Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Microbial Pathogenesis 179 (2023) 106108

Contents lists available at ScienceDirect

Microbial Pathogenesis
journal homepage: www.elsevier.com/locate/micpath

A historical, economic, and technical-scientific approach to the current


crisis in the development of antibacterial drugs: Promising role of
antibacterial peptides in this scenario
Yeimer A.S. Guevara a, Maria H.C. Santos b, Francisco I.R. Gomes b, Sheheryar b, Felipe
P. Mesquita a, b, Pedro F.N. Souza a, b, *
a
Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil
b
Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil

A R T I C L E I N F O A B S T R A C T

Keywords: The emergence of antibiotic resistance (AMR) is a global public health problem. According to estimates, drug-
Antibacterial resistance resistant bacteria infect 2 million patients and perish 23,000 annually. To overcome this problem, antimicro­
Antibiotic crisis bial peptides became a potential solution based on a new mechanism of action against bacteria. This article
Multidrug-resistant pathogens
addresses the phenomenon of antibacterial resistance in most of its nuances, responding to historical, technical-
Synthetic antibacterial peptides
scientific, and economic aspects. Likewise, it explores new therapeutic approaches to combat multi-resistant
pathogens, specifically concerning antibacterial peptides, as a potential therapeutic tool to mitigate the cur­
rent crisis of antibacterial drugs. It is expected that, with technological advances, especially with the advent and
adoption of artificial intelligence, there will be an increase in diversified synthetic peptide production, which can
face the challenges that we have in terms of antibacterial drugs.

1. Introduction antibiotics as growth promoters in feed creates selective pressure that


leads to the emergence of resistant bacteria in cattle that can subse­
According to the World Health Organization, antibiotic resistance is quently colonize or infect humans [9].
a global public health problem [55]. According to estimates in the One of the main concerns of the WHO is multi-resistant bacteria due
United States, drug-resistant bacteria infect 2 million patients and perish to their rapid spread and the difficulty of their treatment [13]. The so­
23,000 of them annually [26]. It is estimated that, if no action is taken, lution to the problem involves, in part, infection prevention and AMR
by 2050, antimicrobial resistance (AMR) could result in 10 million monitoring, the promotion of rational antimicrobial therapy, and the
deaths per year worldwide and accumulate at least US$100 trillion in development of new antimicrobials [17,42]. Unfortunately, in recent
hospital costs and lost productivity [17]. decades, the discovery and development of new antibiotics have slowed
AMR can be innate or acquired. Thus, antibiotic administration drastically to such an extent that no new class has been discovered since
creates an environment where resistance strains have a selective the 1980s, only those obtained from the optimization or combination of
advantage over those not [2,37]. Mechanisms involved in AMR include already known compounds being marketed [12,30,39]. According to
the production of inactivating enzymes, drug efflux and drug entry in­ Spellberg et al. [43], there are 3 leading reasons for discovering new
hibition, target modification, metabolic pathway alteration and chro­ antibiotics: first, there are scientific challenges for the discovery and
mosomal or plasmid-mediated removals [42,48]. development of new agents; second, compared to other classes of drugs,
The unrestricted/excessive misuse in humans and animals acceler­ antibiotics are economically unattractive for investment in research and
ated the natural process of evolution that led to AMR, despite increasing development (R&D) and third, to the difficulties imposed by the Food
pressure on the animal production sector to limit or completely cease the and Drug Administration (FDA) for conducting clinical trials with new
use of antimicrobials antibiotics [6,42]. Additionally, the use of antibiotics [43]. Thus, while nearly 4000 immune-oncology agents are

* Corresponding author. Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará,
60020-181, Brazil.
E-mail addresses: pedrofilhobio@gmail.com, pedrofilhobio@ufc.br (P.F.N. Souza).

https://doi.org/10.1016/j.micpath.2023.106108
Received 10 March 2023; Received in revised form 4 April 2023; Accepted 5 April 2023
Available online 10 April 2023
0882-4010/© 2023 Elsevier Ltd. All rights reserved.
Y.A.S. Guevara et al. Microbial Pathogenesis 179 (2023) 106108

developing, only about 30–40 new antibacterial compounds are the 1960s–1970s, its dissemination in the hospital environment made it
currently in the clinical trial development phases. Less than 25% of the urgent to look for new antibiotic compounds [15].
current drugs in the clinical development pipeline represent a new class The clinical introduction of a variety of third-generation cephalo­
or act through a novel mechanism and are not active against bacteria sporins characterized the 1980s. These agents (mainly cefotaxime, cef­
from the ESKAPE group (Enterococcus, Staphylococcus aureus, Klebsiella tizoxime, ceftriaxone, and ceftazidime) had activity against Enterobacter
species, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter), spp, Citrobacter freundii, and, in the case of ceftazidime, P. aeruginosa,
critically threatened pathogens according to the WHO [27,28]. which were characteristically resistant to β other-lactam strains avail­
Classically, most antibiotics were produced by screening for soil able. The widespread use of these agents was associated with the
microorganisms (Waxman Platform), but this limited resource of cul­ emergence and dissemination of Gram-negative bacilli, most commonly
turable bacteria was superseded by the 1960s and synthetic approaches Klebsiella pneumoniae, which were resistant to expressing β-lactamases of
to producing antibiotics. It has not been able to completely replace it, extended-spectrum (ESBLs) [37]. Linezolid and daptomycin were
except for teixobactin, a depsipeptide with excellent inhibitory potential introduced in the 2000s, and their resistance was reported in five years
against Gram-positive pathogens, including resistant strains [22,25]. [24].
The combination of widespread multidrug resistance, disenchantment Added to the problem of AMR against the antibiotics mentioned
with natural products as sources of new drugs, lack of success using above is the abrupt drop in the production of new molecules with
synthetic compounds, and changing economic and regulatory issues antibacterial properties (Fig. 2). The discovery and development of an­
conspire to move research investment away from the field of antibiotics. tibiotics in recent decades have decreased considerably; for example, 16,
The result is a growing crisis in antibiotic drug discovery that threatens 14, 10 and 7 new antibiotics were approved during 1983–1987,
modern medicine [58]. 1988–1992, 1993–1997 and 1998–2002, respectively, while only 5 and
Finding new antibiotics depends on the ability of scientists to explore 2 were approved during 2003–2007 and 2008–2012, respectively
a new chemical space through novel screening of libraries of diverse and (Fig. 2). This decline was due to the decrease in antibiotic research and
distinct compounds, targeted synthesis or modification of compounds development in major pharmaceutical companies; investment in
with improved physicochemical properties, phenotypic assays, and developing new antibiotics has been hampered by uncertain life cycles
other methods that are explicitly adapted to bacterial pathogens [1]. (associated with the development of antibiotic resistance) of new anti­
Thus, the problem should be approached in several parallel ways, biotics [5,11].
requiring the discovery of novel chemical scaffolds that evade the
resistance mechanisms that circulate in pathogens today and the
exploration of non-traditional anti-infective strategies [44,58]. In this
context, the investigation and arrival on the scene of peptides with
antibacterial properties are emerging as an invaluable tool to mitigate
the current crisis of antibacterial resistance.

2. Antibacterial resistance phenomenon and crisis in the


development of new antibiotic molecules

2.1. Timeline of antimicrobial resistance phenomena

After introducing sulfonamides and penicillin, their respective


resistance was reported in a few years, in mid-1945. Resistance to
tetracycline, streptomycin and chloramphenicol was found in the 1950s
(Fig. 1) [24,36].
Between the 1940’s and 1960’s, several antibiotics were discovered
through screenings of natural microbial products, most effective for
treating Gram-positive bacteria, such as β-lactams, aminoglycosides,
tetracyclines, and macrolides (Fig. 1). Methicillin was introduced in
1959 and methicillin-resistant S. aureus (MRSA) was identified in 1961
[15] (Fig. 1). With the advent of antimicrobial resistance increase from Fig. 2. Rate of production of new antibiotic molecules in the last decades.

