or protein translocation
albicans, while mannan constitutes just 20–25% [163]. It towards the viral surface and by reduced membrane
has been reported that AMPs may have selective anti- fluidity. A virus can be selective for the host cell mem-
fungal activity that is attributed to their binding to glu- brane regions used in forming its cell envelope, prefer-
can, particularly to laminarin, thereby inhibiting cell ring certain lipid rafts. For example, the cholesterol
wall synthesis and generating its thinning [164]. It has content of the viral membrane can be increased com-
been observed that AMP interaction with fungal wall pared to that of host cell, and this has been shown to
components favors subsequent electrostatic interaction be important for a virus’ infective capacity [171]. PC
with negatively-charged phospholipids like PI, which content (the most abundant zwitterionic phospholipid
can lead to membrane disruption and/or cell entry in mammalian cells) usually becomes reduced and PS
where AMPs can affect intracellular targets [155]. and sphingolipid content increased in the viral mem-
Proteins such as mannoprotein and enzymes charac- brane, which could be related to AMP activity [172].
teristic of these microorganisms could also serve as select- Antiviral AMPs such as human neutrophil peptide 1
ive AMP targets [154]. This has been reported for defensin (HNP1) (a defensin that acts against several naked viral
NaD1 from the tobacco plant, which seems to interact species such as HPV and covered species such as HIV)
with mannoprotein of the cell wall hyphae and requires [173,174], can inactivate covered viruses by interacting
wall components to be able to exert its action on the with O- or N-glucans in viral surface glycoproteins in a
membrane and then enter the cell to accelerate cell death lectin-dependent manner [167]. They can thus alter the
[164]. Peptides such as lactoferrin can even reach the ability of glycoproteins to bind to their receptors on tar-
mitochondria in fungal cells, leading to cell death by get cells. Although not a lot is known about naked
apoptosis [165]. D-aa, particularly short peptides 2–10 aa- viruses, it has been suggested that such peptides could
long, may favor passage through the fungal wall [10]. inhibit virion escape from endocytic vesicles but not vir-
ion binding or internalization [167].
Viruses. The presence or lack of an envelope enables a
virus to be classified as covered or naked. The envelope Parasites. AMPs can be active against parasites or
is a membrane formed by glycoprotein-containing against cells invaded by such parasites [175]; their
12 A. BARRETO-SANTAMARIA ET AL.

activity occurs mainly through action against the mem- Although it has been suggested that the positive
brane of eukaryotic cells [176,177]. The peptides’ ability charge of AMPs is responsible for the initial interactions
to inhibit malarial parasite growth and bacteria has a with anionic components on the target cells, the prefer-
certain similarity since modifying active AMPs against ence of AMPs to interact with one component or
bacteria and parasites affects both activities [178]. another goes beyond just this particular property. The
Host cell protein and lipid permeability, fluidity and selectivity of these sequences seems to be the product
composition become altered when parasites like of multiple factors encompassing both the physico-
Plasmodium falciparum invade [179], thereby inducing chemical characteristics of AMPs and the composition
plasma membrane reorganization and altered cytoadher- of the cells with which they can interact. However, stud-
ence in invaded cells; this could be associated with PS ies of AMPs and the components with which they inter-
exposure on the membrane’s external face [180]. PS con- act in bacteria, parasites, viruses, fungi, and mammal
fers a negative charge which leads to AMP activity against cells have extended our knowledge of their MOAs,
their membrane; e.g. peptide NK-2 is active against the which should help in the design of AMPs that are better
membrane of P. falciparum-infected RBCs, subsequently targeted and enable faster optimization of new candi-
crosses the parasitophorous vacuole, and causes damage dates [118,122,124].
to the merozoite membrane [176]. Such PS exposure is For example, it has been suggested that peptides
not produced by all parasites; e.g. the membrane com- that inhibit exotoxin production in S. aureus could pre-
position of macrophages infected with Leishmania amazo- vent colonization by pathogenic S. aureus while not
nensis amastigotes (oval and non-flagellated parasite in interfering with colonization by normal microbiota
intracellular stage of development) is very similar to that (particularly lactobacilli). In addition, the increasing
of non-infected cells, but there is a significant increase in inhibitory capability of these peptides is related to their
lysophosphatidylcholine (LPC) content while PC and PE increasing charge [184].
content becomes reduced [181]. The parasitophorous On the other hand, optimizing the selectivity of
vacuole’s membrane has a similar composition to that of AMPs so that they bind exclusively to pathogens has
the host cell membrane but with a higher concentration been done successfully without affecting commensal
of PC, which is the most abundant phospholipid, followed organisms by attaching them to targeting peptide
by PE, PI, and PS and smaller amounts of sphingolipids regions. Peptides belonging to this class of molecule
and LPC, as well as host cell membrane and parasite pro- are called specifically targeted antimicrobial peptides
tein components (Figure 3) [181]. (STAMPs); they enable pathogens to be killed and they
It seems that even though parasitic cells have similar produce little or no effect on a host’s normal microbiota
membrane composition to mammalian cells, AMPs [185,186]. A multiple-headed STAMP that has activity
could have a greater affinity for the parasite membrane against Pseudomonas aeruginosa and Streptococcus
than for infected cells; this occurs with a dermaseptin mutans has been shown to eliminate both species from
S4 derivative against P. falciparum [182]. Although the a mixed planktonic culture and have little impact on
parasites’ plasma membrane is rich in zwitterionic PC other bacteria because the targeting region enables
(40% of total phospholipid content in Leishmania mem- their nearly exclusive interaction with these two bacter-
brane) and PE (10%), AMPs interact weakly with these ial species while the antimicrobial region causes bacter-
phospholipids, so the interaction could depend on PS ial death [187]. Such strategies targeting exclusive
(1%) and traces of PG, i.e. lipids with which such inter- components of pathogenic microorganisms could over-
action is stronger [131]. It has been suggested that come the limitations of selectivity of AMPs, thus ena-
increasing the positive charge density of AMPs bling personalized treatment against infectious diseases
increases their selectivity against parasite membranes and mitigating the development of resistance and dam-
and the membranes of parasite-infected cells, as seen age to beneficial microbiota [188].
with the AMP NK-2 that are active against free trypano-
somes or within glioblastoma cells [175]. Multi-purpose AMPs
AMP antiparasitic activity could also involve inter-
Studies of AMPs have shown that these peptides have
action with ergosterol on the membrane of fungi and
other types of biological activity apart from their anti-
parasites such as Leishmania and nematodes; ergosterol
microbial activity, thereby reaffirming their impressive
has been shown to be the target of some non-peptide
therapeutic potential. For example, cationic amphi-
antibiotics [183].
pathic peptide [Leu]10-Dec-NH2 has a short sequence
The diversity of composition of the target cell high- (SLLSLIRKLLT-NH2) that has antimicrobial and anticarci-
lights the complexity involved in designing AMPs. nogenic activity, with high selectivity [189]; LL-37-derived
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 13

