Next Article in Journal
Review about Powerful Combinations of Advanced and Hyphenated Sample Introduction Techniques with Inductively Coupled Plasma-Mass Spectrometry (ICP-MS) for Elucidating Trace Element Species in Pathologic Conditions on a Molecular Level
Next Article in Special Issue
Hydrogel Formulations of Antibacterial Pyrazoles Using a Synthesized Polystyrene-Based Cationic Resin as a Gelling Agent
Previous Article in Journal
Curvicollide D Isolated from the Fungus Amesia sp. Kills African Trypanosomes by Inhibiting Transcription
Previous Article in Special Issue
Bactericidal Activity of a Self-Biodegradable Lysine-Containing Dendrimer against Clinical Isolates of Acinetobacter Genus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antimicrobial Peptides and Cationic Nanoparticles: A Broad-Spectrum Weapon to Fight Multi-Drug Resistance Not Only in Bacteria

1
Department of Experimental Medicine (DIMES), General Pathology Section, University of Genoa, 16132 Genoa, Italy
2
Department of Pharmacy, University of Genoa, 16148 Genoa, Italy
3
Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Viale Benedetto XV, 6, 16132 Genova, Italy
4
Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 6108; https://doi.org/10.3390/ijms23116108
Submission received: 10 May 2022 / Revised: 25 May 2022 / Accepted: 27 May 2022 / Published: 29 May 2022

Abstract

:
In the last few years, antibiotic resistance and, analogously, anticancer drug resistance have increased considerably, becoming one of the main public health problems. For this reason, it is crucial to find therapeutic strategies able to counteract the onset of multi-drug resistance (MDR). In this review, a critical overview of the innovative tools available today to fight MDR is reported. In this direction, the use of membrane-disruptive peptides/peptidomimetics (MDPs), such as antimicrobial peptides (AMPs), has received particular attention, due to their high selectivity and to their limited side effects. Moreover, similarities between bacteria and cancer cells are herein reported and the hypothesis of the possible use of AMPs also in anticancer therapies is discussed. However, it is important to take into account the limitations that could negatively impact clinical application and, in particular, the need for an efficient delivery system. In this regard, the use of nanoparticles (NPs) is proposed as a potential strategy to improve therapy; moreover, among polymeric NPs, cationic ones are emerging as promising tools able to fight the onset of MDR both in bacteria and in cancer cells.

1. Bacterial Multi-Drug Resistance: A General Overview

The discovery and use of antibiotics, starting from the second half of the twentieth century, has revolutionized modern medicine, thus allowing the treatment and prevention of infectious diseases and drastically improving people’s quality of life.
However, in the last few years, the phenomenon of antibiotic resistance has increased considerably, becoming nowadays one of the main public health problems worldwide. This entails important clinical implications (increase in morbidity, lethality, duration of the disease, possible development of complications and epidemics) and additional costs too, required for the use of expensive drugs and procedures, for the lengthening of hospital stays and for any disability. In fact, the continuous use of antibiotics promotes the onset and the diffusion of multi-drug resistance (MDR), further reducing the possibility of an effective treatment and increasing mortality [1]. In addition, while, until a few years ago, the presence of bacterial MDR was limited to the hospital environment, nowadays, it is widespread and involves several species of Gram-positive bacteria, such as Staphylococcus aureus, Enterococcus faecium, Enterococcus faecalis and Streptococcus pneumoniae and of Gram-negative pathogens, including the family of Enterobacteriaceae, and non-fermenting Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa [2].
Resistance may be due to intrinsic or acquired mechanisms [3]. While the former occurs naturally, the latter are a consequence of the stress induced by excessive treatment with antibiotics and may be due to genetic or epigenetic alterations [4,5]. In fact, antibiotic resistance may be the result of spontaneous mutations with the acquisition of resistant genes [6,7,8] and of several biochemical alterations, such as the decrease in drug efflux pump activity and membrane porins, leading to a reduction in the intracellular drug concentration [9,10] and drug inactivation through enzymatic action [11,12] or the modulation of metabolic pathways [13,14,15].
Therefore, since antibiotic resistance is one of the major public health problems today, it is crucial to find new strategies able to counteract it.

2. Sometimes They Come Back: Old Strategy to Defeat the Resistance

Although many novel molecules with antimicrobial activity have been synthesized, only a few are undergoing clinical trials and are active against MDR pathogens [16]. For this reason, research has focused on finding other alternative strategies. Various approaches have been proposed, such as the use of “old” antibiotics to which MDR bacteria should no longer be resistant [17], or even more specific ones obtained by changing the molecular structure to restore their activity, as in the case of resistance to vancomycin [18].
Antibiotic adjuvants have been developed and used in therapy to restore the activity of existing drugs no longer functioning, such as, for example, the β-lactamase inhibitors, including clavulanic acid, β-lactam-like sulfones (sulbactam and tazobactam), diazabicyclooctanes (avibactam and relebactam) and boronic acids (vaborbactam) [19]. In fact, unfortunately, the appearance of new variants of β-lactamase enzymes and the emergence and spread of the ultra-broad-spectrum metallo-β-lactamases of class B dramatically reduce the effectiveness of this strategy [19]. Other approaches to overcome MDR include the use of inhibitors of drug efflux pumps and outer membrane permeabilizers able to promote antibiotic uptake and to improve their concentration at target sites [20,21], as well as the employment of membrane disruptor molecules. In this context, the compound 2-((3-[3,6-dichloro-9H-carbazol-9-yl]-2-hydroxypropyl) amino)-2-(hydroxy-methyl) propane-1,3-diol (DCAP) is an antibacterial agent that causes cell lysis by altering membrane potential and permeability. It acts specifically on the membranes of bacteria and has a broad spectrum of action on both Gram-positive and Gram-negative pathogens [22].
Recently, the efficacy of Odilorhabdins against MDR bacteria has been proposed. These compounds interfere with protein synthesis by binding to the small subunit of bacterial ribosomes, which is different from the target subunit of the traditional and no longer active antibiotics [23]. Moreover, it has also been reported that the benzoimidazole moiety, already used for the treatment of other diseases, inhibits peptide deformylase and thus bacteria protein synthesis [24,25] and could represent a novel pharmacophore for the synthesis of new antibiotic agents.
The use of metal-based antibacterial agents has also given promising results, exploiting the multiple mechanisms of action of metal complexes, mainly with ruthenium, gallium, bismuth, silver and copper [26,27]. Another strategy involves the use of bacteriophages, viral agents that exclusively infect bacteria, exerting bactericidal activity; these could have a synergistic action when given in combination with antibiotics [28,29].

3. New Approaches to Defeat the Resistance

The high incidence of the selection of antibiotic-resistant bacterial strains highlights the urgent need to develop effective therapeutic strategies aimed to counteract this phenomenon. In this context, the use of membrane-disruptive peptides/peptidomimetics (MDPs) is receiving particular attention and, among these, antimicrobial peptides (AMPs), both of natural (NAMPs) and synthetic (SAMPs) origin, seem to be promising [30,31]. NAMPs are short cationic peptides that play a fundamental role in the innate immunity [32,33,34]. The amphiphilic structure and the presence of a net positive charge are the most important characteristics that typify NAMPs and that influence their mode of action [35,36]. In particular, NAMPs, as with other cationic materials, act mainly with a rapid and non-specific disruptive action on bacterial membranes, not allowing the pathogens to organize adaptive processes for becoming resistant [37]. Therefore, it is clear that NAMPs, in order to exert their antibacterial action, must be able to easily and firmly interact with the bacterial membranes. In this regard, it has been widely recognized that the electrostatic interactions between NAMPs and the negatively charged lipids of the outer envelope of Gram-negative bacteria and of the surface of Gram-positive bacteria are the main factors responsible for the higher selectivity of NAMPs towards bacteria [38,39,40,41,42,43,44,45,46,47]. Notably, the membrane of eukaryotic cells is mainly composed of lipids with zero net charge, thus drastically lowering the probabilities of electrostatic interaction with cationic materials (Figure 1) [38,39,40,41,42,43,44,45,46,47]. Additionally, mammalian cells are less susceptible to the detrimental effects of NAMPs due to the presence of high amounts of cholesterol, which contributes to stabilizing the membrane (Figure 1) [48].
However, although the primary target of NAMPs is the lipid matrix of the bacterial cell membrane [46], it has been reported that, thanks to their amphiphilic characteristic, several NAMPs also enter the bacterial cells and exert their toxic effect by impairing DNA, RNA and enzymes, thus enhancing their original therapeutic efficacy [49,50,51].
The reduced ability of NAMPs to induce resistance and to exert toxic effects on healthy cells has encouraged the development of SAMPs, which, compared to NAMPs, possess a more easily modifiable chemical structure, allowing one to obtain molecules capable of interacting more strongly with the surface of bacteria, thus achieving greater antimicrobial activity, better physicochemical stability, and greater compatibility with drug delivery methodologies [52]. As for NAMPs, for SAMPs, the determining factor responsible for their antibacterial action is the amphiphilicity and cationic character [53,54,55]. Moreover, it has been found that the enhanced interactions of SAMPs with the negatively charged lipids of bacterial membranes not only induce membrane damage, but also favor the entry of synthetic peptidomimetics into bacteria [56,57], thus exerting a toxic action at the intracellular level [58].
However, the clinical application of these SAMPs as new antibiotics is still hampered by high production costs, elevated hemolytic toxicity and a reduced half-life in serum [59,60,61,62,63,64].

4. Multi-Drug Resistance: A Common Feature of Bacteria and Cancer Cells

4.1. Bacteria and Cancer Cells: What Do They Share?

A very similar phenomenon of drug resistance has been observed in the treatment of various kinds of cancer. In fact, after an initial positive response of the anticancer therapy, the onset of MDR and the negative prognosis occurs [65,66]. Cancer cells have been found to acquire an MDR phenotype through several mechanisms, including increased expression of MDR transporters, which increase drug efflux and/or reduce its uptake; defects in the apoptotic program; promotion of the autophagy process; alteration of drug metabolism and cancer cell metabolism and alteration of intracellular redox homeostasis [67,68,69,70,71,72,73,74]. Notably, many of these mechanisms are the same as those that allow bacteria to develop antibiotic resistance. However, cancer cells, similarly to bacteria, have negatively charged lipids in their cell membranes [75], while healthy mammalian cells have neutrally charged lipids in the cell membrane (Figure 2) [76].
Moreover, using NAMPs as template molecules, SAMPs are continuously developed to produce de novo scaffolds that resemble the parent peptides, which can maintain therapeutic effectiveness with higher biological stability and a greater safety profile. Therefore, it is possible to hypothesize that NAMPs, as well as SAMPs, by selectively interacting with the negatively charged lipids of the tumor cell membrane and preserving healthy cells from injury, might be a promising approach to counteract cancer chemoresistance [77].

4.2. Anticancer Peptides to Fight Cancer Chemoresistance

Several structural and functional studies led to the evidence of cationic low-molecular-weight peptides having antitumor activity, which were then classified as anticancer peptides (ACPs) [78,79,80,81,82]. In fact, cationic peptides, isolated from various organisms and historically known for their antimicrobial activities, were studied and described as potent anticancer agents in 1985 [83]. The use of these peptides to treat cancer has received considerable support since, as mentioned, cancer cells have negatively charged membrane lipids, which are the elective targets of these compounds (Figure 2) [84,85,86]. In addition, cancer cell membranes have many microvilli and therefore a greater surface for interaction with ACPs in comparison with healthy cells (Figure 2) [77,87].
Another factor that could influence the cancer cell response to ACPs is the membrane cholesterol content. In fact, it has been reported that cancer cells (breast and prostate cancers) having high cholesterol levels are protected from the action of APCs, while other ones (leukemia and lung cancer) with low cholesterol content are more susceptible to the ACP-mediated lytic action [88,89,90].
Additionally, ACPs can induce cancer cell death by disrupting mitochondrial membrane integrity, leading to cytochrome C release and apoptosis induction [91,92,93,94,95,96,97,98]. Moreover, it has been demonstrated that ACPs can exert their anticancer activity by activating other mechanisms, such as immunogenic cell death [99], inhibition of DNA polymerase [100] and an anti-angiogenic action [101].
However, although the potential for the clinical use of ACPs is promising, it is necessary to take into account that all the above-mentioned characteristics of cancer cells can interfere with the therapeutic efficacy of this approach.