Fig. 1. Timeline of antibacterial resistance phenomena.

2
Y.A.S. Guevara et al. Microbial Pathogenesis 179 (2023) 106108

2.2. Why did new antibiotic production fall to the detriment of other Gram-negative bacteria such as Pseudomonas aeruginosa and Acineto­
drugs? And what are the consequences of that fact? bacter baumannii, for which treatment options are already limited. This
has raised fears of a “post-antibiotic era,” as it has been estimated that
In recent decades, the discovery and development of new antibiotics between 5 and 20 new antibacterial drugs must enter clinical develop­
have slowed dramatically due to scientific barriers, regulatory chal­ ment to effectively address the current resistance problem [19].
lenges, and declining returns on investment, leading major pharma­ Because of the abandonment by pharmaceutical companies in the
ceutical companies to scale back or abandon drug procurement research production of new antibiotics and the growing phenomena of AMR, it is
antibiotics and focus on research areas with better financial prospects predicted to result in the death of 10 million people by 2050 and
[26,35,39,58] such is the case of AstraZeneca in 2016 and Novartis and cumulatively incur at least $100 trillion in hospital costs and produc­
Sanofi in 2018, which in certain places chose to invest in the research of tivity losses [8,17,52]. Another problem that arises from the misuse of
new drugs for chronic diseases such as diabetes and cancer that guar­ antibiotics is the selective pressure on bacteria, resulting in new modi­
antee better dividends [12,35]. fied strains that seem to reduce the chance that treatments are
Finding novel antibacterial molecules is the challenge, and since adequately effective in patients, for example, compromised by HIV im­
daptomycin and linezolid were launched in the 1980s, only five non- munity, cancer, surgical procedures, and diabetes; causing profound
derivative antibiotics have been approved by the FDA since 2000 consequences that lead to morbidity and mortality or clinical compli­
(Fig. 1) [12,30]. In this context, most current antibiotic development cations [20,45]. Although this is considered a natural process, incom­
programs primarily focus on modifying existing classes of drugs plete knowledge about such a catastrophe triggered the AMR rate [3,
discovered decades ago to circumvent bacterial resistance [39]. An 29]. In this context, the emergence of the ESKAPE pathogens group, for
important reason for the lack of innovation is the absence of a global example, is the leading cause of nosocomial-resistant infections world­
market that guarantees the sustainable income necessary to finance wide, many of which lack clinical treatments and represent currently
research, commercialization, and production at scale [4,46]. In this deadly bacteria with fast-growing multidrug-resistant properties [30,
context, the preclinical stages of antibiotic R&D are the riskiest, ac­ 53].
counting for about 45% of total costs. Although the academic sector can
promote it, the association with external financiers, such as the phar­ 2.3. Is the production of new classes of antibiotics economically viable?
maceutical industry [35]. Unfortunately, this only comes to fruition
after the nomination of extensively validated preclinical candidates, and One of the main problems when it comes to innovating in the pro­
often even these companies require phase I clinical data [28]. duction of new classes of antimicrobial drugs is the economic return. The
In line with the causes of the “market failure” of antibiotics challenge involved in the search for new molecules with the desired
mentioned above, bring up the scientific challenges for the discovery pharmacological characteristics, added to the well-intentioned regula­
and development of new agents and the regulations imposed by the FDA tory policies on using this type of pharmaceutical agent, among other
during the last two decades in clinical trials, which, in the opinion of factors, paints a bleak picture for investors in large companies. Since the
experts, have been taken to irrational extremes based solely on statistical new and effective antibiotics are left as a last resort to prevent the
considerations [43]. In this context, pharmaceutical companies have development of AMR, combined with low market prices conditioned in
stopped producing new antibiotics, a determination that has been part by those established for generics, it leads to unsustainable profits
reinforced by the availability of multiple low-cost generic antibiotics [4].
[38,57] and the adoption by control entities, from the principle of In clinical trials, antimicrobials are generally evaluated for non­
administration, aimed at reducing the general use of antibiotics in the inferiority, rather than superiority, compared to existing treatments, so
hospital setting [26] which ends up impacting the sales of new antibi­ they may not be cost-effective in traditional tests, where the new anti­
otics [3,43]. Also, if that new agent shows promising results but retains microbial is associated with additional cost and a comparable health
some toxicity, it may never see enough patients in a trial, and the result, disregarding the beneficial impact at the population level in
therapeutic index may not be well known [58]. terms of combating AMR [17]. On average, it takes $985 million and
Classically, most antibiotics were produced by screening soil mi­ 10–15 years to bring a new drug to market, and the value of a new
croorganisms Waxman platform. Still, this limited resource of cultivable antibiotic ranges from $486 million to $12 billion [26,30]. A 2017 es­
bacteria was outgrown in the 1960s, and synthetic approaches to pro­ timate puts the cost of developing an antibiotic at around $1.5 billion;
ducing antibiotics have failed to replace it [25,54]. The pharmaceutical meanwhile, industry analysts estimate that the average revenue gener­
industry responded by replacing the Waksman platform with a new ated from the sale of an antibiotic is approximately $46 million per year
chemical modification strategy that unfortunately did not produce the [35].
expected results, causing the need for antibiotics and, in particular, for These estimates are made using the so-called Net Present Value
natural products to decrease [16,57], such disenchantment with natural (NPV), a standard method companies use to prioritize investment stra­
products as sources of new drugs, lack of success in the use of synthetic tegies to calculate the value of a drug over a given period. Recent data
compounds exacerbate the growing crisis in antibiotic drug discovery put the NPV of an antibiotic at a negative $50 million, meaning losses to
that threatens modern medicine [50,58]. developers, to which additional costs per patient must be added due to
However, with advances in next-generation genome sequencing, the AMR phenomenon that ranges between US$5 and more than US
bioinformatics, and analytical chemistry, attempts are being made to $55,000 [38].
advance the renaissance of the antimicrobial industry to address the This financial reality for antibiotic research largely explains the fact
current crisis [57]. Thus, more than 1200 antimicrobial peptides (AMPs) that while there are nearly 4000 immuno-oncology agents in develop­
of various origins have been discovered in recent years. Unfortunately, ment, only about 30–40 new antibacterial compounds are currently in
none of them have been used as antibiotics [12]. Since the Infectious the development phases and clinical trials, and none of them is poten­
Diseases Society of America applied for approval of 10 new systemic tially active against bacteria of the ESKAPE group [28].
antibiotics by 2020 in the United States, there have been six approvals: Given this scenario, fiscal incentive policies are necessary from the
ceftaroline, dalbavancin, oritavancin, tedizolid, ceftolozane-tazoba private or state sector [38]. Multiple academic studies have estimated
ctam, and ceftazidime-avibactam [14,26], however, many of them that the incentives to use a cost-based approach range from $1 billion to
have not been commercialized in the main markets, due to the cost of $5 billion per new antibiotic [32].
registration and commercialization, which are unaffordable by low- and Boluarte & Schulze (2022) [4], using data collected from other
middle-income countries, which have the highest burden of AMR [5,31, antibiotic-specific models and refined with industry experts, calculates a
34]. The previous becomes especially concerning in the case of minimum incentive for the first antibiotic on the market between $2