short AMPs KR-12-a5 and KR-12- a5 (5-DK) have antibio- [202], and other immune mediators. In addition, anti-
film and anti-inflammatory activity [36]; and peptide inflammatory molecules such as IL-10 also become
AR23 and its analog A(A1R, A8R, I17K) have antibiofilm blocked during the inflammatory resolution stage
and antimicrobial activity [14]. [107,201–204]. The hydrophobicity and positive charge
of AMPs can increase their binding capability to LPS
AMPs having antibiofilm activity. Antibiofilm activity [205]. The antimicrobial activity of AMPs in vivo thus
expands the antimicrobial capability of AMPs and pro- results from their ability to interact directly with patho-
vides an advantage over conventional antibiotics gens and cause cell damage as well as their ability to
against bacteria that form organized bacterial com- modulate innate components of host immunity by
plexes (biofilms), which can be resistant to antibiotics decreasing the inflammatory response caused by
and make conventional treatment against them ineffi- pathogenic organisms and reducing sepsis [201].
cient [14,36,190]. This means that AMPs can be used
alone or in combination with conventional antibiotics
From the laboratory to the therapeutic industry
as effective therapies against bacterial biofilm-forming
strains. AMPs can degrade the biofilm and exert their Although around 12,000 articles focused on the devel-
antimicrobial action, and they can cause planktonic opment of new AMPs have been published annually
bacterial membrane disruption and facilitate the entry between 2012 and 2016 [206], few peptides have
of antibiotics into the cell [191–194]. advanced to the clinical stage or entered therapeutic
use [9,10]. It has been argued that, in some cases, the
AMPs having anticancer activity. Cationic antibacter- FDA has played a crucial role in the precarious progress
ial peptides can also act against cancer cells; this may of AMPs in the industry, whilst in others, it has been
be because the membranes of these two types of cells due to pharmaceutical companies’ commercial interests
(bacterial and cancer cells) have a negative net charge. that has led them to discard promising AMPs and to
The membrane asymmetry of cancer cells is lost and PS focus on more profitable chronic diseases [9]. In 2012,
becomes exposed on the external face; this confers a the FDA launched its Antibacterial Drug Development
negative charge and facilitates AMP activity against them Task Force, which facilitates the development of antibi-
[195]. Antifungal peptides that can bind to PI, i.e. phos- otics and supports the use of innovative assays and
phatidylinositol 4,5-bisphosphate (PIP2), on fungi have alternative measures to assess clinical efficacy, yet still
also been reported; these peptides can also bind to PIP2 maintains prior criteria on the need to demonstrate a
in tumor cells and trigger cell lysis [196]. Some AMPs candidate’s superiority over currently available antimi-
affect internal targets in cancer cells [197]; i.e. in mito- crobials [207]. Two programs have emerged since
chondria, an increase in negative charge from increased then. Combatting Antibiotic Resistance in Europe
amounts of the anionic lipid CL on their internal and (COMBACTE), launched in 2013, included development
external membranes could mediate electrostatic inter- of innovative assays for new antibacterial agents [208],
action with positively-charged peptides and trigger apop- and the Global Action Plan on Antimicrobial Resistance,
tosis [198]. Anticancer AMPs are considered to be the approved by the 68th World Health Assembly in 2015,
next generation of chemotherapeutic drugs due to such included, among its strategic objectives, to increase
properties as their short target cell interaction time that investment in new medicines, diagnostic tools and vac-
reduces the probability of resistance being produced, cines [4,209]. These types of initiatives have renewed
their potential low toxicity that reduces possible side the possibilities for the development of AMPs within
effects, and their good tumor penetration [199]. the pharmaceutical industry and extended the pano-
rama regarding the development of new peptides for
AMPs having anti-inflammatory activity. Sequences treating infections. In 2016, it was anticipated that the
such as peptide LL-37 or lactoferrin-derived peptides world market for antimicrobial peptides would continue
can neutralize the inflammatory potential of pathogen to grow at a 7.5% compound annual rate up to
components [200,201]. An example of this would be 2024 [210].
neutralizing LPS by directly binding to it or preventing
its binding to host cell receptors. In this way, the cell
Limitations of AMPs and strategies for
signaling pathway triggered by Toll-like receptor
overcoming them
ligands in antigen-presenting cells is blocked, culminat-
ing in the blockage of expression of many proinflamma- A major difficulty in developing AMPs has been that
tory cytokines (IL-1, IL-6, TNF-a), chemokines (IL-8) peptides isolated from natural sources were evaluated
14 A. BARRETO-SANTAMARIA ET AL.

and then brought too quickly to preclinical and clinical


studies without the peptides having been fully opti-
mized [9]. A long and expensive process thus ended,
leaving them excluded from the therapeutic industry
and slowing down pharmaceutical development.
Improved strategies for identifying unsuitable candi-
dates early on and reducing the number of failures dur-
ing clinical trials have been sought for some time [211].
Design and modification strategies for overcoming AMP
limitations during early stages are supported by the
versatility of peptide-type molecules, making them
viable alternatives for developing new generations of
antibiotics (Figure 4), where the earliest limitation of
AMPs is their toxicity on mammalian cells. However,
even if such limitations are overcome, there are other
drawbacks, as with all peptide-type drugs, mainly due
to their low stability in physiological conditions and
their production costs.

Stability
Most AMPs cannot be administered orally because they
are rapidly degraded by stomach acids or intestinal pep-
tidases, and are not absorbed to fulfill their biological
function [212]. Although some cyclical and/or linear pep-
tides can be administered orally, most are administered
parenterally (by injection); their use also is limited by
their instability in host conditions [213]. Different strat-
egies have been adopted to increase the half-life of
AMPs in plasma [26], such as cutting sequences or clip-
ping them to limit a peptide’s enzymatic degradation;
this also significantly reduces production costs [10,214].
Another strategy involves identifying possible molecular
excision sites and replacing relevant aa or protecting
them by peptide folding or cyclization [90,215–217].
Stapling is carried out by forming covalent bridges
between side-chains using hydrocarbons, triazole, azo-
benzene, thiol, lactam, hydrazine or thioether [218–220].
Substituting natural aa with N-substituted glycines
(peptoids), lysines and arginines, or b-naphthylalanine
or trans-L-4-hydroxyproline has also been suggested as
a useful strategy for developing anti-infective agents
that are resistant to proteolytic degradation [221–224].
Modifications such as acylation, introduction of albu-
Figure 4. Designing and optimizing peptide sequences for min binding elements, or conjugation with antibodies
obtaining AMPs. Once candidate peptides or sequences have have been used to increase peptide half-life by
been selected, an early design step involves improving their in binding to circulating albumin [225,226]. Isolating AMPs
vitro activity and selectivity based on changes in their physical- directly from natural blood sources has been used as a
chemical properties. This is followed by optimizing AMPs, focus-
new approach for obtaining active and stable pepti-
ing on improving their stability in physiological conditions and
on reducing their production costs. The optimized sequences des [227]. Developing alternate administration
are then submitted to preclinical and clinical studies where they routes, such as intranasal and transdermal [228], or
must be shown to be safe and effective in vivo before they can alternative formulation methods by using liposomes,
be used as therapeutic agents against infectious diseases. micelles, or nanoparticles [229] could increase
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 15

possibilities for using these and other peptide-type be successful for optimizing peptide sequences have
therapeutic agents. been used for addressing the limitations regarding their
therapeutic use.
Production cost Although new antimicrobial development has been
delayed, usually for economic reasons, it is now recog-
Some studies have focused in obtaining AMPs having
shorter sequences to reduce their production costs and to nized that the development of new compounds must
improve their stability in physiological conditions [10,230]. be promoted due to the rapid increase in the resistance
An example is lactoferricin, a 25 aa-long AMP with a 6 aa of microorganisms to traditional antibiotics, and gov-
activity minimal motif [231], or 37 aa-long human catheli- ernment programs have been developed for this pur-
cidin with a 12 aa activity minimal motif [232]. pose. The essential characteristics of AMPs (their
Biosynthesis approaches to produce recombinant activity, selectivity, stability in physiological conditions
peptides are economical and easily scalable in principle and low production cost) must thus be considered from
since AMPs can be produced as recombinants or by the start so that advances can be assured during their
chemical synthesis [32,33]. One such approach uses E. development stages. Useful strategies are also needed
coli as the expression system, but this approach has dif- to improve the design of the sequences to be used as
ficulties concerning yield and cost because AMPs could therapeutic agents, thereby avoiding arduous research
have activity against the bacteria expressing it. with candidates that must be discarded.
Furthermore, AMPs could have a strong interaction There are no universal standards for designing
with anionic components of these bacteria and hamper selective AMPs; however, the design and experimental
the purification process [233]. Work is currently focus- evaluation of current peptide analog sequences has
ing on developing more efficient strategies and expres- enabled information to be obtained regarding possible
sion systems for the large-scale production of AMPs, modifications that should lead to sequences with
such as using the yeast, Pichia pastoris, which reduces improved activity, selectivity and diverse properties. It is
processing costs by omitting cell lysis for obtaining the not enough to consider only a peptide’s properties
aa sequence [234], or expression systems based on when designing AMPs since their activity and selectivity
plants such as barley [235]. result from interaction with multiple components of the
Production by chemical synthesis offers a wide range microorganism. Their activity on the membrane of the
of possibilities for the design and modification of peptides microorganism must also be considered because AMPs
that are not available when using recombinant technolo- may interact with multiple targets that are necessary
gies, e.g. including non-natural aa and strategies for mak- for their antimicrobial activity to be effective.
ing these molecules more stable in physiological Strategies for improving a peptide’s half-life under
conditions [45]. Solid-phase peptide synthesis (SPPS), a physiological conditions and reducing production costs
rapid approach for AMP production [236], enables pep- are also necessary to produce AMPs that will be thera-
tide production in small samples for laboratory assays but
peutically useful.
has been considered unprofitable on a large scale, with
an estimated $100 to $1000 cost for 1 mg of AMP [234].
Despite this, large-scale SPPS has become a viable tech- Disclosure statement
nology for producing small- and medium-sized thera- No potential conflict of interest was reported by the authors.
peutic peptides ranging from around 5 to 50 aa, whilst
convergent synthesis is an option for the obtaining pepti-
des having more than 50 aa [212]. Optimizing SPPS meth- ORCID
ods has gained relevance to produce modified peptides Adriana Barreto-Santamarıa http://orcid.org/0000-0003-
which cannot be obtained by recombinant technologies 3485-6934
and for obtaining cleaner products; however, work must Manuel E. Patarroyo http://orcid.org/0000-0002-1119-6040
Hernando Curtidor http://orcid.org/0000-0002-6556-7812
continue on improving methods for producing peptides
on a large scale [237,238].