5. Clinical Development of AMPs and ACPs: An Open Issue

Based on the consideration that both AMPs and ACPs are not traditional drugs and that bacterial and cancer cells share some structural features, the clinical development of both AMPs and ACPs faces similar challenges and limitations. For this reason, we have focused our attention on the factors that could limit the clinical development of AMPs, reasonably assuming that such issues could, potentially, also be found using ACPs.
Firstly, since NAMPs have structures that are not optimized for a specific antibacterial or anticancer action, a fundamental step for their clinical development is their structural optimization. Other critical issues that can limit the employment of NAMPs consist in their poor pharmacokinetic (PK) properties, the high cost of synthesis, their inability to recognize specific receptors and the reduced correlation between in vitro results and those found in animal models [37,102,103,104,105,106].
However, parenterally administrable SAMPs with optimized structures have been already successfully developed [104,105,107,108]. Moreover, although the peptidic nature of AMPs precludes their oral delivery, various specific delivery systems have been engineered to escape the degradative attack of digestive enzymes and to increase their intestinal absorption [77]. Several strategies can be used to enhance the PK properties of AMPs, including the use of the D-enantiomers constituting amino acids to increase stability [109], and the packaging of AMPs in inactive liposomes targeting a specific cancer cell ligand in order to specifically deliver the active peptide [110,111,112,113,114].
With regard to the high cost of production, the problem may be overcome by the application of modern production technologies, such as solid-phase synthesis, and by synthesis on a large scale. Furthermore, the additional difficulty related to the fact that AMPs select their targets by non-receptor-mediated recognition and rather through the membrane properties may represent a strength point since it leads to a lower likelihood of developing resistance.
Despite the above-reported problems, several ACPs show antitumor effects (Table 1) due to their unique mechanism of action, to their relatively high tissue penetration and to the low acquisition of drug resistance; they have many advantages over conventional chemotherapy and some of them are currently being studied for clinical application in oncologic patients (Table 2) [115].

5.1. Insight into the Antitumor Activity of ACPs

5.1.1. ACPs as Membrane Disruptors

It is universally recognized that ACPs act mainly as membrane-active molecules and two models of action have been described. The first one is the “barrel-stave” model [138], in which ACPs induce pore formation in the cell membrane, by rendering cancer cells unable to maintain normal osmotic pressure (Figure 3). Subsequently, the “carpet” model has been proposed, according to which ACPs induce cell death by destroying the cell membrane, thus causing the massive leakage of cytoplasmic content [139], as observed in melanoma, gastric cancer, liver cancer and cervical cancer cells treated with HPRP-A1-TAT, a hybrid peptide [140] (Figure 3). Another example is Temporin-La, which exerts its anticancer activity by destroying the cell membrane [141]. However, the detailed mechanism of interaction between ACPs and the cell membrane needs to be further studied.

5.1.2. ACPs: Other Mechanisms of Action

More in-depth studies have demonstrated that ACPs can also lead to the apoptosis of cancer cells by destroying the mitochondrial membrane and thus favoring the release of cytochrome C. Such a mechanism has been observed, for example, for Ra-V, Dolastatin 10 (Dolabella auricularia) and the Bacillus subtilis lipopeptide. Other peptides, such as KV11, FN070315 and Temporin-1CEa, exert their anticancer activity by inhibiting tumor angiogenesis [142,143,144]. Moreover, immune regulation is the main mechanism of action of Bovine lactoferrin (LfcinB) and MENK (an endogenous neuropeptide) [145,146]. Other studies have shown that MENK acts as an immune booster and can inhibit the proliferation of human cancer cells through cyclin-dependent kinase inhibition pathways [147,148,149].
Collectively, although the development of adequate systems able to facilitate their delivery remains one of the limiting problems, to date, AMPs potentially possess all the essential features to become the only class of drugs able to fight MDR both in bacterial infections and in cancer.

6. Nanoparticle (NP)-Based Delivery Systems: A Solution for the Clinical Application of AMPs?

To make possible the clinical application of AMPs and ACPs, the development of appropriate delivery systems is fundamental and several strategies to design favorable formulations of these peptides have been developed. Among these, nanoscale delivery systems (liposomes, micelles, and NPs) appear to be valid candidates due to their high biocompatibility. Moreover, such systems are also able to improve drug bioavailability by reducing its early elimination and permitting its gradual and sustained release [76]. Concerning this, it is possible to manufacture stimuli-responsive nanocarriers, whose drug release is controlled and triggered by changes in pH or temperature, by alterations in redox state or by the presence of specific microenvironment components. In addition, several NPs offer the possibility of modifying their surface by using poly-(ethylene glycol) (PEG) to increase the NP blood circulation half-life [150,151]. These include the materials used to prepare such NPs, including natural lipids, proteins, and carbohydrates, as well as synthetic polymers, dendrimers, metals, etc. [152,153]. Unlike free drugs, those formulated in NPs have reduced toxicity toward healthy tissue, can be delivered to specific targets, possess improved pharmacological properties, and allow simultaneous combined administration.

6.1. Use of NP-Based Delivery Systems to Overcome Antibiotic Resistance

To improve the therapeutic efficacy of antibiotics and prevent the acquisition of resistance by bacteria, the use of NPs has been proposed as a promising strategy [151,154,155]. To this end, several NPs able to specifically direct antibiotics to bacteria and to prolong their half-life have been developed. Recently, anionic, functionally mesoporous silica NPs able to encapsulate high amounts of polymyxin B, a long-established NAMP, maintaining its antibacterial activity and improving its biocompatibility, have been formulated [156].
Moreover, azithromycin (AZM)-conjugated clustered NPs (AZM-DA NPs), able to release secondary AZM-conjugated PAMAM NPs (PAMAM-AZM NPs) upon disassembling under an acidic biofilm microenvironment, have been reported [157]. In particular, the positive charge and the small size of these PAMAM-AZM NPs were fundamental to improve their penetration and retention within the biofilm produced by bacteria, to make the bacterial cell more easily permeable and to allow the increased uptake of AZM.

6.2. Use of NPs to Counteract Cancer Chemoresistance

Although the research on anticancer polymer/peptide nanocomposites is a recent field of interest, the results obtained so far seem promising. In fact, anticancer polymer/peptide NPs have been shown to be able to reduce the onset of chemoresistance, to inhibit tumor growth and to limit the formation of metastases in vivo [31]. In addition, it has been observed that such anticancer polymer/peptide NPs not only exhibit improved retention times and permeation effects in tumor tissues, but also reduced toxicity towards healthy tissues in respect to non-polymer-modified peptides [31]. Specifically, it has been reported that several polymer-based NAMP- or SAMP-loaded NPs, by facilitating cell uptake, provided enhanced therapeutic effects. Notably, a pH-sensitive ACP-loaded polymeric micelle has been developed by conjugating the synthetic poly (amino ester)-poly-(ethylene glycol) copolymer to the therapeutic peptide CGKRKD (KLAKLAK), to increase the intracellular ACP concentration [158]. Furthermore, in the study of Li’s group, melittin (MLT)-loaded zeolitic imidazolate framework-8 (MLT@ZIF-8) NPs able to improve MLT stability and to inhibit its hemolytic activity were developed [159]. In addition, in respect to free MLT, this nano-formulation displayed enhanced antitumor effects, due to the increased uptake of the drug [159].

6.3. Cationic NPs: Not Only Delivery Systems

Among polymeric NPs, cationic ones are emerging as macromolecules that could play an important role against MDR bacteria, as well as MDR cancer cells. As previously reported, one of the causes of antibiotic resistance is the reduced or nullified capability of the drugs to penetrate bacterial membranes [160]. Gram-positive bacteria—and, even more so, Gram-negative ones—have highly organized external membranes with negative charges [161]. In this context, cationic NPs, used to encapsulate AMPs, are capable of selectively interacting with the net anionic charges of bacterial membranes, rather than with the zwitterionic ones of eukaryotic cells, and considerably reduce AMP-mediated toxic effects [162]. Additionally, cationic NPs can form complexes with other macromolecules with negative charges, such as DNA or proteins exerting intrinsic antibacterial and anticancer activity [163]. A great advantage of cationic NPs, with respect to cationic lipids, is represented by the smaller size and the higher loading capacity, thus facilitating their internalization by target cells. Furthermore, being highly cationic, they are completely water-soluble [164].
A very large number of positively charged NPs and nanogels are currently used for various purposes, including the encapsulation of AMPs, and there are many production procedures and/or polymerization methods, which mainly involve either the use of cationic monomers to prepare polymeric NPs or nanogels or cationic reagents to be covalently bound or absorbed to previously prepared NPs or nanogels [165].
The most efficient and used techniques to synthesize and produce polymeric NPs are those related to hetero-phase polymerization processes, including emulsion polymerization [166]. To obtain cationic NPs, a combination of different reagents is necessary, such as cationic initiators, monomers, polymers, and surfactants [165]. The choice of monomers or polymers to use depends on the type of the desired charge, which could be permanent or pH-dependent. The most used monomers are vinyl-pyridines and quaternary ammonium cationic monomers, while polymers include, but are not limited to, ethyl methacrlates. Notably, even if the synthesis of cationic NPs is mainly surfactant-free, in some cases, surfactants are used since they help to solubilize the monomers by forming micelles and stabilize the polymer during formation. Additionally, surfactants have also a fundamental role in the size and stability of the obtainable NPs and, therefore, the amount of these compounds is crucial for successful preparations [167]. The most common cationic surfactants used are quaternary ammonium salts such as hexadecyltrimethylammonium- or dodecyltrimethylammonium-bromide, as well as cetyltrimethylammonium chloride [168].