3
Y.A.S. Guevara et al. Microbial Pathogenesis 179 (2023) 106108

billion and $3 billion, paid over five to ten years. Fortunately, there are In the same context, the drug’s ability to penetrate the outer mem­
signs that the industry is beginning to respond to the economic problem brane and its susceptibility to efflux mechanisms must be tracked
of antibiotics. The International Federation of Pharmaceutical Manu­ throughout the drug optimization process to develop new antibiotics
facturers and Associations announced the AMR Action Fund involving successfully [19]. Currently, work is being done to target new cellular
24 companies, which aims to bring two to four new antibiotics to market mechanisms with no known resistance and develop structure-based
by 2030 and has committed nearly $1 billion to support needed research “rules” to improve the permanence of the antibiotic inside the path­
to achieve this goal [35]. ogen, especially in Gram-negative bacteria [30,49].
Another way to reduce the limitations in the search for new antibi­
2.4. What are the technical difficulties in producing new antibacterial otics is to change the pharmaceutical industry’s main focus, concen­
compounds? trating on a relatively challenging task of hunting for broad-spectrum
novel antibacterial agents [57]. Expanding the scope of antibiotics to
When addressing the development issues of new antibiotic molecules include narrow-spectrum molecules, syncretic combinations, and viru­
from a merely technical approach, scientific barriers emerge as pro­ lence inhibitors offers an exciting prospect for new drug discovery, along
tagonists, making it necessary to change some of the paradigms that lead with identifying new antibiotic chemical scaffolds that evade resistance
to more interdisciplinary fundamental research. The document: A sci­ mechanisms and continue to develop, exploring non-traditional anti-­
entific road map for antibiotics discovery (2016) emphasizes the need infective strategies [58].
for specific training and experience and a mechanism to transfer
knowledge from industry to scientists to materialize the initiatives [1]. 2.5. What are the advances and perspectives in developing new
Improvements to FDA guidelines are also needed for the execution of antibacterial drugs?
controlled clinical trials with antibiotics that, given the inconvenience of
being executed involving a placebo, entail the need to use extensive Despite the limitations and drawbacks in recent years in obtaining
samples and, consequently, high costs, discouraging research [57]. new antibiotic drugs, several approaches have been proposed and
Microbial natural products, with their privileged properties of mi­ implemented to overcome such limitations. Various methods for culti­
crobial penetration and affinity for bacterial targets, may lead to a new vating organisms that had not been cultivated until then, the rational
era of antibiotic discovery [10,57]. A hallmark of natural antibiotic approach to drug design to improve already known molecules, and the
products is their chemical diversity and complexity. They are based on promotion of technology to obtain synthetic products; have resulted in
producing chemical scaffolds consisting of a peptide, polyketide, car­ obtaining molecules with acceptable pharmacological characteristics.
bohydrate, alkaloid, or terpene backbone that serve as the core structure These efforts have resulted in the achievement of drugs with proven
[47]. Despite these characteristics, pharmaceutical companies have antimicrobial activity. Thus, for example, from the metabolites of acti­
gradually set aside natural compounds for obtaining antibiotics, given nomycetes, walkmycin B, Validamycin and signamycin B were isolated,
that they may not be suitable as drugs once purified. In addition, the with new mechanisms of action such as histidine kinase inhibitors; since
chemical structure of the leading natural product requires rational drug design, more than 10 million new molecules have been
semi-synthesis, which increases the cost and detracts from the attrac­ synthesized. However, few active molecules have reached the market,
tiveness of natural products [57]. particularly anti-tuberculosis drugs [12] such as tubelactomicin A and
Exploring non-cultured bacteria could revive the Waxman Platform caprazamycins, and amycolamycin was identified as an antibiotic of
for discovering antibacterial of natural origin [25]. It is more interesting Staphylococcus aureus resistant to anti methicillin. These compounds’
to investigate, under rational design, synthetic compounds. The rational discovery encourages the search for more natural products active
design of drugs consists of the empirical synthesis of new molecules against antimicrobial-resistant species [18].
designed according to several rules (Lipinski rules) so that they are well Developing new antibiotics to treat life-threatening infections with
absorbed, non-toxic and active against a specific target [12]. Although bacteria resistant to existing antibiotics is highly prioritized [51]. In
Lipinski’s rule does not apply to antibiotics, most pharmaceutical 2016, AMR became only the fourth health issue after HIV, non­
companies have not developed separate synthetic chemical libraries communicable diseases, and Ebola to be discussed by the United Nations
with microbial physiology in mind, making it challenging to develop General Assembly [38]. Faced with the challenge of AMR, antibiotics
similar rules for synthetic compounds that are effective as antibiotics that use novel mechanisms to combat antimicrobial resistance are
[57]. needed.
A more fundamental problem particular to antibiotic discovery is the Teixobactin represents a new class of antibiotics with a unique
need for a better understanding of the scientific drive to build a suc­ chemical scaffolding and no detectable resistance, interfering with lipid
cessful library. Pharmaceutical companies have extensive collections of II, a peptidoglycan precursor [40]. The said molecule, obtained through
compounds that can be used as starting materials to find chemicals that the cultivation of hitherto uncultivated organisms, inhibits peptido­
are active against newly identified biological targets. Given the unique glycan synthesis with excellent activity against Gram-positive patho­
characteristics of antibiotics, the goal of this effort is not to find and gens, including resistant strains [25]. Thus, these new methods to
harvest new sources of natural products or build vast libraries of syn­ activate silent biosynthetic gene clusters using co-culture and to grow
thetic compounds but rather to generate and selectively modify chem­ ‘non-culturable’ bacteria have revitalized natural products as a source of
ical matter suited for the discovery of new antibiotics [1]. antibiotics [30]. On the other hand, in the race against the AMR phe­
One of the main challenges of the antibiotic industry is the AMR nomenon, it is required that scientists working on the discovery of an­
phenomenon. The last 20 years have seen a severe withdrawal of many timicrobials be allowed greater participation in decision-making
pharmaceutical companies from research and development of antimi­ networks within commercial funding sources so that their appreciation
crobial drugs targeting AMR strains [18]. A particular drawback is is hated and taken into consideration [28]. The following steps must
Gram-negative bacteria, most of which have built-in abilities to evade include joint action by the human and animal health sectors; addressing
antibiotics and develop resistance; for example, they have two mem­ antimicrobial resistance is complex. The formal tripartite of WHO, the
branes with orthogonal characteristics, which makes it difficult to Food and Agriculture Organization, and the World Organization for
design drugs that can penetrate both barriers, along with a variety of Animal Health must significantly identify and promote feasible strate­
efflux pumps that push drugs out of the cell, making it difficult to design gies to tackle AMR [21].
new antibiotics [1]. Thus, understanding the orientation and binding of New genomic technologies, including rapid and low-cost DNA
LPS (lipopolysaccharide) molecules to the outside of the outer mem­ sequencing and a growing understanding of the mechanical details of
brane could facilitate the development of cationic molecules to break it. natural product biosynthesis, along with innovation in chemical