References
Conclusions
[1] (a) Ventola CL. The antibiotic resistance crisis: part 1:
AMPs represent promising agents for treating infections causes and threats. P T. 2015;40:277–283.
caused by microorganisms due to their rapid action [2] Fleischmann C, Scherag A, Adhikari NK. Assessment
against them; various modification strategies shown to of global incidence and mortality of hospital-treated
16 A. BARRETO-SANTAMARIA ET AL.

sepsis. current estimates and limitations. Am J Respir peptide with high antibacterial activity. Appl
Crit Care Med. 2016;193:259–272. Microbiol Biotechnol. 2019;103:1765–1775.
[3] O’Neill J. Tackling drug-resistant infections globally: [20] Zasloff M. Magainins, a class of antimicrobial pepti-
Final report and recommendations. Review on anti- des from Xenopus skin: isolation, characterization of
microbial resistance. 2016:84. Availabe from: https:// two active forms, and partial cDNA sequence of a
amr-review.org/sites/default/files/160518_Final%20 precursor. Proc Natl Acad Sci USA. 1987;84:
paper_with%20cover.pdf 5449–5453.
[4] WHO. Global action plan on antimicrobial resistance. [21] Mor A, Hani K, Nicolas P. The vertebrate peptide
2015. Availabe from: https://www.who.int/antimicro- antibiotics dermaseptins have overlapping structural
bial-resistance/global-action-plan/en/ features but target specific microorganisms. J Biol
[5] Haney EF, Mansour SC, Hancock RE. Antimicrobial Chem. 1994;269:31635–31641.
peptides: an introduction. Methods Mol Biol. 2017; [22] Ghosh JK, Shaool D, Guillaud P, et al. Selective cyto-
1548:3–22. toxicity of dermaseptin S3 toward intraerythrocytic
[6] Lai Y, Villaruz AE, Li M, et al. The human anionic anti- Plasmodium falciparum and the underlying molecular
microbial peptide dermcidin induces proteolytic basis. J Biol Chem. 1997;272:31609–31616.
defence mechanisms in staphylococci. Mol Microbiol. [23] Alfred RL, Palombo EA, Panozzo JF, et al. The anti-
2007;63:497–506. microbial domains of wheat puroindolines are cell-
[7] Soblosky L, Ramamoorthy A, Chen Z. Membrane penetrating peptides with possible intracellular
interaction of antimicrobial peptides using E. coli mechanisms of action. PLoS One. 2013;8:e75488.
lipid extract as model bacterial cell membranes and [24] Vriens K, Cammue BP, Thevissen K. Antifungal plant
SFG spectroscopy. Chem Phys Lipids. 2015;187: defensins: mechanisms of action and production
20–33. [Research Support, Non-U.S. Gov’t Review].
[8] Le CF, Fang CM, Sekaran SD. Intracellular targeting Molecules. 2014;19:12280–12303.
mechanisms by antimicrobial peptides. Antimicrob [25] Huang Y, Huang J, Chen Y. Alpha-helical cationic
Agents Chemother. 2017;61:pii: e02340-16. antimicrobial peptides: relationships of structure and
[9] Fox JL. Antimicrobial peptides stage a comeback function. Protein Cell. 2010;1:143–152.
[News]. Nat Biotechnol. 2013;31:379–382. [26] Wang G. Post-translational modifications of natural
[10] Ramesh S, Govender T, Kruger HG, et al. Short anti- antimicrobial peptides and strategies for peptide
microbial peptides (SAMPs) as a class of extraordin- engineering. Curr Biotechnol. 2012;1:72–79.
ary promising therapeutic agents. J Pept Sci. 2016; [27] Kleshchenko YE, Zhigunova AV, Dalin MV, et al.
22:438–451. Peptides selected using phage library variants, effect-
[11] Hu J, Chen C, Zhang S, et al. Designed antimicrobial ively inhibit Trypanosoma cruzi infection. Bull Exp
and antitumor peptides with high selectivity. Biol Med. 2017;163:361–364.
Biomacromolecules. 2011;12:3839–3843. [28] Rondon-Villarreal P, Sierra DA, Torres R. Machine
[12] Rotem S, Mor A. Antimicrobial peptide mimics for learning in the rational design of antimicrobial pepti-
improved therapeutic properties. Biochim Biophys des. Curr Comput Aided Drug Des. 2014;10:183–190.
Acta. 2009;1788:1582–1592. [29] Maccari G, Di Luca M, Nifosı R. In silico design of
[13] Edwards IA, Elliott AG, Kavanagh AM, et al. antimicrobial peptides. Methods Mol Biol. 2015;1268:
Contribution of amphipathicity and hydrophobicity 195–219.
to the antimicrobial activity and cytotoxicity of beta- [30] McCarthy KA, Kelly MA, Li K, et al. Phage display of
hairpin peptides. ACS Infect Dis. 2016;2:442–450. dynamic covalent binding motifs enables facile
[14] Zhang SK, Song JW, Gong F, et al. Design of an development of targeted antibiotics. J Am Chem
alpha-helical antimicrobial peptide with improved Soc. 2018;140:6137–6145.
cell-selective and potent anti-biofilm activity. Sci Rep. [31] Nagarajan D, Nagarajan T, Roy N, et al.
2016;6:27394. Computational antimicrobial peptide design and
[15] Lv Y, Wang J, Gao H, et al. Antimicrobial properties evaluation against multidrug-resistant clinical isolates
and membrane-active mechanism of a potential of bacteria. J Biol Chem. 2017;293:3492–3509.
alpha-helical antimicrobial derived from cathelicidin [32] Wang XJ, Wang XM, Teng D, et al. Recombinant pro-
PMAP-36. PLoS One. 2014;9:e86364. duction of the antimicrobial peptide NZ17074 in
[16] Bahar AA, Ren D. Antimicrobial peptides. Pichia pastoris using SUMO3 as a fusion partner. Lett
Pharmaceuticals (Basel). 2013;6:1543–1575. Appl Microbiol. 2014;59:71–78.
[17] Kasuga G, Tanaka M, Harada Y, et al. Homologous [33] Omardien S, Drijfhout JW, van Veen H, et al.
expression and characterization of Gassericin T and Synthetic antimicrobial peptides delocalize mem-
Gassericin S, a novel class iib bacteriocin produced brane bound proteins thereby inducing a cell enve-
by Lactobacillus gasseri LA327. Appl Environ lope stress response. Biochim Biophys Acta. 2018;
Microbiol. 2019;85:pii: e02815-18. 1860:2416–2427.
[18] Steiner H, Hultmark D, Engstrom A, et al. Sequence [34] Conlon JM, Galadari S, Raza H, et al. Design of
and specificity of two antibacterial proteins involved potent, non-toxic antimicrobial agents based upon
in insect immunity. Nature. 1981;292:246–248. the naturally occurring frog skin peptides, ascaphin-8
[19] Wang M, Lin J, Sun Q, et al. Design, expression, and and peptide XT-7. Chem Biol Drug Des. 2008;72:
characterization of a novel cecropin A-derived 58–64.
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 17