6.3.1. Antibacterial Function of Cationic NPs

Several innovative strategies involving cationic NPs have been developed to counteract MDR bacteria. Notably, Hou et al. have selected from chitosan-grafted oligolysine chains cationic NPs that spontaneously self-assemble in water, with great efficacy both in vitro and in vivo against some methicillin-resistant Staphylococcus aureus and MDR E. coli and P. aeruginosa [169]. These NPs, with multiple positive charges in their polymer chains and a high charge density, are able to form hydrogen bonds between chitosan chains and bacteria, leading to membrane cell injury and thereby exerting their bactericidal activity (Figure 4).
The same mechanism of membrane disruption has been reported for branched polyethyleneimine-functionalized silver nanoclusters, which exert a bactericidal effect against MDR infections, thanks to the synergy of silver’s antimicrobial activity with cationic NPs’ selectivity towards bacterial membranes, resulting in low toxicity for mammalian cell membranes [170]. In this context, Chen et al. have developed NPs of metal–organic frameworks incorporated with titanium, which are able to increase reactive oxygen species (ROS) production and are employed for the photodynamic therapy of chronic wounds infected by MDR bacteria [171].
Two years ago, three non-cytotoxic dendrimers of fifth generation (G5D) were synthetized and proven to be effective against non-fermenting MDR Gram-negative species such as P. aeruginosa, S. maltophilia and A. baumannii [172]. More recently, a lysine-modified cationic polyester-based dendrimer (G5-PDK), capable of electrostatically interacting with bacterial surfaces, thus leading to membrane injury, has been synthetized and characterized [173]. G5-PDK was demonstrated to have bactericidal effects specifically against isolates of the Acinetobacter genus, including A. baumannii, responsible for difficult-to-treat nosocomial infections [173], but also against different species of the Pseudomonas genus [174]. In this latter study, it was established that the antibacterial potency of G5-PDK depended on the capability of Pseudomonas to produce cationic or anionic pigments, thus confirming the above-mentioned mechanism of action of cationic NPs and their high selectivity for bacteria [174].
As previously reported, the overall merit of the cationic NPs consists of their intrinsic antibacterial/bactericidal effects regardless of their association with additional antibacterial agents.
Furthermore, other research groups have also developed strategies for recycling antibiotics that are no longer functioning, taking advantage of NPs. This approach could allow researchers to avoid some procedures in terms of the research and regulatory issues, which are instead mandatory in the case of the development of new antibacterial molecules. In this regard, Si et al. recently proposed a new biodegradable and biocompatible chitosan-derived cationic antibacterial polymer with promising in vitro and in vivo antibacterial activity against MDR bacteria including Listeria monocytogenes, S. aureus, E. coli, K. pneumoniae, P. aeruginosa and A. baumannii. Notably, it showed a synergistic effect with many clinically relevant antibiotics, including amikacin, tobramycin, novobiocin, rifampicin and tazobactam [175]. In addition, Gupta et al. developed NPs able to block the efflux pumps of bacterial membranes, allowing them to regulate the dosage of antibiotics whose optimal efficacy depends on the choice of the cationic surface of the NPs [176].
Additionally, cationic NPs, and particularly dendrimers, with a cationic surface and a non-charged inner matrix, have been recently developed. These formulations possess both hydrophobic internal cavities, able to host hydrophobic small molecules, and hydrophilic exterior functions, which make them highly soluble in water. These macromolecules, in addition to interacting with the bacterial surface, can encapsulate water-insoluble bioactive molecules, otherwise not administrable and not clinically applicable. In this context, ursolic acid or pyrazole derivatives known to be endowed with several pharmacological activities have been successfully encapsulated in fourth- and fifth-generation cationic dendrimers modified with lysine, obtaining drug-loaded nanocomposites with proven high-water solubility and loading capacity [177,178,179]. When tested on bacteria, these nanocomposites displayed potent antibacterial activity, much higher than that of the encapsulated molecules, against Gram-positive MDR isolates of the Staphylococcus [180] and Enterococcus [181] genera, regardless of the intrinsic antibacterial effects of the dendrimers used as encapsulating agents. This achievement can be explained assuming that the cationic property of the NPs enveloping the bioactive molecules allowed their accumulation on the surfaces of bacteria and promoted their entry into the cells via pore formation.

6.3.2. Antitumor Function of Cationic NPs

The compounds and mechanisms of action of cationic NPs used to fight MDR cancer cells are very similar to those used against MDR bacteria. In fact, the use of NPs seems to be promising due to their ability to interact preferentially with the negatively charged membranes of cancer cells, thereby preserving the neutrally charged membranes of healthy cells (Figure 4). Indeed, cationic NPs for cancer therapy can be obtained through polyplexes, which are spontaneous complexes obtained either from the condensation of cationic polymers, directly from cationic polymers, by the coating of non-cationic NPs through copolymerization or by electrostatic interactions with cationic polymers, lipids, surfactants, or dendrimers [182].
Recently, Zhang et al. synthesized a novel compound consisting of cationic NPs based on hyaluronic acid encapsulated with docetaxel, which showed promising effects on MDR breast cancer cells. Specifically, this combination of substances was able to reduce the levels of CYP1B1, an enzyme over-expressed in breast cancer and responsible for resistance, and to improve the bioavailability of the antitumor agent [183].
The great efficacy of these compounds against resistant cancer cells is a promising starting point for the development of new therapies based on cationic NPs. In this context, we recently synthetized and tested on Etoposide-sensitive and Etoposide-resistant neuroblastoma cells two polystyrene-based (P5, P7) cationic nanosized copolymers, containing primary ammonium groups. Both copolymers were able to reduce cell viability and to increase ROS production in both cancer cell lines, demonstrating greater potency against Etoposide-resistant ones [184]. The same copolymers were assayed on more than sixty isolates of both Gram-positive and Gram-negative species and showed ultra-broad-spectrum bactericidal activity [185,186], thus confirming their common mechanism of action on bacteria and cancer cells.

7. Conclusions and Final Remarks

Despite continued efforts to develop new antibiotic and anticancer therapies, the onset of resistance limits the effectiveness of such approaches, having significant repercussions for the health of the world population. For this reason, in recent years, research has increasingly focused on identifying alternative strategies. In this context, the use of MDPs is receiving particular attention and, among these, AMPs and ACPs, of natural or synthetic origin, seem to be promising. In fact, these systems, being able to specifically act on bacteria or cancer cells, limit the drug-induced secondary toxic effects. However, it is necessary to consider that the clinical development of both AMPs and ACPs shows several challenges and limitations, with their delivery identified as the most limiting problem. Among polymeric NPs, cationic ones are emerging and could have a relevant impact, both functioning as efficient delivery systems and exerting potent antibacterial and anticancer effects not only mediated by the cell membrane injury.
Based on these findings and considering that therapy resistance is an alarming concern for global health, great efforts are needed to design and optimize cationic NP-based therapies and to progress them to clinical application.