4
Y.A.S. Guevara et al. Microbial Pathogenesis 179 (2023) 106108

synthesis and analytical methods, can be combined to expand bioactive The first of these efforts, the Infectious Disease Finance Facility, is a
chemical diversity in a fashion that is compatible with high-throughput financing mechanism proposed by the European Investment Bank and
technology and begin to overcome the innovation gap in antibiotic the European Commission and the second is a summary published by
discovery [47]. Another critical approach implemented in obtaining Review on Antimicrobial Resistance, which seeks to identify the charac­
new antimicrobial products is the modification of already known mol­ teristics of an optimal incentive strategy to promote the private devel­
ecules; thus, cefiderocol was developed from cephalosporins, and opment of more antimicrobial compounds. Both potentially offer
Terramycin was obtained from modifying the aminoglycoside sisomicin, valuable contributions to address the current crisis [17]. These and other
both approved by the FDA for urinary infections [12]. efforts that will consolidate future projects for the identification and
The known antibiotic activity of natural products has generally been development of antibiotics require the implementation of a legal
identified by phenotypic screening campaigns that determine activity framework for intellectual property that is agile and dynamic to reduce
against panels of test organisms in standardized assays. The basis for the time needed to negotiate property agreements between the invent­
bioactivity-guided isolation of natural products from complex mixtures, ing institutions so as not to discourage the industry from granting
these screens efficiently recover bioactive compounds when evaluating licenses for the marketing of the new drug [28].
crude extract libraries [28]. Consequently, using natural products and Finally, to determine our position concerning developing new anti­
their derivatives has evolved as a promising approach to combat microbials, as of November 2021, 80 products (46 antibiotics and 34
drug-resistant microbial strains. Furthermore, these natural products, non-traditional antibacterial agents) were in clinical development,
characterized by the presence of novel structures and mechanisms of including in the pre-registration phase [56]. They include 28 antibiotics
action, may guide the development of new potential chemotherapies and 21 non-traditional antibacterial targeting priority pathogens, 13
[18]. antibiotics and 1 non-traditional antibacterial targeting M. tuberculosis,
To fully exploit the privileged nature of natural products in antibiotic and 5 antibiotics and 12 non-traditional antibacterial targets C. difficile
drug discovery, the ability to rapidly identify scaffolds, tailor them using (Fig. 3). In the following link, you can find a table with detailed infor­
a library of modifying enzymes and rapidly purify the resulting com­ mation on these new antibiotics, discriminating by type and name of the
pounds in sufficient quantity to generate libraries of molecules suitable product, class of antibacterial, clinical trials, developer, therapeutic in­
for high throughput screening. The development of synthetic biology dications, route of administration and study phase.
offers an excellent opportunity to address these problems [47]. Despite
these advances, less than 0.1% of the 28,000 natural product antibiotics 3. Antimicrobial peptides
discovered in the last few decades are in clinical use. Many of the
remaining 99.9% will never be suitable as drugs due to intractable ef­ These AMPs presented many biological activities in defense against
ficacy problems, toxicity, stability, etc. [58]. pathogens and thus medical applications based on antimicrobial and
While a molecule may be active in a target-based assay, it will ulti­ anti-larvicidal activities [4], anti-HIV activities [17], inhibitory activity
mately fail to progress if it cannot reach its target in sufficient concen­ against α-amylase and trypsin activity, redox activity [4],
trations. Poor accumulation can be due to low drug cell wall penetration, ribosome-inactivating activity [19].
rapid removal of the drug from inside the cell via efflux pumps, or in the Molecules of natural origin have shown excellent characteristics as
worst-case a combination of both [30]. Intending to remedy the problem antibacterial drugs; many are plant metabolites. They also offer other
of shortage of new antibiotics, faced with the AMR challenge, the in­ possibilities as sources of molecules with antimicrobial potentials, such
dustry responded by focusing on synthetics, and some high-tech ap­ as AMPs, mainly thionins, plant defensins, and cyclotides [62,63] rich in
proaches came together to form the new rational cysteine and disulfide bonds subsequently used as their primary defense
design/genomics/combichem/HTS platform. against microbial infections [61]. AMPs are categorized according to
Conserved essential proteins identified through genomics were sup­ their structure, sequence, or mode of action, such as bacterial killing,
posed to serve as targets for HTS and rational design, producing new immunological regulation, inhibiting biofilm formation, or anti-cancer
antibiotics. However, it did not perform as expected, with oxazolidi­ and anti-viral function [64]. α-Helix, β sheets, and extended/random
nones being the only new class of synthetic compounds developed in coil peptides are three different forms of their secondary structures [65,
recent years [23]. Over the last decade, considerable progress has also 66].
been made in identifying gene products that are essential for bacterial
physiology and pathogenic attributes. As a result, there have been 3.1. Classes of AMPs
numerous suggestions for providing new targets or antibiotics. Howev­
er, there is a considerable gap between identifying an essential bacterial Antimicrobial peptides (AMPs) occur naturally in all living organ­
factor and the inhibitors that can form the basis for developing a new isms, such as bacteria, fungi, plants, and animals, and compose the first
drug [19]. line of defense innate immunity from animals and plants against path­
Various innovative and complementary technologies are required to ogens [83,84]. AMPs are cationic amphipathic, have 10 to 50 residues,
improve access to new product scaffolds for new antimicrobials. Thus, and are classified into several groups according to sequence, 3D struc­
computational tools could provide powerful help at different levels, with ture and function [59]. While in more complex organisms, AMPs play a
artificial intelligence playing a revolutionary role in identifying new critical function in natural immunity and help protect the host from
drug candidates [28]. Fortuitously, the renewed interest in natural diseases. Bacteria eliminate other bacteria that threaten their ecological
products as drug leads is concurrent with the development of new niche [60].
technologies in genomics, bioinformatics, analytical chemistry, and AMPs are classified in thionins [4,7,9,10], defensins [11,12],
chemical synthesis that enable a new assessment of the compatibility of hevein-like peptides [13–15], knottins [15–18], stable-like peptides [19,
natural products with modern drug discovery. Synthetic biology can 20], lipid transfer proteins [21–23], snakins [24–28], and cyclotides
play a leading role in this area. It holds real promise to help overcome [29–32]. These peptides are mainly found in seeds, leaves, flowers,
some challenges for increased integration of natural products in anti­ fruits, and other plant tissues. These AMPs range from 29 to 54 amino
biotic drug discovery [47]. acid residues, the residues being the thionins, the larger and cyclotides,
Together with the technical improvements in the research of new the smaller ones. Additionally, all these molecules were classified into
antibiotics, a greater state and private investment is required. In this subgroups and share many conserved cysteine residues involved in di­
sense, progress has been made, with the proposal of almost 50 incentive sulfide bonds leading to higher stability due to temperature, pH, and
strategies to increase the number of effective antibiotics. The two most proteolytic degradation [11–18]. The three-dimensional is represented
recent large-scale efforts in this area are in the final planning stages. by α-helix, parallel and antiparallel β-sheets, coiled, extended, and

5
Y.A.S. Guevara et al. Microbial Pathogenesis 179 (2023) 106108

Fig. 3. Antibacterial products in clinical development for priority pathogens WHO (2022). The data visualization shows the numbers of antibacterial products
by type (chart A.1), category of non-traditional products (chart A.2), pathogen category and phase of clinical development (chart A.3), expected activity against
priority pathogens (chart B), innovativeness (chart C.1) and if a novel chemical entity is involved or not (chart C.2). accessed on Pipeline of antibacterial products in
clinical development (who.int).