[35] Maturana P, Martinez M, Noguera ME, et al. Lipid and VISA. Antimicrob Agents Chemother. 2018;62:pii:
selectivity in novel antimicrobial peptides: implica- e00157-18.
tion on antimicrobial and hemolytic activity. Colloids [50] Zeitler B, Herrera Diaz A, Dangel A, et al. De-novo
Surf B Biointerfaces. 2017;153:152–159. design of antimicrobial peptides for plant protection.
[36] Kim EY, Rajasekaran G, Shin SY. LL-37-derived short PLoS One. 2013;8:e71687.
antimicrobial peptide KR-12-a5 and its d-amino acid [51] Peng SY, You RI, Lai MJ, et al. Highly potent anti-
substituted analogs with cell selectivity, anti-biofilm microbial modified peptides derived from the
activity, synergistic effect with conventional antibiot- Acinetobacter baumannii phage endolysin LysAB2. Sci
ics, and anti-inflammatory activity. Eur J Med Chem. Rep. 2017;7:11477.
2017;136:428–441. [52] Rathinakumar R, Walkenhorst WF, Wimley WC.
[37] Thery T, O’Callaghan Y, O’Brien N, et al. Optimisation Broad-spectrum antimicrobial peptides by rational
of the antifungal potency of the amidated peptide combinatorial design and high-throughput screen-
H-Orn-Orn-Trp-Trp-NH2 against food contaminants. ing: the importance of interfacial activity. J Am
Int J Food Microbiol. 2018;265:40–48. Chem Soc. 2009;131:7609–7617.
[38] Wiegand I, Hilpert K, Hancock RE. Agar and broth [53] Sitaram N, Chandy M, Pillai VN, et al. Change of glu-
dilution methods to determine the minimal inhibi- tamic acid to lysine in a 13-residue antibacterial and
tory concentration (MIC) of antimicrobial substances. hemolytic peptide results in enhanced antibacterial
Nat Protoc. 2008;3:163–175. activity without increase in hemolytic activity.
[39] Lyu Y, Yang Y, Lyu X, et al. Antimicrobial activity, Antimicrob Agents Chemother. 1992;36:2468–2472.
improved cell selectivity and mode of action of short [54] Chen Y, Mant CT, Farmer SW, et al. Rational design
PMAP-36-derived peptides against bacteria and of alpha-helical antimicrobial peptides with
Candida. Sci Rep. 2016;6:27258. enhanced activities and specificity/therapeutic index.
[40] CLSI CaLSI. Reference method for broth dilution anti- J Biol Chem. 2005;280:12316–12329.
fungal susceptibility testing of filamentous fungi; [55] Munk JK, Uggerhoj LE, Poulsen TJ, et al. Synthetic
Approved Standard - Second Edition. CLSI document analogs of anoplin show improved antimicrobial
M38-A2. Wayne, PA: Clinical and Laboratory activities. J Pept Sci. 2013;19:669–675.
Standards Institute; 2008. [56] Domhan C, Uhl P, Meinhardt A, et al. A novel tool
[41] CLSI CaLSI. Methods for dilution antimicrobial sus- against multiresistant bacterial pathogens – lipopep-
ceptibility tests for bacteria that grow aerobically; tide modification of the natural antimicrobial peptide
Approved Standard – Ninth Edition. CLSI document ranalexin for enhanced antimicrobial activity and
M07-A9. Wayne, PA: Clinical and Laboratory improved pharmacokinetics. Int J Antimicrob Agents.
Standards Institute; 2012. 2018;52:52–62.
[42] Rivas-Santiago B, Rivas Santiago CE, Castaneda- [57] Hwang PM, Vogel HJ. Structure-function relationships
Delgado JE, et al. Activity of LL-37, CRAMP and anti- of antimicrobial peptides. Biochem Cell Biol. 1998;76:
microbial peptide-derived compounds E2, E6 and 235–246.
CP26 against Mycobacterium tuberculosis. Int J [58] Chan DI, Prenner EJ, Vogel HJ. Tryptophan- and
Antimicrob Agents. 2013;41:143–148. arginine-rich antimicrobial peptides: structures and
[43] FDA. The Drug Development Process. 2018. Available mechanisms of action. Biochim Biophys Acta. 2006;
from: https://www.fda.gov/patients/learn-about-drug- 1758:1184–1202.
and-device-approvals/drug-development-process. [59] Abdel Monaim SAH, Jad YE, El-Faham A, et al.
[44] Chou HT, Wen HW, Kuo TY, et al. Interaction of cat- Teixobactin as a scaffold for unlimited new anti-
ionic antimicrobial peptides with phospholipid microbial peptides: SAR study. Bioorg Med Chem.
vesicles and their antibacterial activity. Peptides. 2017;26:2788–2796.
2010;31:1811–1820. [60] Ahn M, Jacob B, Gunasekaran P, et al. Poly-lysine
[45] Sun S, Zhao G, Huang Y, et al. Enantiomeric effect of peptidomimetics having potent antimicrobial activity
d-amino acid substitution on the mechanism of without hemolytic activity. Amino Acids. 2014;46:
action of alpha-helical membrane-active peptides. Int 2259–2269.
J Mol Sci. 2017;19:pii: E67. [61] Barreto-Santamaria A, Curtidor H, Arevalo-Pinzon G,
[46] Gunasekera S, Muhammad T, Stromstedt AA, et al. et al. A new synthetic peptide having two target of
Alanine and lysine scans of the LL-37-derived pep- antibacterial action in E. coli ML35. Front Microbiol.
tide fragment KR-12 reveal key residues for anti- 2016;7:2006.
microbial activity. Chem Biochem. 2018;19:931–939. [62] Amirkia VD, Qiubao P. The Antimicrobial Index: a
[47] Greber KE, Dawgul M. Antimicrobial peptides under comprehensive literature-based antimicrobial data-
clinical trials. Curr Top Med Chem. 2017;17:620–628. base and reference work. Bioinformation. 2011;5:
[48] European Committee for Antimicrobial Susceptibility 365–366.
Testing (EUCAST) of the European Society of Clinical [63] Flamm RK, Rhomberg PR, Simpson KM, et al. In vitro
Microbiology and Infectious Diseases (ESCMID). spectrum of pexiganan activity when tested against
Determination of minimum inhibitory concentrations pathogens from diabetic foot infections and with
(MICs) of antibacterial agents by broth dilution. Clin selected resistance mechanisms. Antimicrob Agents
Microbiol Infect 2003;9:ix–xv. Chemother. 2015;59:1751–1754.
[49] Tran KN, Rybak MJ. Beta-lactams combinations with [64] National Center for Biotechnology Information.
vancomycin provide synergy against VSSA, hVISA, [Internet]. [cited 2018 Apr 13]. Available from:
18 A. BARRETO-SANTAMARIA ET AL.

https://pubchem.ncbi.nlm.nih.gov/compound/ specifically targeted antimicrobial peptide.