Author Contributions

G.E.V., S.A., D.C., C.D. and B.M. contributed equally to this manuscript review. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cosgrove, S.E. The Relationship between Antimicrobial Resistance and Patient Outcomes: Mortality, Length of Hospital Stay, and Health Care Costs. Clin. Infect. Dis. 2006, 42, S82–S89. [Google Scholar] [CrossRef] [Green Version]
  2. Breijyeh, Z.; Jubeh, B.; Karaman, R. Resistance of Gram-Negative Bacteria to Current Antibacterial Agents and Approaches to Resolve It. Molecules 2020, 25, 1340. [Google Scholar] [CrossRef] [Green Version]
  3. Sefton, A.M. Mechanisms of Antimicrobial Resistance. Drugs 2002, 62, 557–566. [Google Scholar] [CrossRef]
  4. Thomas, C.M.; Nielsen, K.M. Mechanisms of, and Barriers to, Horizontal Gene Transfer between Bacteria. Nat. Rev. Microbiol. 2005, 3, 711–721. [Google Scholar] [CrossRef]
  5. Wilson, D.N. Ribosome-targeting antibiotics and mechanisms of bacterial resistance. Nat. Rev. Microbiol. 2014, 12, 35–48. [Google Scholar] [CrossRef]
  6. Rice, L.B. The clinical consequences of antimicrobial resistance. Curr. Opin. Microbiol. 2006, 12, 476–481. [Google Scholar] [CrossRef]
  7. Alekshun, M.N.; Levy, S.B. Molecular Mechanisms of Antibacterial Multidrug Resistance. Cell 2005, 128, 1037–1050. [Google Scholar] [CrossRef] [Green Version]
  8. Soucy, S.M.; Huang, J.; Gogarten, J.P. Horizontal gene transfer: Building the web of life. Nat. Rev. Genet. 2015, 16, 472–482. [Google Scholar] [CrossRef]
  9. Webber, M.A.; Piddock, L.J.V. The importance of efflux pumps in bacterial antibiotic resistance. J. Antimicrob. Chemother. 2003, 51, 9–11. [Google Scholar] [CrossRef]
  10. Levy, S.B. Active efflux mechanisms for antimicrobial resistance. Antimicrob. Agents Chemother. 1992, 36, 695–703. [Google Scholar] [CrossRef] [Green Version]
  11. Bastida, A.; Hidalgo, A.; Chiara, J.L.; Torrado, M.; Corzana, F.; Pérez-Cañadillas, J.M.; Groves, P.; Garcia-Junceda, E.; Gonzalez, C.; Jimenez-Barbero, A.J.; et al. Exploring the Use of Conformationally Locked Aminoglycosides as a New Strategy to Overcome Bacterial Resistance. J. Am. Chem. Soc. 2005, 128, 100–116. [Google Scholar] [CrossRef] [Green Version]
  12. Magnet, S.; Blanchard, J.S. Molecular Insights into Aminoglycoside Action and Resistance. Chem. Rev. 2004, 105, 477–498. [Google Scholar] [CrossRef]
  13. Sköld, O. Sulfonamide resistance: Mechanisms and trends. Drug Resist. Update 2000, 3, 155–160. [Google Scholar] [CrossRef]
  14. Wise, E.M.; Abou-Donia, M.M. Sulfonamide resistance mechanism in Escherichia coli: R plasmids can determine sulfonamide-resistant dihydropteroate synthases. Proc. Natl. Acad. Sci. USA 1975, 72, 2621–2625. [Google Scholar] [CrossRef] [Green Version]
  15. Amyes, S.; Smith, J. R-factor trimethoprim resistance mechanism: An insusceptible target site. Biochem. Biophys. Res. Commun. 1974, 58, 412–418. [Google Scholar] [CrossRef]
  16. Otsuka, Y. Potent Antibiotics Active against Multidrug-Resistant Gram-Negative Bacteria. Chem. Pharm. Bull. 2020, 68, 182–190. [Google Scholar] [CrossRef] [Green Version]
  17. Theuretzbacher, U.; Van Bambeke, F.; Cantón, R.; Giske, C.G.; Mouton, J.W.; Nation, R.L.; Paul, M.; Turnidge, J.D.; Kahlmeter, G. Reviving old antibiotics. J. Antimicrob. Chemother. 2015, 70, 2177–2181. [Google Scholar] [CrossRef] [Green Version]
  18. Mühlberg, E.; Umstätter, F.; Kleist, C.; Domhan, C.; Mier, W.; Uhl, P. Renaissance of vancomycin: Approaches for breaking antibiotic resistance in multidrug-resistant bacteria. Can. J. Microbiol. 2020, 66, 11–16. [Google Scholar] [CrossRef] [Green Version]
  19. Alfei, S.; Zuccari, G. Recommendations to Synthetize Old and New β-Lactamases Inhibitors: A Review to Encourage Further Production. Pharmaceuticals 2022, 15, 384. [Google Scholar] [CrossRef]
  20. Wright, G.D. Antibiotic Adjuvants: Rescuing Antibiotics from Resistance. Trends Microbiol. 2016, 24, 862–871, Erratum in 2016, 24, 862–871. [Google Scholar] [CrossRef]
  21. Douafer, H.; Andrieu, V.; Phanstiel, O.; Brunel, J.M. Antibiotic Adjuvants: Make Antibiotics Great Again. J. Med. Chem. 2019, 62, 8665–8681. [Google Scholar] [CrossRef]
  22. Eun, Y.-J.; Foss, M.H.; Kiekebusch, D.; Pauw, D.A.; Westler, W.M.; Thanbichler, M.; Weibel, D.B. DCAP: A Broad-Spectrum Antibiotic That Targets the Cytoplasmic Membrane of Bacteria. J. Am. Chem. Soc. 2012, 134, 11322–11325. [Google Scholar] [CrossRef] [Green Version]
  23. Racine, E.; Gualtieri, M. From Worms to Drug Candidate: The Story of Odilorhabdins, a New Class of Antimicrobial Agents. Front. Microbiol. 2019, 10, 2893. [Google Scholar] [CrossRef] [Green Version]
  24. Žalubovskis, R.; Winum, J.-Y. Inhibitors of Selected Bacterial Metalloenzymes. Curr. Med. Chem. 2019, 26, 2690–2714. [Google Scholar] [CrossRef]
  25. Kaur, M.; Bansal, G. Antimicrobial Potential of Benzimidazole Derived Molecules. Mini-Rev. Med. Chem. 2019, 19, 624–646. [Google Scholar] [CrossRef]
  26. Turner, R.J. Metal-based antimicrobial strategies. Microb. Biotechnol. 2017, 10, 1062–1065. [Google Scholar] [CrossRef]
  27. Lemire, J.A.; Harrison, J.J.; Turner, R.J. Antimicrobial activity of metals: Mechanisms, molecular targets and applications. Nat. Rev. Microbiol. 2013, 11, 371–384. [Google Scholar] [CrossRef]
  28. Bagińska, N.; Pichlak, A.; Górski, A.; Jończyk-Matysiak, E. Specific and Selective Bacteriophages in the Fight against Multidrug-resistant Acinetobacter baumannii. Virol. Sin. 2019, 34, 347–357. [Google Scholar] [CrossRef]
  29. Torres-Barceló, C. The disparate effects of bacteriophages on antibiotic-resistant bacteria. Emerg. Microbes Infect. 2018, 7, 1–12. [Google Scholar] [CrossRef]
  30. Brogden, N.K.; Brogden, K.A. Will new generations of modified antimicrobial peptides improve their potential as pharmaceuticals? Int. J. Antimicrob. Agents 2011, 38, 217–225. [Google Scholar] [CrossRef] [Green Version]
  31. Tan, J.; Tay, J.; Hedrick, J.; Yang, Y.Y. Synthetic macromolecules as therapeutics that overcome resistance in cancer and microbial infection. Biomaterials 2020, 252, 120078. [Google Scholar] [CrossRef]
  32. Dutta, P.; Das, S. Mammalian Antimicrobial Peptides: Promising Therapeutic Targets against Infection and Chronic Inflammation. Curr. Top. Med. Chem. 2016, 16, 99–129. [Google Scholar] [CrossRef]
  33. Hiemstra, P.S.; Amatngalim, G.; van der Does, A.M.; Taube, C. Antimicrobial Peptides and Innate Lung Defenses. Chest 2016, 149, 545–551. [Google Scholar] [CrossRef]
  34. Shagaghi, N.; Palombo, E.A.; Clayton, A.H.A.; Bhave, M. Archetypal tryptophan-rich antimicrobial peptides: Properties and applications. World J. Microbiol. Biotechnol. 2016, 32, 1–10. [Google Scholar] [CrossRef]
  35. Melo, M.N.; Ferre, R.; Castanho, M.A.R.B. Antimicrobial peptides: Linking partition, activity and high membrane-bound concentrations. Nat. Rev. Genet. 2009, 7, 245–250. [Google Scholar] [CrossRef]
  36. Ahmed, T.; Hammami, R. Recent insights into structure-function relationships of antimicrobial peptides. J. Food Biochem. 2019, 43, e12546. [Google Scholar] [CrossRef] [Green Version]
  37. Alfei, S.; Schito, A.M. Positively Charged Polymers as Promising Devices against Multidrug Resistant Gram-Negative Bacteria: A Review. Polymers 2020, 12, 1195. [Google Scholar] [CrossRef]
  38. Jiang, Y.; Chen, Y.; Song, Z.; Tan, Z.; Cheng, J. Recent advances in design of antimicrobial peptides and polypeptides toward clinical translation. Adv. Drug Deliv. Rev. 2021, 170, 261–280. [Google Scholar] [CrossRef]
  39. Soblosky, L.; Ramamoorthy, A.; Chen, Z. Membrane interaction of antimicrobial peptides using E. coli lipid extract as model bacterial cell membranes and SFG spectroscopy. Chem. Phys. Lipids 2015, 187, 20–33. [Google Scholar] [CrossRef] [Green Version]
  40. Kamimori, H.; Blazyk, J.; Aguilar, M.-I. Lipid Membrane-Binding Properties of Tryptophan Analogues of Linear Amphipathic BETA-Sheet Cationic Antimicrobial Peptides Using Surface Plasmon Resonance. Biol. Pharm. Bull. 2005, 28, 148–150. [Google Scholar] [CrossRef] [Green Version]
  41. Maget-Dana, R. The monolayer technique: A potent tool for studying the interfacial properties of antimicrobial and membrane-lytic peptides and their interactions with lipid membranes. Biochim. Biophys. Acta (BBA)-Biomembr. 1999, 1462, 109–140. [Google Scholar] [CrossRef] [Green Version]
  42. Wang, C.; Chen, T.; Zhang, N.; Yang, M.; Li, B.; Lü, X.; Cao, X.; Ling, C. Melittin, a Major Component of Bee Venom, Sensitizes Human Hepatocellular Carcinoma Cells to Tumor Necrosis Factor-related Apoptosis-inducing Ligand (TRAIL)-induced Apoptosis by Activating CaMKII-TAK1-JNK/p38 and Inhibiting IκBα Kinase-NFκB. J. Biol. Chem. 2009, 284, 3804–3813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Shang, D.; Zhang, Q.; Dong, W.; Liang, H.; Bi, X. The effects of LPS on the activity of Trp-containing antimicrobial peptides against Gram-negative bacteria and endotoxin neutralization. Acta Biomater. 2016, 33, 153–165. [Google Scholar] [CrossRef] [PubMed]
  44. Nan, Y.H.; Bang, J.-K.; Jacob, B.; Park, I.-S.; Shin, S.Y. Prokaryotic selectivity and LPS-neutralizing activity of short antimicrobial peptides designed from the human antimicrobial peptide LL-37. Peptides 2012, 35, 239–247. [Google Scholar] [CrossRef] [PubMed]
  45. Giuliani, A.; Pirri, G.; Rinaldi, A.C. Antimicrobial peptides: The LPS connection. Methods Mol. Biol. 2010, 618, 137–154. [Google Scholar] [CrossRef] [PubMed]
  46. Malanovic, N.; Leber, R.; Schmuck, M.; Kriechbaum, M.; Cordfunke, R.A.; Drijfhout, J.W.; de Breij, A.; Nibbering, P.H.; Kolb, D.; Lohner, K. Phospholipid-driven differences determine the action of the synthetic antimicrobial peptide OP-145 on Gram-positive bacterial and mammalian membrane model systems. Biochim. Biophys. Acta (BBA)-Biomembr. 2015, 1848, 2437–2447. [Google Scholar] [CrossRef] [Green Version]
  47. Ruan, Y.; Shen, T.; Wang, Y.; Hou, M.; Li, J.; Sun, T. Antimicrobial peptide LL-37 attenuates LTA induced inflammatory effect in macrophages. Int. Immunopharmacol. 2013, 15, 575–580. [Google Scholar] [CrossRef]