disordered structures. expression [25]. The authors used as a model the MtDef5, purified from
Medicago truncatula [23]. In this study, authors employed site-directed
3.1.1. Thionins mutagenesis in MtDef5 at the γ-core motif, changing the cationic resi­
Thionins are 45–48 amino acid residue peptides with and molecular dues. A defensin, as a model. The mutated MtDef5 lost antibacterial
weight of around 5 kDa [4]. According to amino acid sequence homol­ activity, revealing this region is important for antibacterial activity.
ogy, 3D structural and disulfide bridge positions can be classified into
five types (I–V) [12]. Several studies have reported the broad-spectrum 3.1.3. Hevein-like peptides
antimicrobial activity of thionins, such as antibacterial [14,16]. Taveira Hevein-like peptides are basic 29–45 amino acid residues peptides.
et al. [16] reported that NsW1 and NsW2 purified from Nigella sativa These peptides were first isolated from Hevea brasiliensis [27]. This
presented an attractive antibacterial potential against Bacillus subtilis group has the chitin-binding domain, antifungal activity and antibac­
and Staphylococcus aureus. NsW1 and NsW2 increased membrane terial activity. These peptides have a 3D structure comprising three
permeability, and atomic force microscopy revealed a change in the antiparallel beta-sheets and a short α-helix, stabilized by 3–5 disulfide
ultrastructure of cells. bonds [28]. Many studies focus on the antibacterial potential of
hevein-like peptides, all regarding antimicrobial activity against phy­
3.1.2. Defensins topathogens [30], which is not the focus of this review.
Defensins are AMPs widely expressed in live organisms, and they are
nontoxic molecules extremely active against bacteria [19]. Defensins 3.1.4. Knottins
have 45–54 amino acid residues and β-sheets. An α-helix 3D structure is Knottins are 30 amino acid residue peptides, including α-amylase,
supported by four disulfide bridges, making them resistant to proteolysis carboxypeptidase, and trypsin inhibitors [31]. These molecules were
and stable to extremes of pH and temperature. These molecules have a first discovered in the 1980s and have an interesting 3D structure with
variety of biological functions, such as inhibiting microbial growth and disulfide bridges conferring high thermal stability. Knottins are named
inhibiting α-amylase and trypsin activity [4]. “promiscuous peptides” because they present a lot of biological func­
Defensins are known based on their antimicrobial activity selectively tions, including antibacterial activity [31]. They can interact with ion
against bacteria at deficient concentrations. For example, The Dm- channels in membranes and cell membranes and acid-sense channels.
AMP1, a defensin from Dahlia merckii, presented an IC50 at 15 μg Those that target membranes can pass through and intracellular attack
mL− 1 for Bacillus subtilis [21]. Other defensins, such as VuDef2 from targets, such as carboxypeptidases, promoting resistance. They also have
Vigna unguiculata, inhibit Staphylococcus aureus at 15 μg mL− 1 [22]. antimicrobial activity against bacteria, fungi, viruses and insects [32].
PaDef, a defensin from avocado (Persea americana var. drymifolia), had Knottins present an amphipathic ability with cationic and hydrophobic
potent antibacterial activity against multidrug-resistant Escherichia coli portions, an essential feature to interact with membranes [33].
and S. aureus. The mechanism of antibacterial action is based on the
destabilization of the bacterial membrane. The most important infor­ 3.1.5. Stable-like peptides
mation about the mechanism of defensin’s antibacterial action came Stable-like peptides are small peptides with a typical Cys motif
from Velivelli et al.’s study [23]. In vitro, MtDef5 induced membrane (XnC1X3C2XnC3X3C4Xn), where X is any amino acid residue except
permeability and inhibited bacteria’s DNA replication and gene cysteine) forming a helix-loop-helix structure [34]. These peptides

6
Y.A.S. Guevara et al. Microbial Pathogenesis 179 (2023) 106108

present a highly conserved 3D structure in a staple-like form. Stable-like include D-amino acids, cyclization, acetylation, and peptidomimetics
peptides present a broad spectrum of biological activity, including [73,74].
antibacterial [34]. Many studies of stable-like peptides focused on their Synthetic peptides are typically more effective than natural peptides
biotechnological application in clinics based on their antibacterial ac­ since they exert more antimicrobial activity at lower concentrations
tivity. These peptides have high activity against E. coli [36]. The than their native natural peptides. For instance, the natural AamAP1
mechanisms of action behind the stable-like peptides did not involve from Androctonus amoreuxi exhibits antimicrobial action against
damage to cell membranes. In contrast, the peptide binds to DNA, Candida albicans, Escherichia coli, and Staphylococcus aureus [75]. The
inhibiting RNA and protein synthesis, producing a bactericidal effect synthesized form of AamAP1, AamAP1-Lysine, is 4–20 times more
[36]. effective than its natural form and has antimicrobial activity against the
same pathogens at concentrations as low as 5–7.5 μM [76]. In another
3.1.6. Lipid transfer proteins (LTPs) example, radish (Raphanus sativus L.) natural peptide Rs-AFP2 exhibits
LTPs are positive-charged peptides with a low molecular weight action against Fusarium culmorum.
(7–10 kDa) [37]. The 3D structures are pockets, with eight cysteines According to the Data Repository of Antimicrobial Peptides
forming four disulfide bonds stabilizing five α-helices [38]. The helices (DRAMP) database, until now, only 77 peptides have been created by
provide a hydrophobic spot essential to bind to lipids, including fatty pharmaceutical companies [77]. Other AMPs used in clinical settings to
acids, phospholipids, prostaglandin B2, hemolytic derivatives, and treat bacteria include those that work against Gram-positive bacteria
acyl-coenzyme A, involved in antibacterial activity [39]. like daptomycin, oritavancin, dalbavancin, teicoplanin, telavancin and
Regarding the activity of LTPs against human bacteria, CmLTP iso­ gramicidins, against Gram-negative bacteria like polymyxins (cyclic
lated from Chelidonium majus was active against Campylobacter jejuni, peptides from Paenibacillus polymyxa) [78]. Successful SAMPs should
Listeria greyi and Clostridium perfringens [41–43]. The mechanism of typically have apparent efficacy and be superior to conventional ther­
antimicrobial activity is based on the transfer of lipids in the membrane, apies. However, SAMPs’ advantage of causing less microbial resistance
leading to pore formation and cell imbalance. than traditional antibiotics is not considered, a feature that should not
be disregarded (reviewed by [79]).
3.1.7. Snakins
Snakins are a class of peptides with cysteine residues involved in 3.3. Synergy between AMPs and antibiotics
disulfide bonds contributing to stability and biological activities. The
general 3D structure of snakins is composed of an N-terminal signal In addition to killing multidrug-resistant (MDR) bacteria like MRSA
peptide, a variable site, and the C-terminal region [44–47]. and P. aeruginosa, several previously identified AMPs are employed with
These peptides can inhibit the growth of bacteria such as Micrococcus traditional antibiotics. Combining two antibacterial medicines results in
luteus and Staphylococcus cohnii. Snakins present prominent bactericidal better efficacy than using either separately, suggesting a synergistic
activity against Gram-negative bacteria. Furthermore, due to their interaction between them. Clinically, this combinational use is signifi­
ability to agglutinate microorganism cells, studies have shown that cant because it offers more alternatives for treating challenging in­
snakins can help to prevent the spread of pathogens in damaged tissues fections [80]. Additionally, AMPs have a more comprehensive range of
[48]. targets than most commonly used antibiotics, making their synergistic
beneficial combination in various contexts [81]. As a result, there is a
3.2. What are antimicrobial peptides (AMPs)? and could they contribute significant therapeutic promise for the clinical use of AMPs in combi­
to the fight against AMR? nation with other antimicrobial drugs, such as traditional antibiotics.
In some cases, combining AMPs with antibiotics enhances their
Multi-resistant bacteria have become a global public health problem, antibacterial and cytotoxic effects. For instance, when combined,
and introducing new antibiotics in the clinical phases is necessary. In gentamicin and AMP PG-1 have a synergistic cytotoxic impact on human
this context, AMPs have gained attention as a potential future thera­ neutrophils. Therefore, effective combinations should be carefully cho­
peutic candidate [67]. They play a crucial role in the body’s natural sen to find the best synergistic combination of AMPs and antibiotics to
defense and immune systems, which carry out a variety of efficient boost the antibacterial effects [82].
methods to eliminate pathogens [68]. Numerous antimicrobial effects
demonstrated by AMPs include disruption of cell membranes, inhibition 4. Conclusion
of protein and DNA synthesis, and suppression of crucial cellular pro­
cesses such as protein folding, cell wall formation, and metabolic turn­ The current antibacterial crisis reveals a bleak scenario that many
over [69]. experts classify as one of the worst health catastrophes that humanity
Some studies have shown the beneficial effects of AMPs in treating can suffer. Even though we already face problems resulting from AMR,
bacterial infections. For example, the VK25 peptide from the plasma of which may worsen in the long run. For this reason, it is vital that science,
the Komodo dragon (Varanus komodoensis) was used as a template to especially pharmaceuticals, lean towards research on new therapeutic
create a novel, short AMP, DRGN1, effective against P. aeruginosa and alternatives. In this context, antibacterial peptides, given their differ­
S. aureus biofilms of both single and mixed species. Pexiganan has entiated characteristics, are emerging as a tool of particular interest in
finished the phase 3 clinical trials for treating DFUs with bacterial in­ combating multi-resistant bacteria. Both clinical and preclinical studies
fections (NCT01590758). Additionally, several AMPs have been effec­ conducted with these molecules show their antibacterial therapeutic
tively created for commercial and pharmaceutical applications [70]. potential significantly when they are improved under a rational design
On the other hand, synthetic AMPs (SAMPs) constitute another scheme to reduce their toxic effects. It is expected that, with techno­
promising alternative to treating bacterial infections. The two goals of logical advances, especially with the advent and adoption of artificial
designing synthetic peptides from natural AMP sequences are to (1) intelligence, there will be an increase in diversified synthetic peptide
extract the better component, which has antimicrobial activity, and (2) production, which can face the challenges that we have in terms of
exclude the worse part, which is poisonous and has low proteolytic antibacterial drugs.
resistance [71]. With the help of in silico technology, specific amino
acids can be modified, and new AMPs can be designed and tested for CRediT authorship contribution statement
enhanced antibacterial activity [72]. For improved antibacterial activ­
ity, some of these alterations include the addition of hydrophobic and Yeimer A.S. Guevara: Writing – review & editing, Writing – original
cationic residues; for increased in vivo stability, other modifications draft, Funding acquisition, Formal analysis, Data curation,