16132253 Antimicrob Agents Chemother. 2011;55:3446–3452.
[65] Katvars LK, Mercer DK, O’Neil DA. NovarifynV R [78] Rothstein DM, Spacciapoli P, Tran LT, et al.
(NP432), a novel antimicrobial peptide rapidly active Anticandida activity is retained in P-113, a 12-amino-
against multi-drug-resistant Acinetobacter baumannii acid fragment of histatin 5. Antimicrob Agents
and methicillin resistant Staphylococcus aureus. Chemother. 2001;45:1367–1373.
Vienna (Austria): European Congress of Clinical [79] Katvars L, Smith D, Duncan V, et al. NP339 as a novel
Microbiology and Infectious Diseases (ECCMID); 2017. approach against respiratory fungal infections.
[66] Pfaller MA, Mendes RE, Sader HS, et al. Activity of Vienna (Austria): European Congress of Clinical
dalbavancin tested against Gram-positive clinical iso- Microbiology and Infectious Diseases ECCMID; 2017.
lates causing skin and skin-structure infections in [80] Li S, Breaker RR. Fluoride enhances the activity of
paediatric patients from US hospitals (2014–2015). J fungicides that destabilize cell membranes. Bioorg
Glob Antimicrob Resist. 2017;11:4–7. Med Chem Lett. 2012;22:3317–3322.
[67] Pfaller MA, Flamm RK, Castanheira M, et al. [81] North JR, Takenaka S, Rozek A, et al. A novel
Dalbavancin in vitro activity obtained against gram- approach for emerging and antibiotic resistant infec-
positive clinical isolates causing bone and joint infec- tions: Innate defense regulators as an agnostic ther-
tions in United States and european hospitals apy. J Biotechnol. 2016;226:24–34.
(2011–2016). Int J Antimicrob Agents. 2018;51: [82] Ramachandran G, Kaempfer R, Chung CS, et al. CD28
608–611. homodimer interface mimetic peptide acts as a pre-
[68] Galluzzo M, D’Adamio S, Bianchi L, et al. ventive and therapeutic agent in models of severe
Pharmacokinetic drug evaluation of dalbavancin for bacterial sepsis and gram-negative bacterial periton-
the treatment of skin infections. Expert Opin Drug itis. J Infect Dis. 2015;211:995–1003.
Metab Toxicol. 2018;14:197–206. [83] Usachev KS, Efimov SV, Kolosova OA, et al.
[69] Kowalski RP, Romanowski EG, Yates KA, et al. An Antimicrobial peptide protegrin-3 adopt an antiparal-
independent evaluation of a novel peptide mimetic, lel dimer in the presence of DPC micelles: a high-
Brilacidin (PMX30063), for Ocular Anti-infective. J
resolution NMR study. J Biomol Nmr. 2015;62:71–79.
Ocul Pharmacol Ther. 2016;32:23–27.
[84] Avitabile C, D’Andrea LD, Romanelli A. Circular
[70] Zhang L, Scheicher S, Harris S, et al. Lipohexapeptide
dichroism studies on the interactions of antimicrobial
HB1345: a novel anti-infective for acne. Chicago (IL):
peptides with bacterial cells. Sci Rep. 2014;4:4293.
American Academy of Dermatology Meeting; 2008.
[85] Chai H, Allen WE, Hicks RP. Spectroscopic investiga-
[71] Citron DM, Tyrrell KL, Merriam CV, et al. In vitro
tions of the binding mechanisms between antimicro-
activities of CB-183,315, vancomycin, and metronida-
bial peptides and membrane models of
zole against 556 strains of Clostridium difficile, 445
Pseudomonas aeruginosa and Klebsiella pneumoniae.
other intestinal anaerobes, and 56
Bioorg Med Chem. 2014;22:4210–4222.
Enterobacteriaceae species. Antimicrob Agents
[86] Wang G. Improved methods for classification, predic-
Chemother. 2012;56:1613–1615.
tion, and design of antimicrobial peptides. Methods
[72] Snydman DR, Jacobus NV, McDermott LA. Activity of
a novel cyclic lipopeptide, CB-183,315, against resist- Mol Biol. 2015;1268:43–66.
ant Clostridium difficile and other Gram-positive aer- [87] Jin L, Bai X, Luan N, et al. designed tryptophan- and
obic and anaerobic intestinal pathogens. Antimicrob lysine/arginine-rich antimicrobial peptide with thera-
Agents Chemother. 2012;56:3448–3452. peutic potential for clinical antibiotic-resistant
[73] Saravolatz LD, Pawlak J, Johnson L, et al. In vitro Candida albicans vaginitis. J Med Chem. 2016;59:
activities of LTX-109, a synthetic antimicrobial peptide, 1791–1799.
against methicillin-resistant, vancomycin-intermediate, [88] Munoz A, Lopez-Garcia B, Perez-Paya E, et al.
vancomycin-resistant, daptomycin-nonsusceptible, and Antimicrobial properties of derivatives of the cationic
linezolid-nonsusceptible Staphylococcus aureus. tryptophan-rich hexapeptide PAF26. Biochem
Antimicrob Agents Chemother. 2012;56:4478–4482. Biophys Res Comm. 2007;354:172–177.
[74] Umerska A, Cassisa V, Bastiat G, et al. Synergistic [89] Gonzalez R, Mendive-Tapia L, Pastrian MB, et al.
interactions between antimicrobial peptides derived Enhanced antimicrobial activity of a peptide derived
from plectasin and lipid nanocapsules containing from human lysozyme by arylation of its tryptophan
monolaurin as a cosurfactant against Staphylococcus residues. J Pept Sci. 2016;22:123–128.
aureus. Int J Nanomedicine. 2017;12:5687–5699. [90] He R, Di Bonaventura I, Visini R, et al. Design, crystal
[75] Fritsche TR, Rhomberg PR, Sader HS, et al. structure and atomic force microscopy study of thio-
Antimicrobial activity of omiganan pentahydrochlor- ether ligated d,l-cyclic antimicrobial peptides against
ide tested against contemporary bacterial pathogens multidrug resistant Pseudomonas aeruginosa. Chem
commonly responsible for catheter-associated infec- Sci. 2017;8:7464–7475.
tions. J Antimicrob Chemother. 2008;61:1092–1098. [91] Li X, Li Y, Han H, et al. Solution structures of human
[76] Ghobrial OG, Derendorf H, Hillman JD. LL-37 fragments and NMR-based identification of a
Pharmacodynamic activity of the lantibiotic MU1140. minimal membrane-targeting antimicrobial and anti-
Int J Antimicrob Agents. 2009;33:70–74. cancer region. J Am Chem Soc. 2006;128:5776–5785.
[77] Kaplan CW, Sim JH, Shah KR, et al. Selective mem- [92] Townsley LE, Tucker WA, Sham S, et al. Structures of
brane disruption: mode of action of C16G2, a gramicidins A, B, and C incorporated into sodium
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 19

dodecyl sulfate micelles. Biochemistry. 2001;40: [108] Gautier R, Douguet D, Antonny B, et al. HELIQUEST: a
11676–11686. web server to screen sequences with specific alpha-
[93] Becucci L, Valensin D, Innocenti M, et al. Dermcidin, helical properties. Bioinformatics. 2008;24:2101–2102.
an anionic antimicrobial peptide: influence of lipid [109] Blazyk J, Wiegand R, Klein J, et al. A novel linear
charge, pH and Zn2þ on its interaction with a bio- amphipathic beta-sheet cationic antimicrobial pep-
mimetic membrane. Soft Matter. 2014;10:616–626. tide with enhanced selectivity for bacterial lipids. J
[94] Wang G. Database-guided discovery of potent pepti- Biol Chem. 2001;276:27899–27906.
des to combat HIV-1 or superbugs. Pharmaceuticals [110] Omardien S, Drijfhout JW, Vaz FM, et al. Bactericidal
(Basel). 2013;6:728–758. activity of amphipathic cationic antimicrobial pepti-
[95] Mulder KC, Lima LA, Miranda VJ, et al. Current scen- des involves altering the membrane fluidity when
ario of peptide-based drugs: the key roles of cationic interacting with the phospholipid bilayer. Biochim
antitumor and antiviral peptides. Front Microbiol.
Biophys Acta Biomembr. 2018;1860:2404–2415.
2013;4:321.
[111] Dziuba B, Dziuba M. New milk protein-derived pepti-
[96] Ueno S, Minaba M, Nishiuchi Y, et al. Generation of
des with potential antimicrobial activity: an approach
novel cationic antimicrobial peptides from natural
based on bioinformatic studies. Int J Mol Sci. 2014;
non-antimicrobial sequences by acid-amide substitu-
15:14531–14545.
tion. Ann Clin Microbiol Antimicrob. 2011;10:11.
[112] Wackett LP. Antimicrobial peptides: an annotated
[97] da Silva AV, De Souza BM, Dos Santos Cabrera MP,
et al. The effects of the C-terminal amidation of mas- selection of world wide web sites relevant to the
toparans on their biological actions and interactions topics in microbial biotechnology. Microb Biotechnol.
with membrane-mimetic systems. Biochim Biophys 2019;12:180–181.
Acta. 2014;1838:2357–2368. [113] Waghu FH, Barai RS, Gurung P, et al. CAMPR3: a
[98] Bea Rde L, Petraglia AF, Johnson LE. Synthesis, anti- database on sequences, structures and signatures of
microbial activity and toxicity of analogs of the scor- antimicrobial peptides. Nucleic Acids Res. 2016;44:
pion venom BmKn peptides. Toxicon. 2015;101: D1094–D1097.
79–84. [114] Wang G, Li X, Wang Z. APD3: the antimicrobial pep-
[99] Jenssen H. Therapeutic approaches using host tide database as a tool for research and education.
defence peptides to tackle herpes virus infections. Nucleic Acids Res. 2016;44:D1087–D1093.
Viruses. 2009;1:939–964. [115] Porto WF, Pires AS, Franco OL. Computational tools
[100] Xiao H, Shao F, Wu M, et al. The application of anti- for exploring sequence databases as a resource for
microbial peptides as growth and health promoters antimicrobial peptides. Biotechnol Adv. 2017;35:
for swine. J Anim Sci Biotechnol. 2015;6:19. 337–349.
[101] Vermeer LS, Lan Y, Abbate V, et al. Conformational [116] Liu S, Fan L, Sun J, et al. Computational resources
flexibility determines selectivity and antibacterial, and tools for antimicrobial peptides. J Pept Sci. 2017;
antiplasmodial, and anticancer potency of cationic 23:4–12.
alpha-helical peptides. J Biol Chem. 2012;287: [117] Torres MDT, Sothiselvam S, Lu TK, et al. Peptide
34120–34133. design principles for antimicrobial applications. J Mol
[102] Wang G, Hanke ML, Mishra B, et al. Transformation Biol. 2019;pii: S0022-2836(18)31289-0.
of human cathelicidin LL-37 into selective, stable, [118] Mishra B, Wang G. Ab initio design of potent anti-
and potent antimicrobial compounds. ACS Chem MRSA peptides based on database filtering technol-
Biol. 2014;9:1997–2002. ogy. J Am Chem Soc. 2012;134:12426–12429.
[103] Hwang H, Hyun S, Kim Y, et al. Reduction of helical [119] Porto WF, Irazazabal L, Alves ESF, et al. In silico opti-
content by insertion of a disulfide bond leads to an mization of a guava antimicrobial peptide enables
antimicrobial peptide with decreased hemolytic
combinatorial exploration for peptide design. Nature
activity. ChemMedChem. 2013;8:59–62.
Comm. 2018;9:1490.
[104] Yeaman MR, Yount NY. Mechanisms of antimicrobial
[120] Chou S, Wang J, Shang L, et al. Short, symmetric-hel-
peptide action and resistance. Pharmacol Rev. 2003;
ical peptides have narrow-spectrum activity with low
55:27–55.
resistance potential and high selectivity. Biomaterials
[105] Son M, Lee Y, Hwang H, et al. Disruption of interac-
tions between hydrophobic residues on nonpolar Sci. 2019;7:2349–2409.
[121] Andreu D, Torrent M. Prediction of bioactive pepti-
faces is a key determinant in decreasing hemolysis
and increasing antimicrobial activities of alpha-helical des using artificial neural networks. Methods Mol
amphipathic peptides. ChemMedChem. 2013;8: Biol. 2015;1260:101–118.
1638–1642. [122] Madanchi H, Akbari S, Shabani AA, et al. Alignment-
[106] Chang TW, Wei SY, Wang SH, et al. Hydrophobic resi- based design and synthesis of new antimicrobial
dues are critical for the helix-forming, hemolytic and Aurein-derived peptides with improved activity
bactericidal activities of amphipathic antimicrobial against Gram-negative bacteria and evaluation of
peptide TP4. PLoS One. 2017;12:e0186442. their toxicity on human cells. Drug Dev Res. 2019;80:
[107] Yang G, Wang J, Lu S, et al. Short lipopeptides spe- 162–170.
cifically inhibit the growth of Propionibacterium acnes [123] Pillong M, Hiss JA, Schneider P, et al. Rational design
with antibacterial and anti-inflammatory dual action. of membrane-pore-forming peptides. Small. 2017;13.
Br J Pharmacol. 2019;176:1603–1618. DOI:10.1002/smll.201701316.
20 A. BARRETO-SANTAMARIA ET AL.