  48. Matsuzaki, K. Why and how are peptide–lipid interactions utilized for self-defense? Magainins and tachyplesins as archetypes. Biochim. Biophys. Acta (BBA)-Biomembr. 1999, 1462, 1–10. [Google Scholar] [CrossRef] [Green Version]
  49. Chen, L.; Harrison, S. Cell-penetrating peptides in drug development: Enabling intracellular targets. Biochem. Soc. Trans. 2007, 35, 821–825. [Google Scholar] [CrossRef]
  50. Mookherjee, N.; Lippert, D.N.D.; Hamill, P.; Falsafi, R.; Nijnik, A.; Kindrachuk, J.; Pistolic, J.; Gardy, J.; Miri, P.; Naseer, M.; et al. Intracellular Receptor for Human Host Defense Peptide LL-37 in Monocytes. J. Immunol. 2009, 183, 2688–2696. [Google Scholar] [CrossRef] [Green Version]
  51. Bechinger, B.; Gorr, S.-U. Antimicrobial Peptides: Mechanisms of Action and Resistance. J. Dent. Res. 2017, 96, 254–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Palermo, E.F.; Vemparala, S.; Kuroda, K. Antimicrobial polymers: Molecular design as synthetic mimics of host-defense pep-tides. ACS Symp. Ser. 2013, 1135, 319–330. [Google Scholar] [CrossRef] [Green Version]
  53. Li, P.; Zhou, C.; Rayatpisheh, S.; Ye, K.; Poon, Y.F.; Hammond, P.T.; Duan, H.; Chan-Park, M.B. Cationic Peptidopolysaccharides Show Excellent Broad-Spectrum Antimicrobial Activities and High Selectivity. Adv. Mater. 2012, 24, 4130–4137. [Google Scholar] [CrossRef] [PubMed]
  54. Palermo, E.F.; Kuroda, K. Structural determinants of antimicrobial activity in polymers which mimic host defense peptides. Appl. Microbiol. Biotechnol. 2010, 87, 1605–1615. [Google Scholar] [CrossRef] [PubMed]
  55. Rahman, A.; Jui, M.S.; Bam, M.; Cha, Y.; Luat, E.; Alabresm, A.; Nagarkatti, M.; Decho, A.W.; Tang, C. Facial Amphiphilicity-Induced Polymer Nanostructures for Antimicrobial Applications. ACS Appl. Mater. Interfaces 2020, 12, 21221–21230. [Google Scholar] [CrossRef] [PubMed]
  56. Lu, C.; Quan, G.; Su, M.; Nimmagadda, A.; Chen, W.; Pan, M.; Teng, P.; Yu, F.; Liu, X.; Jiang, L.; et al. Molecular Architecture and Charging Effects Enhance the In Vitro and In Vivo Performance of Multi-Arm Antimicrobial Agents Based on Star-Shaped Poly(l -lysine). Adv. Ther. 2019, 2, 1900147. [Google Scholar] [CrossRef]
  57. Wang, J.; Lu, C.; Shi, Y.; Feng, X.; Wu, B.; Zhou, G.; Quan, G.; Pan, X.; Cai, J.; Wu, C. Structural Superiority of Guanidinium-Rich, Four-Armed Copolypeptides: Role of Multiple Peptide–Membrane Interactions in Enhancing Bacterial Membrane Perturbation and Permeability. ACS Appl. Mater. Interfaces 2020, 12, 18363–18374. [Google Scholar] [CrossRef]
  58. Lam, S.J.; O’Brien-Simpson, N.M.; Pantarat, N.; Sulistio, A.; Wong, E.H.H.; Chen, Y.Y.; Lenzo, J.C.; Holden, J.A.; Blencowe, A.; Reynolds, E.C.; et al. Combating multidrug-resistant Gram-negative bacteria with structurally nanoengineered antimicrobial peptide polymers. Nat. Microbiol. 2016, 1, 16162. [Google Scholar] [CrossRef]
  59. Findlay, B.; Zhanel, G.G.; Schweizer, F. Cationic Amphiphiles, a New Generation of Antimicrobials Inspired by the Natural Antimicrobial Peptide Scaffold. Antimicrob. Agents Chemother. 2010, 54, 4049–4058. [Google Scholar] [CrossRef] [Green Version]
  60. Hancock, R.E. Cationic peptides: Effectors in innate immunity and novel antimicrobials. Lancet Infect. Dis. 2001, 1, 156–164. [Google Scholar] [CrossRef]
  61. Marr, A.K.; Gooderham, W.J.; Hancock, R.E.W. Antibacterial peptides for therapeutic use: Obstacles and realistic outlook. Curr. Opin. Pharmacol. 2006, 6, 468–472. [Google Scholar] [CrossRef] [PubMed]
  62. Greco, I.; Molchanova, N.; Holmedal, E.; Jenssen, H.; Hummel, B.D.; Watts, J.L.; Håkansson, J.; Hansen, P.R.; Svenson, J. Correlation between hemolytic activity, cytotoxicity and systemic in vivo toxicity of synthetic antimicrobial peptides. Sci. Rep. 2020, 10, 13206. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, Y.; Guarnieri, M.T.; Vasil, A.I.; Vasil, M.L.; Mant, C.T.; Hodges, R.S. Role of Peptide Hydrophobicity in the Mechanism of Action of α-Helical Antimicrobial Peptides. Antimicrob. Agents Chemother. 2007, 51, 1398–1406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Hong, J.; Oren, Z.; Shai, Y. Structure and Organization of Hemolytic and Nonhemolytic Diastereomers of Antimicrobial Peptides in Membranes. Biochemistry 1999, 38, 16963–16973. [Google Scholar] [CrossRef] [PubMed]
  65. Thomas, H.; Coley, H.M. Overcoming Multidrug Resistance in Cancer: An Update on the Clinical Strategy of Inhibiting P-Glycoprotein. Cancer Control 2003, 10, 159–165. [Google Scholar] [CrossRef] [PubMed]
  66. Alfarouk, K.O.; Stock, C.-M.; Taylor, S.; Walsh, M.; Muddathir, A.K.; Verduzco, D.; Bashir, A.H.H.; Mohammed, O.Y.; O ElHassan, G.; Harguindey, S.; et al. Resistance to cancer chemotherapy: Failure in drug response from ADME to P-gp. Cancer Cell Int. 2015, 15, 71. [Google Scholar] [CrossRef] [Green Version]
  67. Gong, J.; Jaiswal, R.; Mathys, J.-M.; Combes, V.; Grau, G.E.R.; Bebawy, M. Microparticles and their emerging role in cancer multidrug resistance. Cancer Treat. Rev. 2012, 38, 226–234. [Google Scholar] [CrossRef]
  68. Monteleone, L.; Speciale, A.; Valenti, G.; Traverso, N.; Ravera, S.; Garbarino, O.; Leardi, R.; Farinini, E.; Roveri, A.; Ursini, F.; et al. PKCα Inhibition as a Strategy to Sensitize Neuroblastoma Stem Cells to Etoposide by Stimulating Ferroptosis. Antioxidants 2021, 10, 691. [Google Scholar] [CrossRef]
  69. Marengo, B.; Garbarino, O.; Speciale, A.; Monteleone, L.; Traverso, N.; Domenicotti, C. MYC Expression and Metabolic Redox Changes in Cancer Cells: A Synergy Able to Induce Chemoresistance. Oxidative Med. Cell. Longev. 2019, 2019, 7346492. [Google Scholar] [CrossRef] [Green Version]
  70. Marengo, B.; Monti, P.; Miele, M.; Menichini, P.; Ottaggio, L.; Foggetti, G.; Pulliero, A.; Izzotti, A.; Speciale, A.; Garbarino, O.; et al. Etoposide-resistance in a neuroblastoma model cell line is associated with 13q14.3 mono-allelic deletion and miRNA-15a/16-1 down-regulation. Sci. Rep. 2018, 8, 13762. [Google Scholar] [CrossRef]
  71. Colla, R.; Izzotti, A.; De Ciucis, C.; Fenoglio, D.; Ravera, S.; Speciale, A.; Ricciarelli, R.; Furfaro, A.L.; Pulliero, A.; Passalacqua, M.; et al. Glutathione-mediated antioxidant response and aerobic metabolism: Two crucial factors involved in determining the multi-drug resistance of high-risk neuroblastoma. Oncotarget 2016, 7, 70715–70737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Marengo, B.; Nitti, M.; Furfaro, A.L.; Colla, R.; De Ciucis, C.; Marinari, U.M.; Pronzato, M.A.; Traverso, N.; Domenicotti, C. Redox Homeostasis and Cellular Antioxidant Systems: Crucial Players in Cancer Growth and Therapy. Oxid. Med. Cell. Longev. 2016, 2016, 13762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Traverso, N.; Ricciarelli, R.; Nitti, M.; Marengo, B.; Furfaro, A.L.; Pronzato, M.A.; Marinari, U.M.; Domenicotti, C. Role of Glutathione in Cancer Progression and Chemoresistance. Oxidative Med. Cell. Longev. 2013, 2013, 972913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhang, M.; Liu, E.; Cui, Y.; Huang, Y. Nanotechnology-based combination therapy for overcoming multidrug-resistant cancer. Cancer Biol. Med. 2017, 14, 212–227. [Google Scholar] [CrossRef] [Green Version]
  75. Leuschner, C. Membrane Disrupting Lytic Peptides for Cancer Treatments. Curr. Pharm. Des. 2004, 10, 2299–2310. [Google Scholar] [CrossRef]
  76. Lin, L.; Chi, J.; Yan, Y.; Luo, R.; Feng, X.; Zheng, Y.; Xian, D.; Li, X.; Quan, G.; Liu, D.; et al. Membrane-disruptive peptides/peptidomimetics-based therapeutics: Promising systems to combat bacteria and cancer in the drug-resistant era. Acta Pharm. Sin. B 2021, 11, 2609–2644. [Google Scholar] [CrossRef]
  77. Deslouches, B.; Di, Y.P. Antimicrobial peptides with selective antitumor mechanisms: Prospect for anticancer applications. Oncotarget 2017, 8, 46635–46651. [Google Scholar] [CrossRef] [Green Version]
  78. Qiao, X.; Wang, Y.; Yu, H. Progress in the mechanisms of anticancer peptides. Chin. J. Biotechnol. 2019, 35, 1391–1400. [Google Scholar] [CrossRef]
  79. Dennison, S.R.; Whittaker, M.; Harris, F.; Phoenix, D.A. Anticancer α-Helical Peptides and Structure / Function Relationships Underpinning Their Interactions with Tumour Cell Membranes. Curr. Protein Pept. Sci. 2006, 7, 487–499. [Google Scholar] [CrossRef]
  80. Mader, J.S.; Hoskin, D.W. Cationic antimicrobial peptides as novel cytotoxic agents for cancer treatment. Expert Opin. Investig. Drugs 2006, 15, 933–946. [Google Scholar] [CrossRef]
  81. Takahashi, H.; Yumoto, K.; Yasuhara, K.; Nadres, E.T.; Kikuchi, Y.; Buttitta, L.; Taichman, R.S.; Kuroda, K. Anticancer polymers designed for killing dormant prostate cancer cells. Sci. Rep. 2019, 9, 1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Gaspar, D.; Veiga, A.S.; Castanho, M.A.R.B. From antimicrobial to anticancer peptides. A review. Front. Microbiol. 2013, 4, 294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Sheu, M.J.; Baldwin, W.W.; Brunson, K.W. Cytotoxicity of rabbit macrophage peptides MCP-1 and MCP-2 for mouse tumor cells. Antimicrob. Agents Chemother. 1985, 28, 626–629. [Google Scholar] [CrossRef] [Green Version]
  84. Utsugi, T.; Schroit, A.J.; Connor, J.; Bucana, C.D.; Fidler, I.J. Elevated expression of phosphatidylserine in the outer membrane leaflet of human tumor cells and recognition by activated human blood monocytes. Cancer Res. 1991, 51, 3062–3066. [Google Scholar] [PubMed]
  85. Gerber, D.E.; Hao, G.; Watkins, L.; Stafford, J.H.; Anderson, J.; Holbein, B.; Oz, O.K.; Mathews, D.; Thorpe, P.E.; Hassan, G.; et al. Tumor-specific targeting by Bavituximab, a phosphatidylserine-targeting monoclonal an-tibody with vascular targeting and immune modulating properties, in lung cancer xenografts. Am. J. Nucl. Med. Mol. Imaging 2015, 5, 493–503. [Google Scholar] [PubMed]
  86. Kenis, H.; Reutelingsperger, C. Targeting phosphatidylserine in anti-cancer therapy. Curr. Pharm. Des. 2009, 15, 2719–2723. [Google Scholar] [CrossRef]