7
Y.A.S. Guevara et al. Microbial Pathogenesis 179 (2023) 106108

Conceptualization. Maria H.C. Santos: Writing – review & editing, [20] B. Khameneh, R. Diab, K. Ghazvini, B.S.F. Bazzaz, Breakthroughs in bacterial
resistance mechanisms and the potential ways to combat them, Microb. Pathog. 95
Writing – original draft, Data curation, Conceptualization. Francisco I.
(2016) 32–42.
R. Gomes: Writing – review & editing, Writing – original draft, Funding [21] T. Lancet, The antimicrobial crisis: enough advocacy, more action, Lancet (London,
acquisition, Formal analysis, Data curation, Conceptualization. She­ England) 395 (2020) 247, 10220.
heryar: Writing – original draft, Formal analysis, Data curation, [22] K. Lewis, Platforms for antibiotic discovery, Nat. Rev. Drug Discov. 12 (2013)
371–387, 5.
Conceptualization. Felipe P. Mesquita: Writing – original draft, Visu­ [23] K. Lewis, New approaches to antimicrobial discovery, Biochem. Pharmacol. 134
alization, Validation, Data curation, Conceptualization. Pedro F.N. (2017) 87–98.
Souza: Writing – review & editing, Writing – original draft, Visualiza­ [24] B. Li, T.J. Webster, Bacteria antibiotic resistance: new challenges and opportunities
for implant-associated orthopedic infections, J. Orthop. Res. 36 (1) (2018) 22–32.
tion, Supervision, Project administration, Methodology, Investigation, [25] L.L. Ling, T. Schneider, A.J. Peoples, A.L. Spoering, I. Engels, B.P. Conlon, K. Lewis,
Funding acquisition, Formal analysis, Data curation, Conceptualization. A new antibiotic kills pathogens without detectable resistance, Nature 517 (7535)
(2015) 455–459.
[26] K.H. Luepke, K.J. Suda, H. Boucher, R.L. Russo, M.W. Bonney, T.D. Hunt, J.
F. Mohr III, Past, present, and future of antibacterial economics: increasing
Declaration of competing interest bacterial resistance, limited antibiotic pipeline, and societal implications,
Pharmacotherapy 37 (1) (2017) 71–84.
[27] G. Mancuso, A. Midiri, E. Gerace, C. Biondo, Bacterial antibiotic resistance: the
The authors declare that they have no known competing financial
most critical pathogens, Pathogens 10 (10) (2021) 1310.
interests or personal relationships that could have appeared to influence [28] M. Miethke, M. Pieroni, T. Weber, M. Brönstrup, P. Hammann, L. Halby, R. Müller,
the work reported in this paper. Towards the sustainable discovery and development of new antibiotics, Nat. Rev.
Chem 5 (10) (2021) 726–749.
[29] D.J. Morgan, I.N. Okeke, R. Laxminarayan, E.N. Perencevich, S. Weisenberg, Non-
Acknowledgments and Funding prescription antimicrobial use worldwide: a systematic review, Lancet Infect. Dis.
11 (9) (2011) 692–701.
[30] K.E. Murphy, G.F. Sloan, G.V. Lawhern, G.E. Volk, J.T. Shumate, A.L. Wolfe,
The authors are grateful to CNPq for providing funding for this work. Advances in antibiotic drug discovery: reducing the barriers for antibiotic
Pedro F. N. Souza is a senior researcher from CNPq at process number development, Future Med. Chem. 12 (22) (2020) 2067–2087.
305003/2022–4. [31] M. Nadimpalli, E. Delarocque-Astagneau, D.C. Love, L.B. Price, B.T. Huynh, J.
M. Collard, D. Guillemot, Combating global antibiotic resistance: emerging one
health concerns in lower-and middle-income countries, Clin. Infect. Dis. 66 (6)
References (2018) 963–969.
[32] K. Outterson, Estimating the appropriate size of global pull incentives for
antibacterial medicines: study examines global antibacterial pull incentives, Health
[1] A scientifc roadmap for antibiotic discovery, A sustained and robust pipeline of
Aff. 40 (11) (2021) 1758–1765.
new antibacterial drugs and therapies is critical to preserve public health (2016).
[33] G.L. Patrick, An Introduction to Medicinal Chemistry, Oxford University Press,
https://www.pewtrusts.org/en/research-and-analysis/reports/2016/05/a-scient
New York. cap, 2005, p. 16.
ific-roadmap-for-antibiotic-discovery.
[34] J. Pierce, A. Apisarnthanarak, N. Schellack, W. Cornistein, A. Al Maani, S. Adnan,
[2] B. Aslam, W. Wang, M.I. Arshad, M. Khurshid, S. Muzammil, M.H. Rasool, M.
M.P. Stevens, Global antimicrobial stewardship with a focus on low-and middle-
A. Nisar, R.F. Alvi, M.A. Aslam, U.M. Qamar, M.K.F. Salamat, Z. Baloch, Antibiotic
income countries: a position statement for the international society for infectious
resistance: a rundown of a global crisis, Infect. Drug Resist. (2018) 1645–1658.
diseases, Int. J. Infect. Dis. 96 (2020) 621–629.
[3] J.B. Bartlett, D.N. Gilbert, B. Spellberg, Seven ways to preserve the miracle of
[35] B. Plackett, No money for new drugs, Nature 586 (7830) (2020) S50–S52.
antibiotics, Clin. Infect. Dis. 56 (10) (2013) 1445–1450.
[36] S.J. Projan, D.M. Shlaes, Antibacterial drug discovery: is it all downhill from here?
[4] Boluarte & Schulze, The case for a subscription model to tackle antimicrobial
Clin. Microbiol. Infection 10 (4) (2014) 18–22.
resistance, Another Pandemic on the Horizon (2022). https://www.bcg.com/pt-br/
[37] L.B. Rice, Antimicrobial stewardship and antimicrobial resistance, Med. Clin. 102
publications/2022/model-for-tackling-antimicrobial-resistance.
(5) (2018) 805–818.
[5] H.W. Boucher, G.H. Talbot, D.K. Benjamin Jr., J. Bradley, R.J. Guidos, R.N. Jones,