[124] Rondon-Villarreal P, Pinzon-Reyes E. Computer aided [138] Zwaal RF, Comfurius P, Bevers EM. Surface exposure
design of non-toxic antibacterial peptides. Curr Top of phosphatidylserine in pathological cells. Cell Mol
Med Chem. 2018;18:1044–1052. Life Sci. 2005;62:971–988.
[125] Yan J, Wang K, Dang W, et al. Two hits are better [139] Whitman WB. The modern concept of the procary-
than one: membrane-active and DNA binding-related ote. J Bacteriol. 2009;191:2000–2005.
double-action mechanism of NK-18, a novel anti- [140] Hobot J, Bacterial ultrastructure. Chapter 2. In: Tang
microbial peptide derived from mammalian NK-lysin. Y-W, Sussman M, Liu D, et al., eds. Molecular medical
Antimicrob Agents Chemother. 2013;57:220–228. microbiology. 2nd ed. Cardiff, UK: Elsevier Science;
[126] Hao G, Shi YH, Tang YL, et al. The intracellular mech- 2014:7–32.
anism of action on Escherichia coli of BF2-A/C, two [141] Epand RF, Savage PB, Epand RM. Bacterial lipid com-
analogues of the antimicrobial peptide Buforin 2. J position and the antimicrobial efficacy of cationic
Microbiol. 2013;51:200–206. steroid compounds (Ceragenins). Biochim Biophys
[127] Dangel A, Ackermann N, Abdel-Hadi O, et al. A de Acta. 2007;1768:2500–2509.
novo-designed antimicrobial peptide with activity [142] Ahmed A, Rushworth JV, Hirst NA, et al. Biosensors
against multiresistant Staphylococcus aureus acting for whole-cell bacterial detection. Clin Microbiol Rev.
on RsbW kinase. FASEB J. 2013;27:4476–4488. 2014;27:631–646.
[128] Malanovic N, Leber R, Schmuck M, et al. [143] Delcour AH. Outer membrane permeability and anti-
Phospholipid-driven differences determine the action biotic resistance. Biochim Biophys Acta. 2009;1794:
of the synthetic antimicrobial peptide OP-145 on 808–816.
Gram-positive bacterial and mammalian membrane [144] Miller SI. Antibiotic resistance and regulation of the
model systems. Biochim Biophys Acta. 2015;1848: gram-negative bacterial outer membrane barrier by
2437–2447. host innate immune molecules. MBio. 2016;7:pii:
[129] Sun S, Zhao G, Huang Y, et al. Specificity and mech- e01541-16.
anism of action of alpha-helical membrane-active [145] Piers KL, Brown MH, Hancock RE. Improvement of
peptides interacting with model and biological mem- outer membrane-permeabilizing and lipopolysacchar-
branes by single-molecule force spectroscopy. Sci ide-binding activities of an antimicrobial cationic
Rep. 2016;6:29145. peptide by C-terminal modification. Antimicrob
[130] Arouri A, Dathe M, Blume A. Peptide induced demix- Agents Chemother. 1994;38:2311–2316.
ing in PG/PE lipid mixtures: a mechanism for the [146] Zawadzka K, Bernat P, Felczak A, et al. Antibacterial
specificity of antimicrobial peptides towards bacterial activity of high concentrations of carvedilol against
membranes? Biochim Biophys Acta. 2009;1788: Gram-positive and Gram-negative bacteria. Int J
650–659. Antimicrob Agents. 2018;51:458–467.
[131] Patino-Marquez IA, Manrique-Moreno M, Patino- [147] Hasper HE, Kramer NE, Smith JL, et al. An alternative
Gonzalez E, et al. Effect of antimicrobial peptides bactericidal mechanism of action for lantibiotic pep-
from Galleria mellonella on molecular models of tides that target lipid II. Sci. 2006;313:1636–1637.
Leishmania membrane. Thermotropic and fluores- [148] Percy MG, Grundling A. Lipoteichoic acid synthesis
cence anisotropy study. J Antibiot (Tokyo). 2018;71: and function in gram-positive bacteria. Annu Rev
642–652. Microbiol. 2014;68:81–100.
[132] Epand RF, Schmitt MA, Gellman SH, et al. Role [149] Saar-Dover R, Bitler A, Nezer R, et al. D-alanylation of
of membrane lipids in the mechanism of lipoteichoic acids confers resistance to cationic pepti-
bacterial species selective toxicity by two des in group B streptococcus by increasing the cell
alpha/beta-antimicrobial peptides. Biochim Biophys wall density. PLoS Pathog. 2012;8:e1002891.
Acta. 2006;1758:1343–1350. [150] Malanovic N, Lohner K. Gram-positive bacterial cell
[133] Dickson RC. Sphingolipid functions in Saccharomyces envelopes: the impact on the activity of antimicro-
cerevisiae: comparison to mammals. Annu Rev bial peptides. Biochim Biophys Acta. 2016;1858:
Biochem. 1998;67:27–48. 936–946.
[134] Daleke DL. Regulation of phospholipid asymmetry in [151] Ernst CM, Peschel A. Broad-spectrum antimicrobial
the erythrocyte membrane. Curr Opin Hematol. 2008; peptide resistance by MprF-mediated aminoacylation
15:191–195. and flipping of phospholipids. Mol Microbiol. 2011;
[135] van der Rest ME, Kamminga AH, Nakano A, et al. The 80:290–299.
plasma membrane of Saccharomyces cerevisiae: struc- [152] Zhang T, Muraih JK, Tishbi N, et al. Cardiolipin pre-
ture, function, and biogenesis. Microbiol Rev. 1995; vents membrane translocation and permeabilization
59:304–322. by daptomycin. J Biol Chem. 2014;289:11584–11591.
[136] Ebenhan T, Gheysens O, Kruger HG, et al. [153] Maria-Neto S, de Almeida KC, Macedo ML, et al.
Antimicrobial peptides: their role as infection-select- Understanding bacterial resistance to antimicrobial
ive tracers for molecular imaging. Biomed Res Int. peptides: from the surface to deep inside. Biochim
2014;2014:867381. Biophys Acta. 2015;1848:3078–3088.
[137] Lee DK, Bhunia A, Kotler SA, et al. Detergent-type [154] Rautenbach M, Troskie AM, Vosloo JA. Antifungal
membrane fragmentation by MSI-78, MSI-367, MSI- peptides: to be or not to be membrane active.
594, and MSI-843 antimicrobial peptides and inhib- Biochimie. 2016;130:132–145.
ition by cholesterol: a solid-state nuclear magnetic [155] Wang K, Yan J, Dang W, et al. Dual antifungal prop-
resonance study. Biochemistry. 2015;54:1897–1907. erties of cationic antimicrobial peptides polybia-MPI:
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 21