  87. Papo, N.; Shai, Y. Host defense peptides as new weapons in cancer treatment. Cell. Mol. Life Sci. 2005, 62, 784–790. [Google Scholar] [CrossRef]
  88. Sok, M.; Šentjurc, M.; Schara, M.; Stare, J.; Rott, T. Cell membrane fluidity and prognosis of lung cancer. Ann. Thorac. Surg. 2002, 73, 1567–1571. [Google Scholar] [CrossRef]
  89. Izzi, V.; Heljasvaara, R.; Pihlajaniemi, T. Understanding the extracellular matrix in acute myeloid leukemia. Haematologica 2017, 102, 1807–1809. [Google Scholar] [CrossRef]
  90. Li, X.; Shen, B.; Chen, Q.; Zhang, X.; Ye, Y.; Wang, F.; Zhang, X. Antitumor effects of cecropin B-LHRH’ on drug-resistant ovarian and endometrial cancer cells. BMC Cancer 2016, 16, 251. [Google Scholar] [CrossRef] [Green Version]
  91. Risso, A.; Zanetti, M.; Gennaro, R. Cytotoxicity and Apoptosis Mediated by Two Peptides of Innate Immunity. Cell. Immunol. 1998, 189, 107–115. [Google Scholar] [CrossRef] [PubMed]
  92. Hsu, J.-C.; Lin, L.-C.; Tzen, J.T.; Chen, J.-Y. Pardaxin-induced apoptosis enhances antitumor activity in HeLa cells. Peptides 2011, 32, 1110–1116. [Google Scholar] [CrossRef] [PubMed]
  93. Kim, S.; Kim, S.S.; Bang, Y.-J.; Kim, S.-J.; Lee, B.J. In vitro activities of native and designed peptide antibiotics against drug sensitive and resistant tumor cell lines. Peptides 2003, 24, 945–953. [Google Scholar] [CrossRef]
  94. Kawamoto, M.; Horibe, T.; Kohno, M.; Kawakami, K. A novel transferrin receptor-targeted hybrid peptide disintegrates cancer cell membrane to induce rapid killing of cancer cells. BMC Cancer 2011, 11, 359. [Google Scholar] [CrossRef] [Green Version]
  95. Hilchie, A.L.; Doucette, C.D.; Pinto, D.M.; Patrzykat, A.; Douglas, S.; Hoskin, D.W. Pleurocidin-family cationic antimicrobial peptides are cytolytic for breast carcinoma cells and prevent growth of tumor xenografts. Breast Cancer Res. 2011, 13, R102. [Google Scholar] [CrossRef] [Green Version]
  96. Chen, Y.-L.S.; Li, J.-H.; Yu, C.-Y.; Lin, C.-J.; Chiu, P.-H.; Chen, P.-W.; Lin, C.-C.; Chen, W.-J. Novel cationic antimicrobial peptide GW-H1 induced caspase-dependent apoptosis of hepatocellular carcinoma cell lines. Peptides 2012, 36, 257–265. [Google Scholar] [CrossRef]
  97. Li, C.; Liu, H.; Yang, Y.; Xu, X.; Lv, T.; Zhang, H.; Liu, K.; Zhang, S.; Chen, Y. N-myristoylation of Antimicrobial Peptide CM4 Enhances Its Anticancer Activity by Interacting with Cell Membrane and Targeting Mitochondria in Breast Cancer Cells. Front. Pharmacol. 2018, 9, 1297. [Google Scholar] [CrossRef] [Green Version]
  98. Feliu, L.; Oliveras, G.; Cirac, A.D.; Besalú, E.; Rosés, C.; Colomer, R.; Bardají, E.; Planas, M.; Puig, T. Antimicrobial cyclic decapeptides with anticancer activity. Peptides 2010, 31, 2017–2026. [Google Scholar] [CrossRef]
  99. Zhou, H.; Forveille, S.; Sauvat, A.; Yamazaki, T.; Senovilla, L.; Ma, Y.; Liu, P.; Yang, H.; Bezu, L.; Müller, K.; et al. The oncolytic peptide LTX-315 triggers immunogenic cell death. Cell Death Dis. 2016, 7, e2134. [Google Scholar] [CrossRef]
  100. Kuriyama, I.; Miyazaki, A.; Tsuda, Y.; Yoshida, H.; Mizushina, Y. Inhibitory effect of novel somatostatin peptide analogues on human cancer cell growth based on the selective inhibition of DNA polymerase β. Bioorganic Med. Chem. 2013, 21, 403–411. [Google Scholar] [CrossRef]
  101. Koskimaki, J.E.; Karagiannis, E.D.; Tang, B.C.; Hammers, H.; Watkins, D.N.; Pili, R.; Popel, A.S. Pentastatin-1, a collagen IV derived 20-mer peptide, suppresses tumor growth in a small cell lung cancer xenograft model. BMC Cancer 2010, 10, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Papo, N.; Seger, D.; Makovitzki, A.; Kalchenko, V.; Eshhar, Z.; Degani, H.; Shai, Y. Inhibition of Tumor Growth and Elimination of Multiple Metastases in Human Prostate and Breast Xenografts by Systemic Inoculation of a Host Defense–Like Lytic Peptide. Cancer Res. 2006, 66, 5371–5378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Camilio, K.A.; Berge, G.; Ravuri, C.S.; Rekdal, O.; Sveinbjørnsson, B. Complete regression and systemic protective immune responses obtained in B16 melanomas after treatment with LTX-315. Cancer Immunol. Immunother. 2014, 63, 601–613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Deslouches, B.; Gonzalez, I.A.; DeAlmeida, D.; Islam, K.; Steele, C.; Montelaro, R.C.; Mietzner, T.A. De novo-derived cationic antimicrobial peptide activity in a murine model of Pseudomonas aeruginosa bacteraemia. J. Antimicrob. Chemother. 2007, 60, 669–672. [Google Scholar] [CrossRef]
  105. Deslouches, B.; Islam, K.; Craigo, J.K.; Paranjape, S.M.; Montelaro, R.C.; Mietzner, T.A. Activity of the De Novo Engineered Antimicrobial Peptide WLBU2 against Pseudomonas aeruginosa in Human Serum and Whole Blood: Implications for Systemic Applications. Antimicrob. Agents Chemother. 2005, 49, 3208–3216. [Google Scholar] [CrossRef] [Green Version]
  106. Makobongo, M.O.; Gancz, H.; Carpenter, B.M.; McDaniel, D.P.; Merrell, D.S. The Oligo-Acyl Lysyl Antimicrobial Peptide C 12 K-2β 12 Exhibits a Dual Mechanism of Action and Demonstrates Strong In Vivo Efficacy against Helicobacter pylori. Antimicrob. Agents Chemother. 2012, 56, 378–390. [Google Scholar] [CrossRef] [Green Version]
  107. Desbois, A.P.; Gemmell, C.G.; Coote, P.J. In vivo efficacy of the antimicrobial peptide ranalexin in combination with the endopeptidase lysostaphin against wound and systemic meticillin-resistant Staphylococcus aureus (MRSA) infections. Int. J. Antimicrob. Agents 2010, 35, 559–565. [Google Scholar] [CrossRef] [Green Version]
  108. Narayana, J.L.; Huang, H.-N.; Wu, C.-J.; Chen, J.-Y. Efficacy of the antimicrobial peptide TP4 against Helicobacter pylori infection: In vitro membrane perturbation via micellization and in vivo suppression of host immune responses in a mouse model. Oncotarget 2015, 6, 12936–12954. [Google Scholar] [CrossRef] [Green Version]
  109. Braunstein, A.; Papo, N.; Shai, Y. In Vitro Activity and Potency of an Intravenously Injected Antimicrobial Peptide and Its dl Amino Acid Analog in Mice Infected with Bacteria. Antimicrob. Agents Chemother. 2004, 48, 3127–3129. [Google Scholar] [CrossRef] [Green Version]
  110. Spinks, C.B.; Zidan, A.S.; Khan, M.; Habib, M.J.; Faustino, P.J. Pharmaceutical characterization of novel tenofovir liposomal formulations for enhanced oral drug delivery: In vitro pharmaceutics and Caco-2 permeability investigations. Clin. Pharmacol. Adv. Appl. 2017, 9, 29–38. [Google Scholar] [CrossRef] [Green Version]
  111. Dawidczyk, C.M.; Russell, L.M.; Hultz, M.; Searson, P.C. Tumor accumulation of liposomal doxorubicin in three murine models: Optimizing delivery efficiency. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1637–1644. [Google Scholar] [CrossRef] [PubMed]
  112. Cern, A.; Marcus, D.; Tropsha, A.; Barenholz, Y.; Goldblum, A. New drug candidates for liposomal delivery identified by computer modeling of liposomes’ remote loading and leakage. J. Control. Release 2017, 252, 18–27. [Google Scholar] [CrossRef] [PubMed]
  113. Abu Lila, A.S.; Ishida, T. Liposomal Delivery Systems: Design Optimization and Current Applications. Biol. Pharm. Bull. 2017, 40, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Li, H.; Yuan, D.; Sun, M.; Ping, Q. Effect of ligand density and PEG modification on octreotide-targeted liposome via somatostatin receptor in vitro and in vivo. Drug Deliv. 2016, 23, 3562–3572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Pan, X.; Xu, J.; Jia, X. Research Progress Evaluating the Function and Mechanism of Anti-Tumor Peptides. Cancer Manag. Res. 2020, 12, 397–409. [Google Scholar] [CrossRef] [Green Version]
  116. Li, Y.; Xiang, Q.; Zhang, Q.; Huang, Y.; Su, Z. Overview on the recent study of antimicrobial peptides: Origins, functions, relative mechanisms and application. Peptides 2012, 37, 207–215. [Google Scholar] [CrossRef]
  117. Meng, S.S. Research progresses of antimicrobial peptides with anticancer activities. Chin. J. Biochem. Pharm. 2011, 32, 77–80. [Google Scholar]
  118. Kim, M.K.; Kang, N.H.; Ko, S.J.; Park, J.; Park, E.; Shin, D.W.; Kim, S.H.; Lee, S.; Lee, J.I.; Ha, E.G.; et al. Antibacterial and Antibiofilm Activity and Mode of Action of Magainin 2 against Drug-Resistant Acinetobacter baumannii. Int. J. Mol. Sci. 2018, 19, 3041. [Google Scholar] [CrossRef] [Green Version]
  119. Dennison, S.; Harris, F.; Phoenix, D.A. The interactions of aurein 1.2 with cancer cell membranes. Biophys. Chem. 2007, 127, 78–83. [Google Scholar] [CrossRef]
  120. Wang, C.; Dong, S.; Zhang, L.; Zhao, Y.; Huang, L.; Gong, X.; Wang, H.; Shang, D. Cell surface binding, uptaking and anti-cancer activity of L-K6, a lysine/ leucine-rich peptide, on human breast cancer MCF-7 cells. Sci. Rep. 2017, 7, 8293. [Google Scholar] [CrossRef]
  121. Ren, S.X.; Shen, J.; Cheng, A.; Lu, L.; Chan, R.L.Y.; Li, Z.J.; Wang, X.J.; Wong, C.C.M.; Zhang, L.; Ng, S.S.M.; et al. FK-16 Derived from the Anticancer Peptide LL-37 Induces Caspase-Independent Apoptosis and Autophagic Cell Death in Colon Cancer Cells. PLoS ONE 2013, 8, e63641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Bellamy, W.; Takase, M.; Yamauchi, K.; Wakabayashi, H.; Kawase, K.; Tomita, M. Identification of the bactericidal domain of lactoferrin. Biochim. Biophys. Acta (BBA)-Protein Struct. Mol. Enzym. 1992, 1121, 130–136. [Google Scholar] [CrossRef]
  123. Yang, X.; Shi, W.R.; Yang, W.J.; He, T.; Hu, Y.S.; Wang, S.S.; Wei, W.M.; Qin, Y.D. Lactoferrin peptides inhibit the proliferation of gastric cancer cells and induce apoptosis in vitro. Acta Univ. Med. Anhui 2017, 52, 781–785. [Google Scholar] [CrossRef]
  124. Hilchie, A.L.; Vale, R.; Zemlak, T.S.; Hoskin, D.W. Generation of a hematologic malignancy-selective membranolytic peptide from the antimicrobial core (RRWQWR) of bovine lactoferricin. Exp. Mol. Pathol. 2013, 95, 192–198. [Google Scholar] [CrossRef] [PubMed]
  125. Meng, L.; Xu, G.; Li, J.; Liu, W.; Jia, W.; Ma, J.; Wei, D. Bovine lactoferricin P13 triggers ROS-mediated caspase-dependent apoptosis in SMMC7721 cells. Oncol. Lett. 2017, 13, 511–517. [Google Scholar] [CrossRef] [Green Version]