[38] L.S. Roope, R.D. Smith, K.B. Pouwels, J. Buchanan, L. Abel, P. Eibich,
B.E. Murray, R.A. Bonomo, D. Gilbert, 10 × ′ 20 progress–development of new
S. Wordsworth, The challenge of antimicrobial resistance: what economics can
drugs active against gram-negative bacilli: an update from the Infectious Diseases
contribute, Science 364 (6435) (2019), eaau4679.
Society of America, Clin. Infect. Dis. 56 (2013) 1685–1694.
[39] C.K. Shore, A. Coukell, Roadmap for antibiotic discovery, Nat. Microbiol. 1 (6)
[6] R.R. Bragg, C.M. Meyburgh, J.Y. Lee, M. Coetzee, Potential treatment options in a
(2016) 1–2.
post-antibiotic era, Infect. Dis. Nanomed. III (2018) 51–61.
[40] R. Shukla, F. Lavore, S. Maity, M.G. Derks, C.R. Jones, B.J. Vermeulen,
[7] D.M. Brogan, E. Mossialos, A critical analysis of the review on antimicrobial
M. Weingarth, Teixobactin kills bacteria by a two-pronged attack on the cell
resistance report and the infectious disease financing facility, Glob. Health 12 (1)
envelope, Nature 608 (7922) (2022) 390–396.
(2016) 1–7.
[41] L.L. Silver, Appropriate targets for antibacterial drugs, Cold Spring Harbor
[8] E. Calbo, L. Boix-Palop, J. Garau, Clinical and economic impact of bacterial
perspectives in Med. 6 (12) (2016) a030239.
resistance: an approach to infection control and antimicrobial stewardship
[42] T. Singhal, Antimicrobial resistance: the’Other’Pandemic, Indian J. Pediatr. (2022)
solutions, Curr. Opin. Infect. Dis. 33 (6) (2020) 458–463.
1–7.
[9] S.T. Cole, Who will develop new antibacterial agents? Phil. Trans. Biol. Sci. 369
[43] B. Spellberg, The future of antibiotics, Crit. Care 18 (3) (2014) 228, https://doi.
(1645) (2014), 20130430.
org/10.1186/cc13948.PMID:25043962. PMCID: PMC4075146.
[10] J. Dai, R. Han, Y. Xu, N. Li, J. Wang, W. Dan, Recent progress of antibacterial
[44] B. Spellberg, J. Bartlett, R. Wunderink, D.N. Gilbert, Novel approaches are needed
natural products: future antibiotics candidates, Bioorg. Chem. 101 (2020), 103922.
to develop tomorrow’s antibacterial therapies, Am. J. Respir. Crit. Care Med. 191
[11] J.A. DiMasi, H.G. Grabowski, R.W. Hansen, Innovation in the pharmaceutical
(2) (2015) 135–140.
industry: new estimates of R&D costs, J. Health Econ. 47 (2016) 20–33.
[45] F.C. Tenover, Mechanisms of antimicrobial resistance in bacteria, Am. J. Infect.
[12] G.A. Durand, D. Raoult, G. Dubourg, Antibiotic discovery: history, methods and
Control 34 (5) (2006).
perspectives, Int. J. Antimicrob. Agents 53 (4) (2019) 371–382.
[46] A.K. Thabit, J.L. Crandon, D.P. Nicolau, Antimicrobial resistance: impact on
[13] L. Escolà-Vergé, I. Los-Arcos, B. Almirante, New antibiotics for the treatment of
clinical and economic outcomes and the need for new antimicrobials, Expet Opin.
infections by multidrug-resistant microorganisms, Med. Clínica 154 (9) (2020)
Pharmacother. 16 (2) (2015) 159–177.
351–357.
[47] M.N. Thaker, G.D. Wright, Opportunities for synthetic biology in antibiotics:
[14] L. Escolà-Vergé, C. Pigrau, B. Almirante, Ceftolozane/tazobactam for the treatment
expanding glycopeptide chemical diversity, ACS Synth. Biol. 4 (3) (2015) 195–206.
of complicated intra-abdominal and urinary tract infections: current perspectives
[48] U. Theuretzbacher, Accelerating resistance, inadequate antibacterial drug pipelines
and place in therapy, Infect. Drug Resist. 12 (2019) 1853.
and international responses, Int. J. Antimicrob. Agents 39 (4) (2012) 295–299.
[15] P. Fernandes, Antibacterial discovery and development the failure of success? Nat.
[49] U. Theuretzbacher, Antibiotic innovation for future public health needs, Clin.
Biotechnol. 24 (12) (2006) 1497–1503.
Microbiol. Infection 23 (10) (2017) 713–717.
[16] A. González, J. Casado, Á. Lanas, Fighting the antibiotic crisis: flavonoids as
[50] G.J. Tortora, B.R. Funke, C.L. Case, Microbiology, 10 Ed., Artmed, Porto Alegre,
promising antibacterial drugs against Helicobacter pylori infection, Front. Cell.
2012.
Infect. Microbiol. (2021) 671.
[51] D.J. Toth, M.H. Samore, R.E. Nelson, Economic evaluations of new antibiotics: the
[17] J. Gordon, O. Darlington, P. McEwan, M. Lumley, A. Taie, M. Hicks, M. Wilcox,
high potential value of reducing healthcare transmission through decolonization,
Estimating the value of new antimicrobials in the context of antimicrobial
Clin. Infect. Dis. 72 (Supplement_1) (2021) S34–S41.
resistance: development and application of a dynamic disease transmission model,
[52] T.I. Verhoef, S. Morris, Cost-effectiveness and pricing of antibacterial drugs, Chem.
Pharmacoeconomics 38 (8) (2020) 857–869.
Biol. Drug Des. 85 (1) (2015) 4–13.
[18] M. Igarashi, New natural products to meet the antibiotic crisis: a personal journey,
[53] C. Wang, H. Lu, X. Li, Y. Zhu, Y. Ji, W. Lu, C. Tan, Identification of an anti-
J. Antibiot. 72 (12) (2019) 890–898.
virulence drug that reverses antibiotic resistance in multidrug resistant bacteria,
[19] N. Jackson, L. Czaplewski, L.J. Piddock, Discovery and development of new
Biomed. Pharmacother. 153 (2022), 113334.
antibacterial drugs: learning from experience? J. Antimicrob. Chemother. 73 (6)
(2018) 1452–1459.

8
Y.A.S. Guevara et al. Microbial Pathogenesis 179 (2023) 106108

[54] W.R. Wong, A.G. Oliver, R.G. Linington, Development of antibiotic activity profile from choice of the best sequences to action mechanisms, Biochimie 175 (2020)
screening for the classification and discovery of natural product antibiotics, Chem. 132–145.
Biol. 19 (11) (2012) 1483–1495. [71] D. Ciumac, H. Gong, X. Hu, J.R. Lu, Membrane targeting cationic antimicrobial
[55] World Health Organization (WHO, Ten Threats to Global Health in 2019, 2019. peptides, J. Colloid Interface Sci. 537 (2019) 163–185.
[56] World Health Organization (WHO, Antibacterial Products in Clinical Development [72] P. Kumar, J.N. Kizhakkedathu, S.K. Straus, Antimicrobial peptides: diversity,
for Priority Pathogens, 2022. mechanism of action and strategies to improve the activity and biocompatibility in
[57] G.D. Wright, Something old, something new: revisiting natural products in vivo, Biomolecules 8 (1) (2018) 4.
antibiotic drug discovery, Can. J. Microbiol. 60 (3) (2014) 147–154. [73] M.D. Torres, S. Sothiselvam, T.K. Lu, C. de la Fuente-Nunez, Peptide design
[58] G.D. Wright, Opportunities for natural products in 21 st century antibiotic principles for antimicrobial applications, J. Mol. Biol. 431 (18) (2019) 3547–3567.
discovery, Nat. Prod. Rep. 34 (7) (2017) 694–701. [74] A. Almaaytah, M. Zhou, L. Wang, T. Chen, B. Walker, C. Shaw, Antimicrobial/
[59] [a] M. Pirtskhalava, A. Gabrielian, P. Cruz, H.L. Griggs, R.B. Squires, D.E. Hurt, cytolytic peptides from the venom of the North African scorpion, Androctonus
M. Tartakovsky, DBAASP v. 2: an enhanced database of structure and amoreuxi: biochemical and functional characterization of natural peptides and a
antimicrobial/cytotoxic activity of natural and synthetic peptides, Nucleic Acids single site-substituted analog, Peptides 35 (2) (2012) 291–299.
Res. 44 (D1) (2016) D1104–D1112; [75] A. Almaaytah, S. Tarazi, A. Abu-Alhaijaa, Y. Altall, N. Alshar’i, K. Bodoor, Q. Al-
[b] M. Hassan, M. Kjos, I.F. Nes, D.B. Diep, F. Lotfipour, Natural antimicrobial Balas, Enhanced antimicrobial activity of AamAP1-Lysine, a novel synthetic
peptides from bacteria: characteristics and potential applications to fight against peptide analog derived from the scorpion venom peptide AamAP1,
antibiotic resistance, J. Appl. Microbiol. 113 (4) (2012) 723–736. Pharmaceuticals 7 (5) (2014) 502–516.
[60] J.P. Tam, S. Wang, K.H. Wong, W.L. Tan, Antimicrobial peptides from plants, [76] G. Shi, X. Kang, F. Dong, Y. Liu, N. Zhu, Y. Hu, H. Xu, X. Lao, H. Zheng, Dramp 3.0:
Pharmaceuticals 8 (4) (2015) 711–757. an enhanced comprehensive data repository of antimicrobial peptides, Nucleic
[61] R. Nawrot, J. Barylski, G. Nowicki, J. Broniarczyk, W. Buchwald, A. Goździcka- Acids Res. 50 (D1) (2022) D488–D496.
Józefiak, Plant antimicrobial peptides, Folia Microbiol. 59 (3) (2014) 181–196. [77] N. Malanovic, K. Lohner, Antimicrobial peptides targeting gram-positive bacteria,
[62] H.U. Stotz, J. Thomson, Y. Wang, Plant defensins: defense, development and Pharmaceuticals 9 (3) (2016) 59.
application, Plant Signal. Behav. 4 (11) (2009) 1010–1012. [78] G.S. Dijksteel, M.M. Ulrich, E. Middelkoop, B.K. Boekema, Lessons learned from
[63] G. Jin, A. Weinberg, Human antimicrobial peptides and cancer, in: Seminars in Cell clinical trials using antimicrobial peptides (AMPs), Front. Microbiol. 12 (2021),
& Developmental Biology, 88, Academic Press, 2019, April, pp. 156–162. 616979.
[64] L.T. Nguyen, E.F. Haney, H.J. Vogel, The expanding scope of antimicrobial peptide [79] S.A. Li, Y. Xiang, Y.J. Wang, J. Liu, W.H. Lee, Y. Zhang, Naturally occurring
structures and their modes of action, Trends Biotechnol. 29 (9) (2011) 464–472. antimicrobial peptide OH-CATH30 selectively regulates the innate immune
[65] M. Pasupuleti, A. Schmidtchen, M. Malmsten, Antimicrobial peptides: key response to protect against sepsis, J. Med. Chem. 56 (22) (2013) 9136–9145.
components of the innate immune system, Crit. Rev. Biotechnol. 32 (2) (2012) [80] M. Cassone, L. Otvos Jr., Synergy among antibacterial peptides and between
143–171. peptides and small-molecule antibiotics, Expert Rev. Anti-infect. Ther. 8 (6) (2010)
[66] M. Zasloff, Antimicrobial peptides of multicellular organisms, Nature 415 (6870) 703–716.
(2002) 389–395. [81] L. Otvos Jr., V. de Olivier Inacio, J.D. Wade, P. Cudic, Prior antibacterial peptide-
[67] N.Y. Yount, M.R. Yeaman, Structural congruence among membrane-active host mediated inhibition of protein folding in bacteria mutes resistance enzymes,
defense polypeptides of diverse phylogeny, Biochimica. et Biophysica. Acta (BBA)- Antimicrob. Agents Chemother. 50 (9) (2006) 3146–3149.
Biomembr. 1758 (9) (2006) 1373–1386. [82] M.D.T. Torres, S. Sothiselvam, T.K. Lu, C. de la Fuente-Nunez, Peptide design
[68] C.F. Le, C.M. Fang, S.D. Sekaran, Intracellular targeting mechanisms by principles for antimicrobial applications, J. Mol. Biol. (2019), https://doi.org/
antimicrobial peptides, Antimicrob. Agents Chemother. 61 (4) (2017) e02340, 16. 10.1016/j.jmb.2018.12.015.
[69] H.Y. Wang, J.W. Cheng, H.Y. Yu, L. Lin, Y.H. Chih, Y.P. Pan, Efficacy of a novel [83] P.G. Lima, J.T. Oliveira, J.L. Amaral, C.D. Freitas, P.F. Souza, Synthetic
antimicrobial peptide against periodontal pathogens in both planktonic and antimicrobial peptides: characteristics, design, and potential as alternative
polymicrobial biofilm states, Acta Biomater. 25 (2015) 150–161. molecules to overcome microbial resistance, Life Sci. 278 (2021), 119647.
[70] P.F. Souza, L.S. Marques, J.T. Oliveira, P.G. Lima, L.P. Dias, N.A. Neto, F.E. Lopes, [84] Bechinger, S.-U. Gorr, Antimicrobial peptides: mechanisms of action and
J.S. Sousa, A.F. Silva, R.F. Caneiro, J.L. Lopes, Synthetic antimicrobial peptides: resistance, J. Dent. Res. 96 (2017) 254–260, https://doi.org/10.1177/
0022034516679973.

You might also like