membrane integrity disruption and inhibition of bio- [172] Lorizate M, Krausslich HG. Role of lipids in virus repli-
film formation. Peptides. 2014;56:22–29. cation. Cold Spring Harbor Perspect Biol. 2011;3:
[156] Henderson CM, Block DE. Examining the role of a004820.
membrane lipid composition in determining the [173] Buck CB, Day PM, Thompson CD, et al. Human
ethanol tolerance of Saccharomyces cerevisiae. Appl alpha-defensins block papillomavirus infection. Proc
Environ Microbiol. 2014;80:2966–2972. Nat Acad Sciences USA. 2006;103:1516–1521.
[157] Feigin AM, Schagina LV, Takemoto JY, et al. The [174] Chang TL, Vargas J, Jr., DelPortillo A, et al. Dual role
effect of sterols on the sensitivity of membranes to of alpha-defensin-1 in anti-HIV-1 innate immunity. J
the channel-forming antifungal antibiotic, syringomy- Clin Invest. 2005;115:765–773.
cin E. Biochim Biophys Acta. 1997;1324:102–110. [175] Jacobs T, Bruhn H, Gaworski I, et al. NK-lysin and its
[158] Czub J, Baginski M. Comparative molecular dynamics shortened analog NK-2 exhibit potent activities
study of lipid membranes containing cholesterol and against Trypanosoma cruzi. Antimicrob Agents
ergosterol. Biophys J. 2006;90:2368–2382. Chemother. 2003;47:607–613.
[159] Smits GJ, Kapteyn JC, van den Ende H, et al. Cell [176] Gelhaus C, Jacobs T, Andra J, et al. The antimicrobial
wall dynamics in yeast. Curr Opin Microbiol. 1999;2: peptide NK-2, the core region of mammalian NK-
348–352. lysin, kills intraerythrocytic Plasmodium falciparum.
[160] Endo M, Takesako K, Kato I, et al. Fungicidal action Antimicrob Agents Chemother. 2008;52:1713–1720.
of aureobasidin A, a cyclic depsipeptide antifungal [177] Hernandez C, Mor A, Dagger F, et al. Functional and
antibiotic, against Saccharomyces cerevisiae. structural damage in Leishmania mexicana exposed
Antimicrob Agents Chemother. 1997;41:672–676. to the cationic peptide dermaseptin. Eur J Cell Biol.
[161] Levitz SM, Selsted ME, Ganz T, et al. In vitro killing of 1992;59:414–424.
spores and hyphae of Aspergillus fumigatus and [178] Mor A. Multifunctional host defense peptides: anti-
Rhizopus oryzae by rabbit neutrophil cationic pepti- parasitic activities. FEBS J. 2009;276:6474–6482.
des and bronchoalveolar macrophages. J Infect Dis. [179] Sherman IW, Prudhomme J, Tait JF. Altered mem-
1986;154:483–489. brane phospholipid asymmetry in Plasmodium falcip-
[162] Osumi M. The ultrastructure of yeast: cell wall struc- arum-infected erythrocytes. Parasitol Today (Regul.
ture and formation. Micron. 1998;29:207–233. Ed.). 1997;13:242–243.
[163] Ruiz-Herrera J, Mormeneo S, Vanaclocha P, et al. [180] Eda S, Sherman IW. Cytoadherence of malaria-
Structural organization of the components of the cell infected red blood cells involves exposure of phos-
wall from Candida albicans. Microbiology. 1994;140: phatidylserine. Cell Physiol Biochem. 2002;12:
1513–1523. 373–384.
[164] van der Weerden NL, Hancock RE, Anderson MA. [181] Henriques C, Atella GC, Bonilha VL, et al. Biochemical
Permeabilization of fungal hyphae by the plant analysis of proteins and lipids found in parasitopho-
defensin NaD1 occurs through a cell wall-dependent rous vacuoles containing Leishmania amazonensis.
process. J Biol Chem. 2010;285:37513–37520. Parasitol Res. 2003;89:123–133.
[165] Yeaman MR, Buttner S, Thevissen K. Regulated cell [182] Krugliak M, Feder R, Zolotarev VY, et al. Antimalarial
death as a therapeutic target for novel antifungal activities of dermaseptin S4 derivatives. Antimicrob
peptides and biologics. Oxid Med Cell Longev. 2018; Agents Chemother. 2000;44:2442–2451.
2018:1. [183] Marechal E, Riou M, Kerboeuf D, et al. Membrane lip-
[166] Shartouny JR, Jacob J. Mining the tree of life: host idomics for the discovery of new antiparasitic drug
defense peptides as antiviral therapeutics. Semin Cell targets. Trends Parasitol. 2011;27:496–504.
Dev Biol. 2019;88:147–155. [184] Merriman JA, Nemeth KA, Schlievert PM. Novel anti-
[167] Falco A, Ortega-Villaizan M, Chico V, et al. microbial peptides that inhibit gram positive bacter-
Antimicrobial peptides as model molecules for the ial exotoxin synthesis. PLoS One. 2014;9:e95661.
development of novel antiviral agents in aquacul- [185] Sarma P, Mahendiratta S, Prakash A, et al. Specifically
ture. Mini Rev Med Chem. 2009;9:1159–1164. targeted antimicrobial peptides: a new and promis-
[168] Hsieh IN, Hartshorn KL. The role of antimicrobial pep- ing avenue in selective antimicrobial therapy. Indian
tides in influenza virus infection and their potential J Pharmacol. 2018;50:1–3.
as antiviral and immunomodulatory therapy. [186] Huo L, Huang X, Ling J, et al. Selective activities of
Pharmaceuticals. 2016;9:pii: E53. STAMPs against Streptococcus mutans. Exp Ther Med.
[169] Owen SM, Rudolph DL, Wang W, et al. RC-101, a ret- 2018;15:1886–1893.
rocyclin-1 analogue with enhanced activity against [187] He J, Anderson MH, Shi W, et al. Design and activity
primary HIV type 1 isolates. AIDS Res Hum of a ’dual-targeted’ antimicrobial peptide. Int J
Retroviruses. 2004;20:1157–1165. Antimicrob Agents. 2009;33:532–537.
[170] Tripathi S, Wang G, White M, et al. Antiviral activity [188] de la Fuente-Nunez C, Torres MD, Mojica FJ, et al.
of the human cathelicidin, ll-37, and derived pepti- Next-generation precision antimicrobials: towards
des on seasonal and pandemic influenza A viruses. personalized treatment of infectious diseases. Curr
PLoS One. 2015;10:e0124706. Opin Microbiol. 2017;37:95–102.
[171] Aloia RC, Tian H, Jensen FC. Lipid composition and [189] Torres MDT, Andrade GP, Sato RH, et al. Natural and
fluidity of the human immunodeficiency virus enve- redesigned wasp venom peptides with selective anti-
lope and host cell plasma membranes. Proc Nat tumoral activity. Beilstein J Org Chem. 2018;14:
Acad Sci USA. 1993;90:5181–5185. 1693–1703.
22 A. BARRETO-SANTAMARIA ET AL.