  126. Gaspar, D.; Freire, J.M.; Pacheco, T.R.; Barata, J.T.; Castanho, M.A. Apoptotic human neutrophil peptide-1 anti-tumor activity revealed by cellular biomechanics. Biochim. Biophys. Acta 2015, 1853, 308–316. [Google Scholar] [CrossRef] [Green Version]
  127. Veldhuizen, E.J.A.; Schneider, V.A.F.; Agustiandari, H.; Van Dijk, A.; Bokhoven, J.L.M.T.-V.; Bikker, F.; Haagsman, H.P. Antimicrobial and Immunomodulatory Activities of PR-39 Derived Peptides. PLoS ONE 2014, 9, e95939. [Google Scholar] [CrossRef]
  128. Bae, S.; Oh, K.; Kim, H.; Kim, Y.; Kim, H.R.; Hwang, Y.I.; Lee, D.S.; Kang, J.S.; Lee, W.J. The effect of alloferon on the en-hancement of NK cell cytotoxicity against cancer via the up-regulation of perforin/granzyme B secretion. Immunobiology 2013, 218, 1026–1033. [Google Scholar] [CrossRef]
  129. Ryu, M.-J.; Anikin, V.; Hong, S.-H.; Jeon, H.; Yu, Y.G.; Yu, M.-H.; Chernysh, S.; Lee, C. Activation of NF-κB by alloferon through down-regulation of antioxidant proteins and IκBα. Mol. Cell. Biochem. 2008, 313, 91–102. [Google Scholar] [CrossRef]
  130. Wang, H.M.; Li, Y.Y.; Hou, J.C.; Jiang, C.G.; Li, D.F.; Lu, J.; Zhao, Y.H.; Feng, S.Y. Biological activity and stability of new peptide derivative KW-WK from Bovine lactoferricin. Food Sci. 2018, 39, 57–62. [Google Scholar] [CrossRef]
  131. Jeon, H.; Le, M.T.; Ahn, B.; Cho, H.-S.; Le, V.C.Q.; Yum, J.; Hong, K.; Kim, J.-H.; Song, H.; Park, C. Copy number variation of PR-39 cathelicidin, and identification of PR-35, a natural variant of PR-39 with reduced mammalian cytotoxicity. Gene 2019, 692, 88–93. [Google Scholar] [CrossRef]
  132. Hu, E.; Wang, D.; Chen, J.; Tao, X. Novel cyclotides from Hedyotis diffusa induce apoptosis and inhibit proliferation and migration of prostate cancer cells. Int. J. Clin. Exp. Med. 2015, 8, 4059–4065. [Google Scholar] [PubMed]
  133. Zhang, G.; Liu, S.; Liu, Y.; Wang, F.; Ren, J.; Gu, J.; Zhou, K.; Shan, B. A novel cyclic pentapeptide, H-10, inhibits B16 cancer cell growth and induces cell apoptosis. Oncol. Lett. 2014, 8, 248–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Wang, Y.; Guo, D.; He, J.; Song, L.; Chen, H.; Zhang, Z.; Tan, N. Inhibition of fatty acid synthesis arrests colorectal neoplasm growth and metastasis: Anti-cancer therapeutical effects of natural cyclopeptide RA-XII. Biochem. Biophys. Res. Commun. 2019, 512, 819–824. [Google Scholar] [CrossRef] [PubMed]
  135. Kollár, P.; Rajchard, J.; Balounová, Z.; Pazourek, J. Marine natural products: Bryostatins in preclinical and clinical studies. Pharm. Biol. 2014, 52, 237–242. [Google Scholar] [CrossRef] [Green Version]
  136. Wali, A.F.; Majid, S.; Rasool, S.; Shehada, S.B.; Abdulkareem, S.K.; Firdous, A.; Beigh, S.; Shakeel, S.; Mushtaq, S.; Akbar, I.; et al. Natural products against cancer: Review on phytochemicals from marine sources in preventing cancer. Saudi Pharm. J. 2019, 27, 767–777. [Google Scholar] [CrossRef]
  137. Kitagaki, J.; Shi, G.; Miyauchi, S.; Murakami, S.; Yang, Y. Cyclic depsipeptides as potential cancer therapeutics. Anti-Cancer Drugs 2015, 26, 259–271. [Google Scholar] [CrossRef]
  138. Ehrenstein, G.; Lecar, H. Electrically gated ionic channels in lipid bilayers. Q. Rev. Biophys. 1977, 10, 1–34. [Google Scholar] [CrossRef]
  139. Pouny, Y.; Rapaport, D.; Mor, A.; Nicolas, P.; Shai, Y. Interaction of antimicrobial dermaseptin and its fluorescently labeled analogs with phospholipid membranes. Biochemistry 1992, 31, 12416–12423. [Google Scholar] [CrossRef]
  140. Hao, X.; Yan, Q.; Zhao, J.; Wang, W.; Huang, Y.; Chen, Y. TAT modification of α-helical anticancer peptides to improve specificity and efficacy. PLoS ONE 2015, 10, e0138911. [Google Scholar] [CrossRef] [Green Version]
  141. Diao, Y.; Han, W.; Zhao, H.; Zhu, S.; Liu, X.; Feng, X.; Gu, J.; Yao, C.; Liu, S.; Sun, C.; et al. Designed synthetic analogs of the α-helical peptide temporin-La with improved antitumor efficacies via charge modification and incorporation of the integrin αvβ3 homing domain. J. Pept. Sci. 2012, 18, 476–486. [Google Scholar] [CrossRef] [PubMed]
  142. Yi, Z.-F.; Cho, S.-G.; Zhao, H.; Wu, Y.-Y.; Luo, J.; Li, D.; Yi, T.; Xu, X.; Wu, Z.; Liu, M. A novel peptide from human apolipoprotein(a) inhibits angiogenesis and tumor growth by targeting c-Src phosphorylation in VEGF-induced human umbilical endothelial cells. Int. J. Cancer 2009, 124, 843–852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jang, J.-P.; Jung, H.J.; Han, J.M.; Jung, N.; Kim, Y.; Kwon, H.J.; Ko, S.-K.; Soung, N.-K.; Jang, J.-H.; Ahn, J.S. Two cyclic hexapeptides from Penicillium sp. FN070315 with antiangiogenic activities. PLoS ONE 2017, 12, e0184339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Wang, C.; Chen, Y.W.; Zhang, L.; Gong, X.G.; Zhou, Y.; Shang, D.J. Melanoma cell surface-expressed phosphatidylserine as a therapeutic target for cationic anticancer peptide, temporin-1CEa. J. Drug Target 2016, 24, 548–556. [Google Scholar] [CrossRef]
  145. Zhang, Y.; Nicolau, A.; Lima, C.F.; Rodrigues, L.R. Bovine Lactoferrin Induces Cell Cycle Arrest and Inhibits Mtor Signaling in Breast Cancer Cells. Nutr. Cancer 2014, 66, 1371–1385. [Google Scholar] [CrossRef]
  146. Wolf, J.S.; Li, G.; Varadhachary, A.; Petrak, K.; Schneyer, M.; Li, D.; Ongkasuwan, J.; Zhang, X.; Taylor, R.J.; Strome, S.E.; et al. Oral Lactoferrin Results in T Cell–Dependent Tumor Inhibition of Head and Neck Squamous Cell Carcinoma In vivo. Clin. Cancer Res. 2007, 13, 1601–1610. [Google Scholar] [CrossRef] [Green Version]
  147. Li, X.; Meng, Y.; Plotnikoff, N.P.; Youkilis, G.; Griffin, N.; Wang, E.; Lu, C.; Shan, F. Methionine enkephalin (MENK) inhibits tumor growth through regulating CD4+Foxp3+ regulatory T cells (Tregs) in mice. Cancer Biol. Ther. 2015, 16, 450–459. [Google Scholar] [CrossRef] [Green Version]
  148. Zhao, D.; Plotnikoff, N.; Griffin, N.; Song, T.; Shan, F. Methionine enkephalin, its role in immunoregulation and cancer therapy. Int. Immunopharmacol. 2016, 37, 59–64. [Google Scholar] [CrossRef]
  149. Zagon, I.S.; Donahue, R.N.; McLaughlin, P.J. Opioid growth factor-opioid growth factor receptor axis is a physiological de-terminant of cell proliferation in diverse human cancers. Am. J. Physiol. 2009, 297, R1154–R1161. [Google Scholar] [CrossRef]
  150. Teixeira, M.C.; Carbone, C.; Sousa, M.C.; Espina, M.; Garcia, M.L.; Sanchez-Lopez, E.; Souto, E.B. Nanomedicines for the Delivery of Antimicrobial Peptides (AMPs). Nanomaterials 2020, 10, 560. [Google Scholar] [CrossRef] [Green Version]
  151. Wu, Q.; Feng, D.; Huang, Z.; Chen, M.; Yang, D.; Pan, X.; Lu, C.; Quan, G.; Wu, C. Supersaturable organic-inorganic hybrid matrix based on well-ordered mesoporous silica to improve the bioavailability of water insoluble drugs. Drug Deliv. 2020, 27, 1292–1300. [Google Scholar] [CrossRef] [PubMed]
  152. Zhou, L.; Qiu, T.; Lv, F.; Liu, L.; Ying, J.; Wang, S. Self-Assembled Nanomedicines for Anticancer and Antibacterial Applications. Adv. Health Mater. 2018, 7, e1800670. [Google Scholar] [CrossRef] [PubMed]
  153. Alfei, S.; Marengo, B.; Zuccari, G. Nanotechnology application in food packaging: A plethora of opportunities versus pending risks assessment and public concerns. Food Res. Int. 2020, 137, 109664. [Google Scholar] [CrossRef] [PubMed]
  154. Chagri, S.; Ng, D.Y.W.; Weil, T. Designing bioresponsive nanomaterials for intracellular self-assembly. Nat. Rev. Chem. 2022, 6, 320–338. [Google Scholar] [CrossRef]
  155. Baptista, P.V.; McCusker, M.P.; Carvalho, A.; Ferreira, D.A.; Mohan, N.M.; Martins, M.; Fernandes, A.R. Nano-Strategies to Fight Multidrug Resistant Bacteria—A Battle of the Titans. Front. Microbiol. 2018, 9, 1441. [Google Scholar] [CrossRef] [Green Version]
  156. Gounani, Z.; Asadollahi, M.A.; Meyer, R.L.; Arpanaei, A. Loading of polymyxin B onto anionic mesoporous silica nanoparticles retains antibacterial activity and enhances biocompatibility. Int. J. Pharm. 2018, 537, 148–161. [Google Scholar] [CrossRef]
  157. Gao, Y.; Wang, J.; Chai, M.; Li, X.; Deng, Y.; Jin, Q.; Ji, J. Size and Charge Adaptive Clustered Nanoparticles Targeting the Biofilm Microenvironment for Chronic Lung Infection Management. ACS Nano 2020, 14, 5686–5699. [Google Scholar] [CrossRef]
  158. Mozhi, A.; Ahmad, I.; Okeke, C.I.; Li, C.; Liang, X.J. pH-sensitive polymeric micelles for the co-delivery of proapoptotic peptide and anticancer drug for synergistic cancer therapy. RSC Adv. 2017, 7, 12886–12896. [Google Scholar] [CrossRef] [Green Version]
  159. Li, Y.; Xu, N.; Zhu, W.; Wang, L.; Liu, B.; Zhang, J.; Xie, Z.; Liu, W. Nanoscale Melittin@Zeolitic Imidazolate Frameworks for Enhanced Anticancer Activity and Mechanism Analysis. ACS Appl. Mater. Interfaces 2018, 10, 22974–22984. [Google Scholar] [CrossRef]
  160. Moffatt, J.H.; Harper, M.; Boyce, J.D. Mechanisms of Polymyxin Resistance. Adv. Exp. Med. Biol. 2019, 1145, 55–71. [Google Scholar] [CrossRef]
  161. Nikaido, H. Molecular Basis of Bacterial Outer Membrane Permeability Revisited. Microbiol. Mol. Biol. Rev. 2003, 67, 593–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Epand, R.M.; Walker, C.; Epand, R.F.; Magarvey, N.A. Molecular mechanisms of membrane targeting antibiotics. Biochim. Biophys. Acta 2016, 1858, 980–987. [Google Scholar] [CrossRef] [PubMed]
  163. Alfei, S.; Schito, A.M. From Nanobiotechnology, Positively Charged Biomimetic Dendrimers as Novel Antibacterial Agents: A Review. Nanomaterials 2020, 10, 2022. [Google Scholar] [CrossRef] [PubMed]
  164. Elouahabi, A.; Ruysschaert, J.-M. Formation and Intracellular Trafficking of Lipoplexes and Polyplexes. Mol. Ther. 2005, 11, 336–347. [Google Scholar] [CrossRef] [PubMed]
  165. Ramos, J.; Forcada, J.; Hidalgo-Alvarez, R. Cationic Polymer Nanoparticles and Nanogels: From Synthesis to Biotechnological Applications. Chem. Rev. 2013, 114, 367–428. [Google Scholar] [CrossRef] [PubMed]