[190] Rasamiravaka T, Labtani Q, Duez P, et al. The forma- [206] Ageitos JM, Sanchez-Perez A, Calo-Mata P, et al.
tion of biofilms by Pseudomonas aeruginosa: a Antimicrobial peptides (AMPs): ancient compounds
review of the natural and synthetic compounds that represent novel weapons in the fight against
interfering with control mechanisms. Biomed Res Int. bacteria. Biochem Pharmacol. 2017;133:117–138.
2015;2015:1. [207] Food and Drug Administration. Antibacterial Drug
[191] Pletzer D, Coleman SR, Hancock R. Anti-biofilm pepti- Development Task Force. 2013. Availabe from:
des as a new weapon in antimicrobial warfare. Curr https://www.fda.gov/drugs/development-resources/
Opin Microbiol. 2016;33:35–40. antibacterial-drug-development-task-force
[192] Ribeiro SM, de la Fuente-Nunez C, Baquir B, et al. [208] COMBACTE. Combatting antimicrobial resistance.
Antibiofilm peptides increase the susceptibility of Available from: https://www.combacte.com/about/
carbapenemase-producing Klebsiella pneumoniae [209] World Health Organization. Resolution WHA68.7.
clinical isolates to beta-lactam antibiotics. Antimicrob Global action plan on antimicrobial resistance. In:
Agents Chemother. 2015;59:3906–3912. Sixty-eighth World Health Assembly. Geneva. 2015.
[193] Yasir M, Willcox MDP, Dutta D. Action of antimicro- Available from: https://www.who.int/antimicrobial-
bial peptides against bacterial biofilms. Materials. resistance/global-action-plan/en/
2018;11:pii: E2468. [210] Goldstein-Research. Anti microbial peptides market
[194] de la Fuente-Nunez C, Cardoso MH, de Souza CE, outlook 2024: Global opportunity and demand ana-
et al. Synthetic antibiofilm peptides. Biochim Biophys lysis, market forecast, 2016-2024. 2017. p. 236.
Acta. 2016;1858:1061–1069. Available from: https://www.goldsteinresearch.com/
[195] Bevers EM, Comfurius P, Zwaal RF. Regulatory mech- report/anti-microbial-peptides-market-outlook-2024-
anisms in maintenance and modulation of trans- global-opportunity-and-demand-analysis-market-fore-
membrane lipid asymmetry: pathophysiological cast-2016-2024
implications. Lupus. 1996;5:480–487. [211] Muller PY, Milton MN. The determination and inter-
[196] Baxter AA, Richter V, Lay FT, et al. The tomato defen-
pretation of the therapeutic index in drug develop-
sin TPP3 binds phosphatidylinositol (4,5)-bisphosphate
ment. Nat Rev Drug Discov. 2012;11:751–761.
via a conserved dimeric cationic grip conformation to
[212] Vlieghe P, Lisowski V, Martinez J, et al. Synthetic
mediate cell lysis. Mol Cell Biol. 2015;35:1964–1978.
therapeutic peptides: science and market. Drug
[197] Tsai TL, Li AC, Chen YC, et al. Antimicrobial peptide
Discov Today. 2010;15:40–56.
m2163 or m2386 identified from Lactobacillus casei
[213] Santos GB, Ganesan A, Emery FS. Oral administration
ATCC 334 can trigger apoptosis in the human colo-
of peptide-based drugs: beyond Lipinski’s rule.
rectal cancer cell line SW480. Tumor Biol. 2015;36:
ChemMedChem. 2016;11:2245–2251.
3775–3789.
[214] Svenson J, Vergote V, Karstad R, et al. Metabolic fate
[198] O’Connor S, Szwej E, Nikodinovic-Runic J, et al. The
of lactoferricin-based antimicrobial peptides: effect of
anti-cancer activity of a cationic anti-microbial pep-
truncation and incorporation of amino acid analogs
tide derived from monomers of polyhydroxyalka-
on the in vitro metabolic stability. J Pharmacol Exp
noate. Biomaterials. 2013;34:2710–2718.
[199] Felicio MR, Silva ON, Goncalves S, et al. Peptides Therap. 2010;332:1032–1039.
with dual antimicrobial and anticancer activities. [215] Falanga A, Nigro E, De Biasi MG, et al. Cyclic pepti-
Front Chem. 2017;5:5. des as novel therapeutic microbicides: engineering
[200] Into T, Inomata M, Shibata K, et al. Effect of the anti- of human defensin mimetics. Molecules. 2017;22:pii:
microbial peptide LL-37 on Toll-like receptors 2-, 3- E1217.
and 4-triggered expression of IL-6, IL-8 and CXCL10 [216] Liu B, Zhang W, Gou S, et al. Intramolecular cycliza-
in human gingival fibroblasts. Cell Immunol. 2010; tion of the antimicrobial peptide Polybia-MPI with
264:104–109. triazole stapling: influence on stability and bioactiv-
[201] Drago-Serrano ME, Campos-Rodriguez R, Carrero JC, ity. J Pept Sci. 2017;23:824–832.
et al. Lactoferrin and peptide-derivatives: antimicro- [217] Silva AF, Torres MT, Silva LS, et al. Angiotensin II-
bial agents with potential use in nonspecific immun- derived constrained peptides with antiplasmodial
ity modulation. Curr Pharm Des. 2018;24:1067–1078. activity and suppressed vasoconstriction. Sci Rep.
[202] Turner MD, Nedjai B, Hurst T, et al. Cytokines and 2017;7:14326.
chemokines: at the crossroads of cell signalling and [218] Migon D, Neubauer D, Kamysz W. Hydrocarbon
inflammatory disease. Biochim Biophys Acta. 2014; stapled antimicrobial peptides. Protein J. 2018;37:
1843:2563–2582. 2–12.
[203] Lee SH, Jun HK, Lee HR, et al. Antibacterial and lipo- [219] Jenner ZB, Crittenden CM, Gonzalez M, et al.
polysaccharide (LPS)-neutralising activity of human Hydrocarbon-stapled lipopeptides exhibit selective
cationic antimicrobial peptides against periodonto- antimicrobial activity. Biopolymers. 2017;108. doi:10.
pathogens. Int J Antimicrob Agents. 2010;35:138–145. 1002/smll.201701316.
[204] Lee EY, Lee MW, Wong GCL. Modulation of toll-like [220] Lau YH, de Andrade P, Wu Y, et al. Peptide stapling
receptor signaling by antimicrobial peptides. Semin techniques based on different macrocyclisation
Cell Dev Biol. 2019;88:173–184. chemistries. Chem Soc Rev. 2015;44:91–102.
[205] Sun Y, Shang D. Inhibitory effects of antimicrobial [221] Czyzewski AM, Jenssen H, Fjell CD, et al. In vivo, in
peptides on lipopolysaccharide-induced inflamma- vitro, and in silico characterization of peptoids as
tion. Mediators Inflamm. 2015;2015:1. antimicrobial agents. PLoS One. 2016;11:e0135961.
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 23

[222] Bhosle GS, Nawale L, Yeware AM, et al. Antibacterial [230] Seo M-D, Won H-S, Kim J-H, et al. Antimicrobial pep-
and anti-TB tat-peptidomimetics with improved effi- tides for therapeutic applications: a review.
cacy and half-life. Eur J Med Chem. 2018;152: Molecules. 2012;17:12276–12286.
358–369. [231] Fang B, Guo HY, Zhang M, et al. The six amino acid
[223] Arias M, Piga KB, Hyndman ME, et al. Improving the antimicrobial peptide bLFcin6 penetrates cells and
activity of Trp-rich antimicrobial peptides by Arg/Lys delivers siRNA. FEBS J. 2013;280:1007–1017.
substitutions and changing the length of cationic [232] Wang G. Structures of human host defense
residues. Biomolecules. 2018;8:19. cathelicidin LL-37 and its smallest antimicrobial pep-
[224] Chu HL, Yu HY, Yip BS, et al. Boosting salt resistance tide KR-12 in lipid micelles. J Biol Chem. 2008;283:
of short antimicrobial peptides. Antimicrob Agents 32637–32643.
Chemother. 2013;57:4050–4052. [233] Bommarius B, Jenssen H, Elliott M, et al. Cost-effect-
[225] Xie D, Yao C, Wang L, et al. An albumin-conjugated ive expression and purification of antimicrobial and
peptide exhibits potent anti-HIV activity and long in host defense peptides in Escherichia coli. Peptides.
vivo half-life. Antimicrob Agents Chemother. 2010;54: 2010;31:1957–1965.
191–196. [234] Cao J, de la Fuente-Nunez C, Ou RW, et al. Yeast-
[226] Sivertsen A, Isaksson J, Leiros HK, et al. Synthetic cat- based synthetic biology platform for antimicrobial
ionic antimicrobial peptides bind with their hydro- peptide production. ACS Synth Biol. 2018;7:896–902.
phobic parts to drug site II of human serum [235] Holaskova E, Galuszka P, Micuchova A, et al.
albumin. BMC Struct Biol. 2014;14:4. Molecular farming in barley: development of a novel
[227] Pei J, Feng Z, Ren T, et al. Purification, characteriza- production platform to produce human antimicrobial
tion and application of a novel antimicrobial peptide peptide LL-37. Biotechnol J. 2018;13:e1700628.
from Andrias davidianus blood. Lett Appl Microbiol. [236] Merrifield B. Solid phase synthesis. Science. 1986;232:
2018;66:38–43. 341–347.
[228] Fosgerau K, Hoffmann T. Peptide therapeutics: cur- [237] Behrendt R, White P, Offer J. Advances in Fmoc
rent status and future directions. Drug Discov Today. solid-phase peptide synthesis. J Pept Sci. 2016;22:
2015;20:122–128. 4–27.
[229] Carmona-Ribeiro AM, de Melo Carrasco LD. Novel [238] Made V, Els-Heindl S, Beck-Sickinger AG. Automated
formulations for antimicrobial peptides. Int J Mol Sci. solid-phase peptide synthesis to obtain therapeutic
2014;15:18040–18083. peptides. Beilstein J Org Chem. 2014;10:1197–1212.

You might also like