  166. Jenjob, R.; Phakkeeree, T.; Seidi, F.; Theerasilp, M.; Crespy, D. Emulsion Techniques for the Production of Pharmacological Nanoparticles. Macromol. Biosci. 2019, 19, e1900063. [Google Scholar] [CrossRef]
  167. Matsuoka, K.; Nakatani, Y.; Yoshimura, T.; Akasaki, T. Superoxide Scavenging Activity of Gold, Silver, and Platinum Nanoparticles Capped with Sugar-based Nonionic Surfactants. J. Oleo Sci. 2019, 68, 847–854. [Google Scholar] [CrossRef] [Green Version]
  168. Chakraborty, M.; Hsiao, F.-W.; Naskar, B.; Chang, C.-H.; Panda, A.K. Surfactant-Assisted Synthesis and Characterization of Stable Silver Bromide Nanoparticles in Aqueous Media. Langmuir 2012, 28, 7282–7290. [Google Scholar] [CrossRef]
  169. Hou, Z.; Shankar, Y.V.; Liu, Y.; Ding, F.; Subramanion, J.L.; Ravikumar, V.; Zamudio-Vázquez, R.; Keogh, D.; Lim, H.; Tay, M.Y.F.; et al. Nanoparticles of Short Cationic Peptidopolysaccharide Self-Assembled by Hydrogen Bonding with Antibacterial Effect against Multidrug-Resistant Bacteria. ACS Appl. Mater. Interfaces 2017, 9, 38288–38303. [Google Scholar] [CrossRef]
  170. Huma, Z.-E.; Gupta, A.; Javed, I.; Das, R.; Hussain, S.Z.; Mumtaz, S.; Hussain, I.; Rotello, V.M. Cationic Silver Nanoclusters as Potent Antimicrobials against Multidrug-Resistant Bacteria. ACS Omega 2018, 3, 16721–16727. [Google Scholar] [CrossRef] [Green Version]
  171. Chen, M.; Long, Z.; Dong, R.; Wang, L.; Zhang, J.; Li, S.; Zhao, X.; Hou, X.; Shao, H.; Jiang, X. Titanium Incorporation into Zr-Porphyrinic Metal–Organic Frameworks with Enhanced Antibacterial Activity against Multidrug-Resistant Pathogens. Small 2020, 16, e1906240. [Google Scholar] [CrossRef] [PubMed]
  172. Schito, A.M.; Alfei, S. Antibacterial Activity of Non-Cytotoxic, Amino Acid-Modified Polycationic Dendrimers against Pseudomonas aeruginosa and Other Non-Fermenting Gram-Negative Bacteria. Polymers 2020, 12, 1818. [Google Scholar] [CrossRef] [PubMed]
  173. Alfei, S.; Caviglia, D.; Piatti, G.; Zuccari, G.; Schito, A. Bactericidal Activity of a Self-Biodegradable Lysine-Containing Dendrimer against Clinical Isolates of Acinetobacter Genus. Int. J. Mol. Sci. 2021, 22, 7274. [Google Scholar] [CrossRef] [PubMed]
  174. Schito, A.M.; Piatti, G.; Caviglia, D.; Zuccari, G.; Zorzoli, A.; Marimpietri, D.; Alfei, S. Bactericidal Activity of Non-Cytotoxic Cationic Nanoparticles against Clinically and Environmentally Relevant Pseudomonas spp. Isolates. Pharmaceutics 2021, 13, 1411. [Google Scholar] [CrossRef] [PubMed]
  175. Si, Z.; Hou, Z.; Vikhe, Y.S.; Thappeta, K.R.V.; Marimuthu, K.; De, P.P.; Ng, O.T.; Li, P.; Zhu, Y.; Pethe, K.; et al. Antimicrobial Effect of a Novel Chitosan Derivative and Its Synergistic Effect with Antibiotics. ACS Appl. Mater. Interfaces 2021, 13, 3237–3245. [Google Scholar] [CrossRef] [PubMed]
  176. Gupta, D.; Singh, A.; Khan, A.U. Nanoparticles as Efflux Pump and Biofilm Inhibitor to Rejuvenate Bactericidal Effect of Conventional Antibiotics. Nanoscale Res. Lett. 2017, 12, 454. [Google Scholar] [CrossRef] [PubMed]
  177. Alfei, S.; Schito, A.M.; Zuccari, G. Considerable Improvement of Ursolic Acid Water Solubility by Its Encapsulation in Dendrimer Nanoparticles: Design, Synthesis and Physicochemical Characterization. Nanomaterials 2021, 11, 2196. [Google Scholar] [CrossRef]
  178. Alfei, S.; Brullo, C.; Caviglia, D.; Zuccari, G. Preparation and Physicochemical Characterization of Water-Soluble Pyrazole-Based Nanoparticles by Dendrimer Encapsulation of an Insoluble Bioactive Pyrazole Derivative. Nanomaterials 2021, 11, 2662. [Google Scholar] [CrossRef]
  179. Alfei, S.; Spallarossa, A.; Lusardi, M.; Zuccari, G. Successful Dendrimer and Liposome-Based Strategies to Solubilize an Antiproliferative Pyrazole Otherwise Not Clinically Applicable. Nanomaterials 2022, 12, 233. [Google Scholar] [CrossRef]
  180. Alfei, S.; Brullo, C.; Caviglia, D.; Piatti, G.; Zorzoli, A.; Marimpietri, D.; Zuccari, G.; Schito, A.M. Pyrazole-Based Water-Soluble Dendrimer Nanoparticles as a Potential New Agent against Staphylococci. Biomedicines 2022, 10, 17. [Google Scholar] [CrossRef]
  181. Schito, A.M.; Caviglia, D.; Piatti, G.; Zorzoli, A.; Marimpietri, D.; Zuccari, G.; Schito, G.C.; Alfei, S. Efficacy of Ursolic Acid-Enriched Water-Soluble and Not Cytotoxic Nanoparticles against Enterococci. Pharmaceutics 2021, 13, 1976. [Google Scholar] [CrossRef] [PubMed]
  182. Bilensoy, E. Cationic nanoparticles for cancer therapy. Expert Opin. Drug Deliv. 2010, 7, 795–809. [Google Scholar] [CrossRef] [PubMed]
  183. Zhang, J.; Song, J.; Liang, X.; Yin, Y.; Zuo, T.; Chen, D.; Shen, Q. Hyaluronic acid-modified cationic nanoparticles overcome enzyme CYP1B1-mediated breast cancer multidrug resistance. Nanomedicine 2019, 14, 447–464. [Google Scholar] [CrossRef] [PubMed]
  184. Alfei, S.; Marengo, B.; Valenti, G.; Domenicotti, C. Synthesis of Polystyrene-Based Cationic Nanomaterials with Pro-Oxidant Cytotoxic Activity on Etoposide-Resistant Neuroblastoma Cells. Nanomaterials 2021, 11, 977. [Google Scholar] [CrossRef]
  185. Alfei, S.; Piatti, G.; Caviglia, D.; Schito, A. Synthesis, Characterization, and Bactericidal Activity of a 4-Ammoniumbuthylstyrene-Based Random Copolymer. Polymers 2021, 13, 1140. [Google Scholar] [CrossRef]
  186. Schito, A.; Piatti, G.; Caviglia, D.; Zuccari, G.; Alfei, S. Broad-Spectrum Bactericidal Activity of a Synthetic Random Copolymer Based on 2-Methoxy-6-(4-Vinylbenzyloxy)-Benzylammonium Hydrochloride. Int. J. Mol. Sci. 2021, 22, 5021. [Google Scholar] [CrossRef]
Figure 1. Comparison among membrane lipid composition of healthy mammalian cells, Gram-positive and Gram-negative bacteria.
Figure 1. Comparison among membrane lipid composition of healthy mammalian cells, Gram-positive and Gram-negative bacteria.
Ijms 23 06108 g001
Figure 2. Comparison among membrane lipid composition of healthy mammalian cells and cancer cells.
Figure 2. Comparison among membrane lipid composition of healthy mammalian cells and cancer cells.
Ijms 23 06108 g002
Figure 3. Models of ACPs’ action. In the carpet model (left), ACPs bind to the cell membrane via electrostatic interactions and, subsequently, ACPs may enter the cell membrane, inducing membrane disruption. In the barrel-stave model (right), peptides self-aggregate and form a transmembrane pore, leading to membrane depolarization.
Figure 3. Models of ACPs’ action. In the carpet model (left), ACPs bind to the cell membrane via electrostatic interactions and, subsequently, ACPs may enter the cell membrane, inducing membrane disruption. In the barrel-stave model (right), peptides self-aggregate and form a transmembrane pore, leading to membrane depolarization.
Ijms 23 06108 g003
Figure 4. NPs’ interaction with cell membrane and their killing ability on cancer cells and bacteria.
Figure 4. NPs’ interaction with cell membrane and their killing ability on cancer cells and bacteria.
Ijms 23 06108 g004
Table 1. ACPs showing antitumor and/or antiviral effects, classified according to their structural characteristics into four categories: α-helical, β-pleated sheets, random coil and cyclic [116,117].
Table 1. ACPs showing antitumor and/or antiviral effects, classified according to their structural characteristics into four categories: α-helical, β-pleated sheets, random coil and cyclic [116,117].
CategoryFeatureACPModel
α-Helical
[118,119,120,121]
Peptides short in length
Simple in structure
Magainin IILung cancer cells
(A549)
AureinGlioblastoma cells
(T98G)
L-K6Breast cancer cells
(MCF-7)
LL37Colorectal cancer cells (HCT116)
FK-16
β-Pleated sheet
[122,123,124,125,126]
Two or more disulfide bonds Good stability
Structures more complex than α-helical
Bovine lactoferrin (LfcinB)Gastric cancer cells
(MGC803)
MPLfcinB6Human T leukemia cells
MPLfcin-P13Hepatocellular carcinoma cells (HepG2)
Human neutrophil peptide
(HNP-1)
Prostate cancer cells
(PC-3)
Random coil ACPs
[127,128,129,130,131]
Rich in proline and glycine
Lack of a typical secondary structure
AlloferonHerpes simplex virus
Human papillomavirus
KW-WKN.R.
PR39N.R.
PR35N.R.
Cyclic ACPs
[132,133,134]
Closed peptides composed of a head-to-tail cyclization backbone or disulfide bonds that form cystine knots
More stable than linear structures
Hedyotis diffusa Cytide 1–3Prostate cancer cells
(PC3, DU145, LNCap)
H-10Malignant melanoma cells (B16)
RA-XIIColorectal tumor cells (HCT116)
N.R. = not reported.
Table 2. ACPs in clinical trials ongoing or already approved and marketed.
Table 2. ACPs in clinical trials ongoing or already approved and marketed.
ACPs in DevelopmentPhase of DevelopmentTarget TumorAdvantages
Bryostatin 1
[135,136]
Phase 1Melanoma
Lymphoma
Ovarian carcinoma
Aplidine (plitidepsin)
[137]
Phase 1 (completed) Well tolerated
Low toxicity
Phase 2 (in progress)Advanced medullary thyroid carcinoma
Advanced malignant melanoma
Small-cell lung cancer
Advanced renal carcinoma
Kyprolis [115]Approved by FDA and EMA (marketed)N.R.N.S.
SomaKit TOC [115]
Lutathera [115]
Gallium Dotatoc Ga68 [115]
N.R. = not reported; N.S. = not specified.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Valenti, G.E.; Alfei, S.; Caviglia, D.; Domenicotti, C.; Marengo, B. Antimicrobial Peptides and Cationic Nanoparticles: A Broad-Spectrum Weapon to Fight Multi-Drug Resistance Not Only in Bacteria. Int. J. Mol. Sci. 2022, 23, 6108. https://doi.org/10.3390/ijms23116108

AMA Style

Valenti GE, Alfei S, Caviglia D, Domenicotti C, Marengo B. Antimicrobial Peptides and Cationic Nanoparticles: A Broad-Spectrum Weapon to Fight Multi-Drug Resistance Not Only in Bacteria. International Journal of Molecular Sciences. 2022; 23(11):6108. https://doi.org/10.3390/ijms23116108

Chicago/Turabian Style

Valenti, Giulia E., Silvana Alfei, Debora Caviglia, Cinzia Domenicotti, and Barbara Marengo. 2022. "Antimicrobial Peptides and Cationic Nanoparticles: A Broad-Spectrum Weapon to Fight Multi-Drug Resistance Not Only in Bacteria" International Journal of Molecular Sciences 23, no. 11: 6108. https://doi.org/10.3390/ijms23116108

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop