Next Article in Journal
A Retrospective Analysis of Intravenous Push versus Extended Infusion Meropenem in Critically Ill Patients
Previous Article in Journal
Real-World Clinical Characteristics and Outcomes with Daptomycin Use in Pediatric Patients: A Retrospective Case Series
Previous Article in Special Issue
Bacteriophage Therapy in Companion and Farm Animals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Innovative Strategies in Drug Repurposing to Tackle Intracellular Bacterial Pathogens

by
Blanca Lorente-Torres
1,
Jesús Llano-Verdeja
1,
Pablo Castañera
1,
Helena Á. Ferrero
1,
Sergio Fernández-Martínez
1,
Farzaneh Javadimarand
1,
Luis M. Mateos
1,2,
Michal Letek
1,3,* and
Álvaro Mourenza
1,*
1
Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
2
Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
3
Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, 24071 León, Spain
*
Authors to whom correspondence should be addressed.
Antibiotics 2024, 13(9), 834; https://doi.org/10.3390/antibiotics13090834
Submission received: 28 July 2024 / Revised: 26 August 2024 / Accepted: 29 August 2024 / Published: 2 September 2024

Abstract

:
Intracellular bacterial pathogens pose significant public health challenges due to their ability to evade immune defenses and conventional antibiotics. Drug repurposing has recently been explored as a strategy to discover new therapeutic uses for established drugs to combat these infections. Utilizing high-throughput screening, bioinformatics, and systems biology, several existing drugs have been identified with potential efficacy against intracellular bacteria. For instance, neuroleptic agents like thioridazine and antipsychotic drugs such as chlorpromazine have shown effectiveness against Staphylococcus aureus and Listeria monocytogenes. Furthermore, anticancer drugs including tamoxifen and imatinib have been repurposed to induce autophagy and inhibit bacterial growth within host cells. Statins and anti-inflammatory drugs have also demonstrated the ability to enhance host immune responses against Mycobacterium tuberculosis. The review highlights the complex mechanisms these pathogens use to resist conventional treatments, showcases successful examples of drug repurposing, and discusses the methodologies used to identify and validate these drugs. Overall, drug repurposing offers a promising approach for developing new treatments for bacterial infections, addressing the urgent need for effective antimicrobial therapies.

1. Introduction

The global burden of infectious diseases caused by intracellular pathogens remains a significant challenge to public health [1]. Pathogens such as Listeria monocytogenes, Mycobacterium tuberculosis, and Staphylococcus aureus have developed sophisticated mechanisms to survive and replicate within host cells [2,3], evading both the host immune response and traditional antimicrobial therapies. This evasion complicates treatment strategies and underscores the urgent need for innovative approaches to combat these infections [4].
Drug repurposing, the process of identifying new therapeutic uses for existing drugs, typically FDA-approved with established information on their toxicity, formulation, pharmacology, and potential side effects [5,6], has emerged as a promising strategy to address this challenge (Figure 1), especially pushed by the vast amount of information surrounding approved drugs [7]. In that sense, the emergence of COVID-19 has accelerated the application of drug repurposing strategies to find new and effective treatments for combating diseases [8,9]. Drug repurposing had primarily been accidental, lacking a systematic approach; however, the discovery of Viagra as an effective treatment for erectile dysfunction, instead of its original purpose as an antihypertensive drug, opened the door to a new and lucrative market, which was further propelled by the COVID-19 pandemic [10,11]. Repurposing offers a cost-effective and time-efficient alternative to the lengthy and expensive process of novel drug discovery [12] that reduces the investment needed for the development of new treatments and makes repurposing an attractive alternative for pharmaceutical companies [13]. Recent advances in high-throughput screening, bioinformatics, and systems biology have facilitated the identification of candidate drugs with potential efficacy against intracellular pathogens [14,15,16].
It is crucial to acknowledge that drug repurposing faces several significant challenges, with intellectual property barriers being foremost among them. From a profitability perspective, repurposed drugs often present difficulties, as generating profit can be challenging [10]. However, it is possible to secure patents and achieve profitability for a repurposed drug when a novel medical use is demonstrated. This requires successfully completing clinical trials to confirm the drug’s efficacy in its new application [10].
This review aims to provide a comprehensive overview of the current landscape of drug repurposing efforts targeting intracellular bacterial pathogens, which is an evolving field (Figure 2). We will discuss the mechanisms by which these pathogens evade conventional therapies, highlight key examples of successfully repurposed drugs, and explore the methodological approaches employed in identifying and validating these candidates.

2. Staphylococcus aureus

Staphylococcus aureus is a significant human pathogen responsible for a variety of infections. Approximately 30% of the human population is estimated to carry S. aureus, and it is the leading cause of bacteremia [17,18]. This pathogen expresses numerous virulence factors that aid in the colonization of eukaryotic cells. Attempts to develop vaccines targeting various S. aureus virulence factors have failed due to the bacterium’s ability to evade the immune response from multiple angles, rendering these vaccines ineffective [19]. Moreover, the misuse of antimicrobials has led to the emergence of multi-drug resistant bacteria [20]. Methicillin-resistant S. aureus (MRSA), in particular, was responsible for approximately 100,000 deaths related to antimicrobial resistance in 2019 [18]. The rapid evolution of bacteria poses significant challenges for developing new antimicrobials, making it often economically inefficient. Consequently, drug repurposing has emerged as a viable alternative [21]. This strategy has shown promise, particularly in the treatment of S. aureus infections.
S. aureus initially infects the skin and mucosal surfaces, and can eventually infect internal organs, causing pneumonia, endocarditis, osteomyelitis, and bacteremia [19]. More interestingly, S. aureus can act as both an extracellular and an intracellular pathogen, which significantly increases the difficulty of treating S. aureus infections. Once inside the cells, S. aureus can evade the phagosome and become free in the cytoplasm. Additionally, it controls the host adaptive immune response through a complex system of virulence factors expressed during infection [19]. For these reasons, S. aureus is a model for targeting host-directed therapies (HDT), as reactivating the immune system could be an effective strategy to kill intracellular bacteria.
A plethora of drugs target intracellular S. aureus, and interestingly, some have been extensively used for other purposes and are now repurposed to target intracellular infections (Table 1). Chlorpromazine (CPZ), originally used to treat psychosis, was the first drug discovered with anti-S. aureus activity among other intracellular pathogens [22]. It was demonstrated that the drug accumulates in macrophages, particularly in lysosomes, at higher concentrations than needed for its in vitro activity. The fusion between the phagosome (where the bacteria are usually located) and the lysosome triggers the antimicrobial activity of CPZ. However, the doses required for in vivo activity are beyond those clinically achievable, and CPZ becomes toxic when used chronically. Fortunately, another neuroleptic agent, thioridazine (TZ), has shown antimicrobial activity at clinically relevant doses with a similar mechanism of action as CPZ [22,23]. Thioridazine, a phenothiazine, exhibits activity in vitro against S. aureus and also acts against phagocytosed S. aureus by accumulating in macrophages [22,24].
Antimicrobials active against cell walls or cell wall synthesis are commonly used. Clomiphene, a fertility drug, antagonizes wall teichoic acid (WTA), showing activity against S. aureus and synergism with cell-wall-targeting antimicrobials [25].
Repurposing antimicrobial agents against new targets is another strategy to utilize existing compounds. Some agents are used alone or in combination with other antimicrobials to mitigate resistance. Interestingly, ebselen, a non-toxic seleno-organic drug with anti-inflammatory, antioxidant, and anti-atherosclerotic properties, has demonstrated activity against MRSA [26]. Additionally, while ebselen does not show activity against Gram-negative bacteria, it is effective against intracellular MRSA, highlighting its potential as a new anti-MRSA therapy [27].
The anthelmintic drug selamectin showed a Gram-positive antimicrobial profile with high activity against S. aureus, a low toxicity profile, and an 81.3% reduction in intracellular S. aureus loading, exhibiting a synergistic effect with ampicillin [28].
Celecoxib, an anti-inflammatory drug, inhibits cyclooxygenase-2 (COX-2) and also inhibits multidrug efflux pumps in S. aureus, increasing sensitivity to other antimicrobials. However, the antimicrobial mechanism of celecoxib remains unknown [29]. Diflunisal, another anti-inflammatory drug and a prostaglandin synthetase inhibitor, acts on the Agr quorum-sensing system, which plays a key role in pathological bone remodeling in S. aureus osteomyelitis [30]. Inactivation of the Agr system leads to significant bone destruction reduction in osteomyelitis patients [30].
Repurposing anticancer drugs as antimicrobials has several advantages, primarily due to their established safety spectrum and guaranteed intracellular activity. However, repurposing drugs from cancer to antibacterial therapies shares a common handicap: the appearance of resistance in both prokaryotic and eukaryotic cells [31]. Floxuridine, an inhibitor of riboside phosphorylase, and streptozotocin, a DNA synthesis inhibitor, were both repurposed to treat intracellular S. aureus. These compounds inhibit the SaeRS two-component system and demonstrated a significant survival rate in S. aureus-infected mice compared to the commonly used anthracycline antibiotic doxorubicin [32].
As an intracellular pathogen, S. aureus is exposed to reactive oxygen and nitrogen species (RONS), which usually reduce its viability [33]. Staphylotoxin, the golden carotenoid pigment of S. aureus, aids in ROS detoxification during infection. Interestingly, BPH-652, a cholesterol-lowering agent, binds to the dehydrosqualene synthase of S. aureus, reducing staphylotoxin and restoring the toxic effect of ROS against S. aureus [34]. Artemisinin, extensively used as an antimalarial drug, exhibits antibacterial activity via ROS generation; the addition of Cu+ ions enhances its activity through increased free radical production and DNA damage [35].
Ebselen, which shows non-selective activity against different cysteine-containing proteins, specifically inhibits thioredoxin reductase (TrxR) and thioredoxin (Trx) in S. aureus, resulting in toxic bactericidal activity against MRSA [36]. Similarly, auranofin decreases the reducing capacity of MRSA by inhibiting the TrxR system, making the bacteria more sensitive to oxidative stress [37]. Auranofin, an FDA-approved organogold drug, has proven effective in vivo against MRSA and intracellular M. tuberculosis at low micromolar concentrations [37]. It clears over 60% of intracellular S. aureus, demonstrating significantly higher efficacy compared to isoniazid and vancomycin, which only achieve a 30% clearance of intracellular bacteria [38].
Another strategy to fight intracellular pathogens is the induction of autophagy in infected cells, a treatment known as host-directed therapy [15,16]. Several compounds exhibit this activity. Ibrutinib, used for lymphocytic leukemia, decreases intracellular Mycobacterium tuberculosis and S. aureus, reducing MRSA viability by 90%. Ibrutinib inhibits the BTK/Akt/mTOR pathway during M. tuberculosis infections, triggering autophagy in macrophages [39]. Ibrutinib’s activity was further analyzed with other host-directed therapy compounds like dasatinib and crizotinib against S. aureus, acting as kinase inhibitors and blocking bacterial infection and intracellular proliferation [15]. Interestingly, crizotinib targets ATP production and presents broad-spectrum activity against Gram-positive bacteria, showing a synergistic effect with clindamycin and gentamicin [15,40].
Raloxifene, an estrogenic receptor agonist, has been repurposed against intracellular S. aureus due to its autophagy-inducing activity [41]. Raloxifene also prevents neutrophil cell death in response to phorbol 12-myristate 13-acetate (PMA), inhibiting PMA-induced ERK phosphorylation but not ROS production, which triggers the formation of neutrophil extracellular traps (NETs), known to contain antimicrobial substances aiding in intracellular pathogen clearance [41,42].
During infection, MRSA activates the MAPK/ERK pathway, which acts as a sensor for cell energy homeostasis and amino acid deprivation. Intriguingly, S. aureus appears capable of inducing autophagy to facilitate its own replication and proliferation [43]. Building on this hypothesis, dorsomorphin, an AMPK inhibitor, has shown activity against S. aureus as a host-directed therapy. It reduces autophagy and thereby decreases S. aureus intracellular survival [43]. Similarly, thapsigargin, an inhibitor of the SERCA protein (a Ca2+ pump in the endoplasmic reticulum), blocks autophagy, increases host cell viability, and reduces intracellular MRSA survival by two-fold [16].
Table 1. List of repurposed drugs against S. aureus: classification and mechanism of action.
Table 1. List of repurposed drugs against S. aureus: classification and mechanism of action.
DrugTypeMechanism of ActionReference
ArtemisininAntimalarialGenerates ROS and DNA damage[35]
AuranofinAnti-inflammatoryInhibits thioredoxin reductase[37]
BPH-652Cholesterol-loweringBinds dehydrosqualene synthase (CrtM)[34]
CelecoxibAnti-inflammatoryInhibits multidrug efflux pumps[29]
ChlorpromazineAntipsychoticAccumulates in lysosomes, triggering antimicrobial activity[22]
ClomipheneFertilityAntagonizes wall teichoic acid[25]
CrizotinibAntitumoralATP production[5,28]
DasatinibAntitumoralTyrosine kinase inhibitor[15]
DiflunisalAnti-inflammatoryInhibits the Agr system[30]
DorsomorphinAMPK inhibitorReduces autophagy and intracellular survival[43]
EbselenAnti-inflammatoryInhibits thioredoxin reductase and thioredoxin[26]
FloxuridineAntitumoralInhibits riboside phosphorylase[32]
IbrutinibAntitumoralControls the MEK/ERK/c-JUN signaling pathway[15]
Phenothiazine AntipsychoticAntagonism of dopamine D2 receptors[11,13]
RaloxifeneEstrogen receptor agonistInduces autophagy and inhibits neutrophil cell death[41]
SelamectinAnthelminticShows high activity against S. aureus and M. tuberculosis[28,41]
StreptozotocinAntitumoralDNA synthesis inhibitor[28,32]
ThapsigarginCa2+ endoplasmic pump inhibitorIncreases host cell viability and reduces intracellular survival[16,32]
ThioridazineAntipsychoticAccumulates in macrophages, triggering antimicrobial activity[16,22,23]

3. Mycobacterium tuberculosis

M. tuberculosis is the bacterium responsible for tuberculosis (TB) and is considered the leading cause of death due to bacterial infections [14,44]. M. tuberculosis is adept at colonizing and surviving inside alveolar macrophages, modulating immune responses, and controlling macrophage maturation. Its metabolism is adapted to the challenging environment it encounters during infection. Recent reviews have focused on drug repurposing for M. tuberculosis treatment, specifically on drugs that can cross eukaryotic membranes to target intracellular M. tuberculosis [12,14,26]. One of the most prominent strategies includes host-directed therapies (Table 2) that have been extensively used against M. tuberculosis [12,14,45,46,47].
During infection, M. tuberculosis controls lysosome protease fusion proteins, downregulating them to aid colonization. Saquinavir, an anti-HIV drug, has been proposed as a repurposed drug against M. tuberculosis because it can restore cathepsin protease activity, including cathepsin S, in a dose-dependent manner in M. tuberculosis-infected macrophages [48]. Additionally, M. tuberculosis utilizes cholesterol, a crucial component of mammalian cell membranes, as an energy source essential for colonization. Cholesterol accumulation decreases membrane permeability to anti-tubercular drugs and blocks phagosome maturation by direct cholesterol uptake [49]. Consequently, statins, which treat hypercholesterolemia and atherosclerotic cardiovascular disease, have emerged as new anti-tubercular drugs. Atorvastatin, a 3-hydroxy-3-methyl-glutaryl-coenzume A (HMG-CoA) reductase inhibitor, enhances macrophage bactericidal effects [50]. Atorvastatin started a phase 2 clinical trial (ref. NCT06199921) this year. Simvastatin, another HMG-CoA inhibitor, reduces cholesterol uptake by M. tuberculosis, sensitizing the bacteria to other antimicrobial compounds and showing synergy with isoniazid and rifampicin [51].
Inhibiting efflux pumps is another strategy to enhance antimicrobial effects. M. tuberculosis detoxifies itself via a bacterial efflux pump-mediated process. Verapamil, an FDA-approved cardiovascular drug, has been successfully used as an adjuvant for TB treatment, acting as a mycobacterial efflux pump inhibitor and increasing the toxicity of isoniazid and rifampicin [52]; this drug was used in a recently finished Phase 1/2 clinical trial (ref. IRCT20170210032478N1). During M. tuberculosis infection, patients often suffer inflammatory responses, leading to tissue damage from neutrophil infiltration. Anti-inflammatory drugs, such as ibuprofen, have shown potential in reducing bacterial load in the lungs and improving survival in animal models, despite having no direct effect on M. tuberculosis [53]. Ibuprofen has progressed to two distinct Phase 2 clinical trials as an adjunctive treatment: one in combination with acetylsalicylic acid (currently ongoing, ref. NCT02781909), and another as a standalone treatment, which was completed in 2019 (ref. NCT03891901).
The activation of Toll-like receptors (TLR) is part of the innate defence against bacterial infections, but M. tuberculosis downregulates TLR activity. Vitamin D, which activates TLR, has shown anti-TB activity, highlighting the importance of sunlight in bacterial infections [54]. Repurposing anticancer drugs is another strategy against M. tuberculosis. Tamoxifen, an established anticancer drug, has been repurposed to induce autophagy in infected cells, promoting bacterial degradation in lysosomes [55]. Fluspirilene and pimozide, two diphenylbutylpiperidine-class antipsychotic drugs, elicit autophagy in infected cells, promoting bacterial clearance. Pimozide also affects ROS generation and inhibits STAT5, reducing the presence of cytokine-inducible SH2-containing protein on infected phagosomes [56].
M. tuberculosis uses ABL and other related host tyrosine kinases for entry and intracellular survival. Imatinib, a tyrosine kinase inhibitor used in cancer therapy, has been successfully tested against M. tuberculosis in vitro and in vivo [57] and it was in a Phase 2 clinical trial testing its safety, pharmacokinetics and hematologic effect alone or in combination with isoniazid and rifabutin, finishing the study in 2022 (ref. NCT03891901). The development of kinase inhibitors has been pursued for years due to their importance in intracellular pathogen colonization. Kuijl et al. (2007) have developed several kinase inhibitors with good antimicrobial profiles against M. tuberculosis and Salmonella enterica serovar Typhimurium [58].
Promoting autophagy in infected cells is a classic method to combat intracellular pathogens. Bazedoxifene, a selective estrogen receptor modulator (SERM) used in cancer therapy, has been repurposed against M. tuberculosis, reducing the intracellular growth of the bacteria in THP-1 macrophages [59]. Vitamin D also shows antimicrobial activity via autophagy induction by activating the human cathelicidin microbial peptide (CAMP) [60]. Vitamin D3 induces antimicrobial activity through autophagy and nitric oxide production, acting through cathelicidin and TLRs, which induce a nitric oxide burst [54,61]. The good activity shown against M. tuberculosis opened the door to three different clinical trials (refs. NCT00507000 and NCT01130311); one of them was finalized in 2023 (ref. NCT01992263).
Nitroimidazofurans, originally used as radiosensitizers in cancer chemotherapy, are bicyclic molecules that show anti-TB activity in the nanomolar range, similar to isoniazid [62]. PA-824 has demonstrated activity in murine models treated orally with the compound, thus PA-824 was used in a Phase 2 clinical trial that finalized in 2022 (ref. NCT02256696). Chronic inflammation, often resulting from infection, aids M. tuberculosis infection [63]. Reducing deleterious inflammation can make current antibiotherapies more effective. Metformin, a biguanide used for type 2 diabetes, has emerged as a new HDT therapy by activating a major energy-sensing kinase and reducing inflammation by limiting the proliferation of inflammatory cells [63]. Metformin was in a Phase 1/2 clinical trial that finalized in 2023 (ref. NCT05215990). Metformin, in combination with statins, has shown synergistic effects [64].
Once M. tuberculosis colonizes pulmonary tissues, it often forms granulomas characterized by hypoxia and necrosis, similar to solid tumors. This induces the production of vascular endothelial growth factor (VEGF), promoting new vessel formation and aiding TB progression. Bevacizumab, a humanized monoclonal antibody used to neutralize VEGF in some cancers, has been used to reduce TB progression and disease lesions in animal models [65].
Reintroducing “old” antimicrobials is an effective strategy against multidrug-resistant bacteria due to their known pharmacokinetics and pharmacodynamics. Sulphonamides have been studied as new TB therapies. Sulfadiazine, an analogue of para-aminobenzoic acid, acts as a dihydropteroate synthase inhibitor, blocking the folic acid synthesis pathway in M. tuberculosis and showing high in vitro activity, suggesting its potential as a second-line antibiotic in combination with other treatments [66].
Targeting cell wall synthesis is a common strategy against bacterial infections. Arabinosyl transferase (EmbC) is crucial for lipoarabinomannan biosynthesis, a major polysaccharide in the cell envelope. Ethambutol, which targets EmbC, is part of TB treatment but often encounters resistance. Terlipressin, originally a vasoactive drug for low blood pressure, has been identified as a new anti-TB drug targeting EmbC in a different region than ethambutol, showing high binding affinity and reduced resistance potential [67]. In silico analysis has identified 29 compounds with anti-TB activity, with fluvastatin showing intracellular activity at moderate concentrations, highlighting the power of bioinformatics in antimicrobial discovery [68].
Anthelmintic drugs like avermectins, used against helminths, insects, and arachnids, have shown activity against mycobacteria. Ivermectin, selamectin, and moxidectin have demonstrated in vitro activity against MDR M. tuberculosis, showing high specificity and oral activity, making them promising antimicrobial therapies [69]. Nitazoxanide, an FDA-approved anthelmintic and antiparasitic drug, inhibits intracellular M. tuberculosis growth by modulating host cell immune responses [70] and it was in a Phase 2 clinical trial to study its bactericidal activity (ref. NCT02684240).
Phenothiazine-derived antipsychotic drugs have shown high antitubercular activity with minimal side effects, showing no cytotoxicity in vitro and in vivo [71]. Carbapenems, such as biapenem, which was involved in a Phase 1 clinical trial (ref. NCT01702649), and tebipenem, have shown activity against M. tuberculosis by targeting L,D-transpeptidases, [72]. Faropenem has shown even better transpeptidase affinity and good in vivo antimicrobial activity [72], results that opened the door to clinical trials; thus, Faropenem was used in a Phase 3 clinical trial (ref. NCT01937832). Carbapenems show synergistic effects with rifampin, with doripenem, biapenem, and rifampin combinations showing the best results [73].
Artemisinin and its derivative artesunate, known for their anti-malarial properties, also show anti-TB activity with confirmed low toxicity and good pharmacokinetic profiles in murine models [74]. Imidazopyridine amides, such as compound Q203, target the cytochrome bc1 complex and have shown effective inhibition of M. tuberculosis in vitro and in mouse models [75]. Moreover, Q203 was utilized in a Phase 2 clinical trial finished in 2019 that evaluated its bactericidal activity, safety, tolerability, and pharmacokinetics following multiple oral doses (ref. NCT03563599).
M. tuberculosis experiences oxidative stress inside macrophages, making its redox mechanisms, including NADH dehydrogenase type II and thioredoxin reductase (TrxR), critical. Chlorpromazine, an NADH inhibitor, and gatifloxacin, a fluoroquinolone, that was in a Phase 3 clinical trial (ref NCT00216385), have shown effective synergistic antimicrobial effects, indicating their potential as dual therapies [76]. Auranofin, a TrxR inhibitor, has shown significant efficacy against both intracellularly replicating and non-replicating M. tuberculosis [37]. In fact, Auranofin was used in a Phase 2 clinical trial (ref. NCT02968927) that finalized in 2020.
Table 2. List of repurposed drugs against M. tuberculosis: classification and mechanism of action.
Table 2. List of repurposed drugs against M. tuberculosis: classification and mechanism of action.
DrugTypeMechanism of ActionReference
ArtemisininAntimalarialGenerates ROS and DNA damage[74]
ArtesunateAntimalarialUnknown[74]
AtorvastatinStatinEnhances macrophage bactericidal effects[50]
AuranofinAnti-inflammatoryInhibits thioredoxin reductase[37]
BazedoxifeneEstrogen receptor modulatorReduces intracellular growth[59]
BevacizumabAntitumoralNeutralizes VEGF[65]
BiapenemAntimicrobialTargets cell wall synthesis[72]
ChlorpromazineAntipsychoticNADH dehydrogenase type II inhibitor[76]
FaropenemAntimicrobialTargets cell wall synthesis[72]
FluspirileneAntipsychoticElicits autophagy[56]
FluvastatinStatinTargets intracellular activity[68]
GatifloxacinAntimicrobialDNA gyrase[76]
IbuprofenAnti-inflammatoryReduces inflammatory response[53]
IbrutinibAntitumoralInhibits the BTK/Akt/mTOR pathway, triggering autophagy[39]
ImatinibAntitumoralInhibits tyrosine kinases[57]
Imidazopyridine amides (Q203)AntimicrobialCytochrome bc1 complex[61]
IvermectinAnthelminticInhibits intracellular growth[69]
MetforminAntidiabeticActivates energy-sensing kinase and reduces inflammation[63]
MoxidectinAnthelminticUnknown mechanism of action[55]
Nitroimidazofuran (PA-824)AntimicrobialBacterial nitroreduction[49]
NitazoxanideAnthelminticModulates host immune responses[70]
PhenothiazineAntipsychoticNADH inhibitor[57,62]
PimozideAntipsychoticAffects ROS generation and inhibits STAT5[56]
SaquinavirAntiviralRestores cathepsin protease activity[48]
SelamectinAnthelminticUnknown mechanism of action[55]
SimvastatinStatinReduces cholesterol uptake, showing synergy with antimicrobials[51]
SulfadiazineAntimicrobialInhibits folic acid synthesis[66]
TamoxifenAntitumoralInduces autophagy[55]
TebipenemAntimicrobialTargets cell wall synthesis[72]
TerlipressinVasoactiveTargets EmbC for cell wall synthesis[67]
VerapamilCardiovascularInhibits bacterial efflux pump[39]
Vitamin DVitaminActivates TLR and induces autophagy[54]

4. Listeria monocytogenes

Listeria monocytogenes (LMO) is a Gram-positive, saprophytic bacterium that can exist both intracellularly and extracellularly, categorizing it as a facultative intracellular pathogen. It is the causative agent of listeriosis, a serious infection typically leading to meningitis, septicemia, and neonatal death [77]. Upon infection, LMO penetrates the intestinal epithelial barrier and migrates to the lamina propria [78]. LMO enters host cells via endocytosis; once inside the phagosome, it escapes into the cytoplasm where it can reside [79]. LMO infection triggers an inflammatory response, apoptosis, necrosis, pyroptosis, and autophagy in the infected cells [78]. As a host response, both major histocompatibility complex (MHC) I and II molecules produce immune responses involving CD4 and CD8 T cells and the production of interferon γ (IFN γ).
Given the high fatality rate of LMO among food-borne pathogens (13%) and a 97.1% hospitalization rate [80], repurposing drugs to combat LMO infections could be a cost-effective strategy. Similar to other intracellular pathogens like S. aureus and M. tuberculosis, LMO can control infection by modulating the host-cell immune response, making host-directed therapies (HDT) potentially effective alone or in combination with antimicrobials for new treatments.
Neurological compounds have shown promise against LMO (Table 3). In a screening of 68 neurological compounds, 26 exhibited inhibitory effects on LMO internalization in eukaryotic cells. Notably, thioridazine (an antipsychotic) and bepridil (a calcium channel inhibitor) inhibited LMO’s escape from the vacuole, resulting in bacterial death [81]. Pimozide, previously reported, blocks LMO entry into host cells, phagosome escape, and cell-to-cell spread, indicating its potential as an effective treatment by targeting multiple stages of infection [82]. However, no in vivo results for pimozide have been reported.
Repurposing antimicrobials is another strategy against LMO (Table 3). Griseofulvin, an antifungal drug, has demonstrated activity against LMO, although it lacks activity against S. aureus or Bacillus subtilis. This discovery has led to modifications of its structure to increase the activity of various derivatives [83]. These compounds preferentially target GyrB, DNA topoisomerase IV, and thymidylate kinase, showing low or negligible cytotoxicity [83]. Despite these efforts, no in vivo studies have been conducted on these compounds, and their stability remains untested.

5. Salmonella enterica Serovar Typhimurium

Salmonella Typhimurium is a Gram-negative bacterium that causes gastroenteritis and severe systemic infections in humans and other animals. It infects both epithelial cells and macrophages. Epithelial cell infection is primarily driven by the Salmonella Pathogenicity Island 1 (SPI-1), whereas infection of macrophages and neutrophils occurs through phagocytosis, leading to the formation of a modified phagosome known as the Salmonella-containing vacuole (SCV) [84]. Within host cells, S. Typhimurium releases various effectors to aid its intracellular survival [85]. Additionally, S. Typhimurium infection alters host cholesterol biosynthesis [86].
The use of host-directed therapies (HDT) is widely applied for Salmonella due to its role as a model for host-pathogen interactions [86] (Table 4). Statins, which affect host cholesterol metabolism, have been shown to negatively impact bacterial growth. For instance, lovastatin blocked intracellular proliferation of S. Typhimurium in macrophages and murine models [86].
Anti-inflammatory drugs are another powerful tool for repurposing against S. Typhimurium as they modulate the host-cell response. Diclofenac sodium, an anti-inflammatory drug that inhibits prostaglandin G/H synthase 1 and 2, exhibited antimicrobial activity against S. Typhimurium and other bacteria both in vitro and in vivo. This compound, alone or in combination with other drugs, showed significant antimicrobial activity against several pathogens, including Shigella spp. and Salmonella spp. Diclofenac sodium demonstrated a synergistic effect with streptomycin and trifluoperazine, reducing mortality in mice from 100% without treatment to less than 10% in treated groups [87].
High-throughput screening (HTS) of various compounds has been a promising method for identifying effective repurposed drugs against S. Typhimurium. Doxapram, amoxapine, and trifluoperazine were identified using this technique. Trifluoperazine, a dopamine D2 receptor blocker used as an antipsychotic, accumulates in macrophages and reduces S. Typhimurium intracellular survival by potentially targeting the host autophagy pathway [88]. In the same line, loperamide, an antidiarrheal drug, blocked the intracellular replication of S. Typhimurium not by directly affecting the bacteria, but by inducing autophagy in S. Typhimurium-infected cells [89].
Bioinformatic tools are also utilized to identify inhibitors of specific bacterial proteins or virulence factors. For instance, Joshi et al. (2022) analyzed over 1900 compounds as potential S. Typhimurium dihydrofolate reductase inhibitors. From this, eight compounds were further analyzed, and four (duvelisib, amenamevir, lifitegrast, and nilotinib) were identified as the most promising inhibitors [90].
HTS of different antimicrobials against intracellular S. Typhimurium yielded interesting results. Since antimicrobials can have different effects in vitro versus during infection, a new technique was developed to evaluate the activity of 1600 antimicrobials against intracellular S. enterica. Nucleoside analogs, including doxifluridine, fluorouracil, azacitidine, and carmofur, were among the most effective. Additionally, bromperidol, metergoline, ciclopirox and ethopropazine showed significant activity at the lowest concentration of 64 µg/mL [84]. Metergoline, the most promising agent, disrupted the cytoplasmic membrane potential decreasing ATP levels. This alkaloid, produced by fungi, was effective in murine models, particularly in the spleen, liver, cecum, and colon [84].
Table 4. List of repurposed drugs against S. enterica: classification and mechanism of action.
Table 4. List of repurposed drugs against S. enterica: classification and mechanism of action.
DrugTypeMechanism of ActionReference
AmoxapineAntidepressantIncrease the levels of norepinephrine and serotonin[74]
AmenamevirAntiviralInhibits helicase-primase complex [75]
AzacitidineAntitumoralDisrupts cytoplasmic membrane potential[84]
BromperidolAntipsychoticDopamine D2 receptor antagonist[70]
CarmofurAntitumoralInhibitor dihydropyrimidine dehydrogenase[84]
CiclopiroxAntifungalInhibits iron-dependent enzymes[70]
Diclofenac sodiumAnti-inflammatoryInhibits prostaglandin G/H synthase[74]
DoxapramRespiratory stimulantStimulates respiratory chemoreceptors[74]
DoxifluridineAntitumoralInhibits thymidylate synthase[70]
DuvelisibAntitumoralInhibits δ and γ isoforms PI3K [75]
EthopropazineAnticholinergicBlocks muscarinic acetylcholine receptors[70]
FluorouracilAntitumoralDisrupts cytoplasmic membrane potential[84]
LifitegrastAnti-inflammatoryIntegrin agonist[75]
LoperamideAntidiarrhealPromotes autophagy [89]
LovastatinAnti-cholesterolInhibits HMG-CoA reductase[72]
MetergolineAntipsychoticSerotonin receptor antagonist[70]
NilotinibAntitumoralInhibits tyrosine kinase[75]
TrifluoperazineAntipsychoticAccumulates in macrophages, targets autophagy pathway[88]

6. Other Intracellular Pathogens

Intracellular pathogens have unique strategies for surviving and proliferating within host cells. For instance, Chlamydia forms inclusions; Legionella and Coxiella occupy lysosomes; and Shigella and Rickettsia inhabit the cytosol [84].
Shigella flexneri, a common gastrointestinal pathogen, has been targeted for drug repurposing. As an intracellular pathogen, S. flexneri is susceptible to drugs that induce autophagy in host cells. Capsaicin, a herbal compound known to induce autophagy, has shown effectiveness in reducing S. flexneri intracellular growth both in vitro and in vivo, with a good safety profile in murine models [91]. Diclofenac, an anti-inflammatory drug, has also demonstrated activity against S. flexneri in murine models, prompting further research that advanced to clinical trials. These efforts culminated in a Phase 4 clinical trial, which revealed diclofenac’s efficacy in women with urinary tract infections (UTIs) who were treated for 30 days. The trial results suggest that diclofenac may have potential as a therapeutic option for UTI management, warranting further exploration and consideration in clinical practice [92].
In efforts to combat Yersinia pestis, the causative agent of plague, a library of 780 FDA-approved compounds was screened for their ability to block intracellular colonization. Among the most effective compounds were doxapram, amoxapine, and trifluoperazine, which reduced intracellular survival in vitro and increased animal survivability in vivo, despite not having direct antimicrobial activity [88].
The concept of broad-spectrum host-directed therapies (HDT) is particularly relevant for intracellular pathogens. Broad-spectrum HDT drugs can assist host cells in mounting an immune response against various intracellular bacteria (Table 5). However, it is crucial to first test the toxicity of these compounds, as they often alter host cell behavior and can be toxic.
Czyz et al. identified several repurposed FDA-approved drugs with broad-spectrum activity against Legionella pneumophila, Brucella abortus, Rickettsia conorii, and Coxiella burnetii. Notably, drugs related to GPCR signaling, kinases, calcium inhibitors, and sterol/hormones emerged as potential intracellular antimicrobial agents. The effectiveness of these drugs varied depending on the pathogen, highlighting the importance of understanding each bacterium’s infection mechanism. For example, blocking cholesterol trafficking was effective against C. burnetti and L. pneumophila but not against B. abortus and R. conorii [93]. Statins, used for lowering cholesterol levels, have shown effectiveness against various pathogens, as previously mentioned. Simvastatin, in particular, was also effective against Chlamydia pneumoniae, a human respiratory pathogen. By decreasing host cell isoprenoid and cholesterol levels, simvastatin disrupted the intracellular trafficking of cholesterol, reducing the intracellular survival of C. pneumoniae. This mechanism also applied to Salmonella enterica, which lacks a cholesterol synthesis pathway [94].
Trifluoperazine, initially identified as a repurposed drug against Yersinia pestis, did not exhibit direct antimicrobial activity but showed the best in vivo activity. It was hypothesized that its mechanism of action could be effective against other pathogens. Indeed, trifluoperazine was also tested in vivo against Salmonella Typhimurium and Clostridium difficile increasing animal survivability in both cases [88].
Table 5. List of repurposed drugs against several intracellular pathogens: classification, mechanism of action, and target pathogens.
Table 5. List of repurposed drugs against several intracellular pathogens: classification, mechanism of action, and target pathogens.
DrugTypeMechanism of ActionPathogenReference
CapsaicinHerbal compoundInduces autophagyS. flexneri[91]
Diclofenac sodiumAnti-inflammatoryInhibits prostaglandin G/H synthaseShigella sp.[87]
DoxapramRespiratoryBlocks intracellular colonizationY. pestis[88]
SimvastatinStatinHMG-CoA inhibitorC. pneumoniae[94]
TrifluoperazineAntipsychoticAccumulates in macrophages, targets autophagy pathwayC. pneumoniae, C. difficile, Y. pestis[88]

7. Discussion

The rapid evolution of bacteria and the increasing prevalence of antimicrobial resistance pose significant challenges to the development of new antimicrobials. This situation has led many pharmaceutical companies to deprioritize further research in this area, increasing the risk of new pandemics emerging worldwide [18,20]. Drug repurposing has emerged as a viable and cost-effective strategy to address these challenges by using existing drugs with known safety profiles (Figure 1). This review has explored various repurposed treatments against a range of intracellular pathogens, including S. aureus, M. tuberculosis, L. monocytogenes, S. enterica, S. flexneri, Y. pestis, C. pneumoniae, C. difficile and Shigella sp., focusing on drugs initially developed for cancer, anti-inflammation, autophagy modulators, ROS related drugs, and antimicrobials that have been repurposed against other bacteria. The limited number of repurposed drugs currently in clinical trials, with only two ongoing studies and one successfully completed suggests that prior approval for other uses does not necessarily expedite their authorization for new indications [8]. This could be attributed to various factors, such as the lack of efficacy in human trials or the possibility that these trials have progressed beyond the clinical stage and are advancing to the next phase. Another challenge may lie in the fact that the bactericidal mechanisms of action for many of these drugs are still under investigation [11]. Additionally, the continued effectiveness of existing antibiotic therapies for most patients, the limited understanding of potential resistance that microorganisms might develop against repurposed drugs, and the lack of sufficient funding could also contribute to the slow progress. Despite these challenges, drug repurposing remains a faster and more cost-effective approach to drug research and development [13].
This review focuses on repurposed drugs that are active against intracellular pathogens. These microorganisms share a common trait: they can alter cellular responses to evade bacterial clearance once inside the host cells, often by modifying autophagy or inflammatory responses. As a result, some of the drugs discussed here are host-directed therapies (HDTs) because they do not directly target the bacteria but instead modulate the immune response of the eukaryotic cell. This strategy has shown promising results due to its broad-spectrum effects, including cross-species activity and effectiveness against multidrug-resistant pathogens. While there is no definitive evidence yet on how host-directed therapy (HDT) will reduce antimicrobial resistance, many experts support the idea that targeting eukaryotic cells may make the development of resistance less likely. Since the mechanisms of resistance in pathogens primarily involve prokaryotic processes, the likelihood of resistance emerging against HDT is considered to be lower [4,14,15,45]. Consequently, some of the drugs under study (such as the antipsychotics pimozide, phenothiazine, thioridazine, trifluoperazine, and chlorpromazine; the anti-inflammatory drugs auranofin and diclofenac; the classic antimalarial drug artemisinin; the anticancer drug ibrutinib; and the anthelmintic selamectin) are active against various microorganisms. Their broad-spectrum activity is linked to their mechanisms of action. For example, some anticancer and antipsychotic drugs induce autophagy in infected cells, leading to bacterial clearance, although the exact mechanisms by which these drugs induce autophagy remain unclear [28,39,69,74].
The mechanisms of action of anti-inflammatory drugs are more diverse, often involving the inhibition of bacterial efflux pumps, which can enhance the effectiveness of conventional antibiotic therapies. Additionally, the anti-inflammatory effects of these drugs benefit patients by reducing systemic inflammation, which can otherwise be fatal. Finally, intracellular pathogens are exposed to oxidative bursts that can damage proteins and kill bacteria, making bacterial redox mechanisms crucial for their survival. Several of the drugs discussed, such as auranofin and ebselen, inhibit essential bacterial redox mechanisms such as TrxR, thereby reducing intracellular survival [26,37].
Drug repurposing is an active field of research that is growing exponentially in the number of published papers and, consequently, in the number of new repurposed drug candidates. Despite these efforts, several gaps remain to be addressed, such as the emergence of bacterial resistance or tolerance, potential side effects in patients with other underlying conditions, and the need for optimization of dosing and formulation. Additionally, the challenge of securing adequate funding and navigating the complex regulatory pathways for approval further complicates the development and widespread adoption of repurposed drugs. Addressing these challenges will be critical to fully realizing the potential of drug repurposing as a viable and efficient approach to treating a wide range of diseases [5,11,13].
Overall, drug repurposing offers a promising avenue for developing new treatments against bacterial infections. By leveraging the existing pharmacological knowledge and safety profiles of these drugs, we can accelerate the discovery of effective therapies and address the urgent need for new antimicrobials in the fight against bacterial pathogens.

Author Contributions

L.M.M. and M.L. conceptualization. B.L.-T., P.C., J.L.-V. and Á.M. made the bibliographic search; B.L.-T. and M.L. designed and drew the figures; Á.M. wrote the first draft; B.L.-T., P.C., J.L.-V., H.Á.F., S.F.-M., F.J., L.M.M., M.L. and Á.M. reviewed and edited. All authors have read and agreed to the published version of the manuscript.

Funding

We would like to extend our heartfelt thanks for the grant CNS2022-135378 funded by MCIN/AEI/10.13039/501100011033 and by European Union NextGenerationEU/PRTR; to the Junta de Castilla y León (JCyL), Spain, for funding our research through the grant LE044P20, and acknowledge the fellowships that have supported our team: B.L.-T. received a predoctoral fellowship from the JCyL; J.L.-V. and H.Á.F. earned the Beca de Colaboración from the Spanish Ministry of Education, with J.L.-V. also securing an FPU fellowship (FPU2022-00432); P.C. was awarded a Ph.D. fellowship by ULE; S.F.-M. was awarded with AECC summer scholarship; Á.M. received a Margarita Salas postdoctoral fellowship and M.L. was recipient of a Beatriz Galindo grant (ref. BEAGAL18/00068—BGP18/00033).

Acknowledgments

We are thankful to the anonymous reviewers for reading our manuscript and their insightful comments and suggestions.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Thakur, A.; Mikkelsen, H.; Jungersen, G. Intracellular Pathogens: Host Immunity and Microbial Persistence Strategies. J. Immunol. Res. 2019, 2019, 1356540. [Google Scholar] [CrossRef] [PubMed]
  2. Leseigneur, C.; Le-Bury, P.; Pizarro-Cerda, J.; Dussurget, O. Emerging Evasion Mechanisms of Macrophage Defenses by Pathogenic Bacteria. Front. Cell Infect. Microbiol. 2020, 10, 577559. [Google Scholar] [CrossRef] [PubMed]
  3. Chandra, P.; Grigsby, S.J.; Philips, J.A. Immune evasion and provocation by Mycobacterium tuberculosis. Nat. Rev. Microbiol. 2022, 20, 750–766. [Google Scholar] [CrossRef] [PubMed]
  4. Shapira, T.; Christofferson, M.; Av-Gay, Y. The antimicrobial activity of innate host-directed therapies: A systematic review. Int. J. Antimicrob. Agents 2024, 63, 107138. [Google Scholar] [CrossRef] [PubMed]
  5. Konreddy, A.K.; Rani, G.U.; Lee, K.; Choi, Y. Recent Drug-Repurposing-Driven Advances in the Discovery of Novel Antibiotics. Curr. Med. Chem. 2019, 26, 5363–5388. [Google Scholar] [CrossRef]
  6. Quezada, H.; Martinez-Vazquez, M.; Lopez-Jacome, E.; Gonzalez-Pedrajo, B.; Andrade, A.; Fernandez-Presas, A.M.; Tovar-Garcia, A.; Garcia-Contreras, R. Repurposed anti-cancer drugs: The future for anti-infective therapy? Expert. Rev. Anti Infect. Ther. 2020, 18, 609–612. [Google Scholar] [CrossRef]
  7. Ahmad, Z.; Rauf, A.; Naz, S.; Hemeg, H.A. Introduction to Drug Repurposing: Exploring New Applications for Existing Drugs; InTech Open: London, UK, 2024. [Google Scholar]
  8. Galindez, G.; Matschinske, J.; Rose, T.D.; Sadegh, S.; Salgado-Albarran, M.; Spath, J.; Baumbach, J.; Pauling, J.K. Lessons from the COVID-19 pandemic for advancing computational drug repurposing strategies. Nat. Comput. Sci. 2021, 1, 33–41. [Google Scholar] [CrossRef]
  9. Kulkarni, V.S.; Alagarsamy, V.; Solomon, V.R.; Jose, P.A.; Murugesan, S. Drug Repurposing: An Effective Tool in Modern Drug Discovery. Russ. J. Bioorg. Chem. 2023, 49, 157–166. [Google Scholar] [CrossRef]
  10. Pushpakom, S.; Iorio, F.; Eyers, P.A.; Escott, K.J.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; McNamee, C.; et al. Drug repurposing: Progress, challenges and recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef]
  11. Juarez-Lopez, D.; Schcolnik-Cabrera, A. Drug Repurposing: Considerations to Surpass While Re-directing Old Compounds for New Treatments. Arch. Med. Res. 2021, 52, 243–251. [Google Scholar] [CrossRef]
  12. Tiberi, S.; du Plessis, N.; Walzl, G.; Vjecha, M.J.; Rao, M.; Ntoumi, F.; Mfinanga, S.; Kapata, N.; Mwaba, P.; McHugh, T.D.; et al. Tuberculosis: Progress and advances in development of new drugs, treatment regimens, and host-directed therapies. Lancet Infect. Dis. 2018, 18, e183–e198. [Google Scholar] [CrossRef] [PubMed]
  13. Jampilek, J. Drug repurposing to overcome microbial resistance. Drug Discov. Today 2022, 27, 2028–2041. [Google Scholar] [CrossRef] [PubMed]
  14. Mourenza, A.; Gil, J.A.; Mateos, L.M.; Letek, M. Novel Treatments against Mycobacterium tuberculosis Based on Drug Repurposing. Antibiotics 2020, 9, 550. [Google Scholar] [CrossRef] [PubMed]
  15. Bravo-Santano, N.; Stolting, H.; Cooper, F.; Bileckaja, N.; Majstorovic, A.; Ihle, N.; Mateos, L.M.; Calle, Y.; Behrends, V.; Letek, M. Host-directed kinase inhibitors act as novel therapies against intracellular Staphylococcus aureus. Sci. Rep. 2019, 9, 4876. [Google Scholar] [CrossRef]
  16. Bravo-Santano, N.; Capilla-Lasheras, P.; Mateos, L.M.; Calle, Y.; Behrends, V.; Letek, M. Identification of novel targets for host-directed therapeutics against intracellular Staphylococcus aureus. Sci. Rep. 2019, 9, 15435. [Google Scholar] [CrossRef]
  17. Tong, S.Y.; Davis, J.S.; Eichenberger, E.; Holland, T.L.; Fowler, V.G., Jr. Staphylococcus aureus infections: Epidemiology, pathophysiology, clinical manifestations, and management. Clin. Microbiol. Rev. 2015, 28, 603–661. [Google Scholar] [CrossRef]
  18. Antimicrobial Resistance, C. Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef]
  19. Hajam, I.A.; Liu, G.Y. Linking S. aureus Immune Evasion Mechanisms to Staphylococcal Vaccine Failures. Antibiotics 2024, 13, 410. [Google Scholar] [CrossRef]
  20. Yu, H.; Xu, Y.; Imani, S.; Zhao, Z.; Ullah, S.; Wang, Q. Navigating ESKAPE Pathogens: Considerations and Caveats for Animal Infection Models Development. ACS Infect. Dis. 2024, 10, 2336–2355. [Google Scholar] [CrossRef]
  21. Das, S.; Dasgupta, A.; Chopra, S. Drug repurposing: A new front in the war against Staphylococcus aureus. Future Microbiol. 2016, 11, 1091–1099. [Google Scholar] [CrossRef]
  22. Martins, M.; Bleiss, W.; Marko, A.; Ordway, D.; Viveiros, M.; Leandro, C.; Pacheco, T.; Molnar, J.; Kristiansen, J.E.; Amaral, l. Clinical Concentrations of Thioridazine Enhance the Killing of Intracellular Methicillin-resistant Staphylococcus aureus: An In Vivo, Ex Vivo and Electron Microscopy Study. In Vivo 2004, 18, 787–794. [Google Scholar] [PubMed]
  23. Amaral, L.; Kristiansen, J.E.; Abebe, L.S.; Millett, W. Inhibition of the respiration of multi-drug resistant clinical isolates of Mycobacterium tuberculosis by thioridazine: Potential use for initial therapy of freshly diagnosed tuberculosis. J. Antimicrob. Chemother. 1996, 38, 1049–1053. [Google Scholar] [CrossRef] [PubMed]
  24. Ordwaya, D.; Viveiros, M.; Leandro, C.; Arroz, M.J.; Amaral, L. Intracellular activity of clinical concentrations of phenothiazines including thioridiazine against phagocytosed Staphylococcus aureus. Int. J. Antimicrob. Agents 2002, 20, 34–43. [Google Scholar] [CrossRef]
  25. Farhaa, M.A.; Czarnya, T.L.; Myersa, C.L.; Worrallc, L.J.; Frencha, S.; Conradyc, D.G.; Wangd, Y.; Oldfieldd, E.; Strynadkac, N.C.J.; Brown, E.D. Antagonism screen for inhibitors of bacterial cell wall biogenesis uncovers an inhibitor of undecaprenyl diphosphate synthase. Proc. Natl. Acad. Sci. USA 2015, 112, 11048–11053. [Google Scholar] [CrossRef] [PubMed]
  26. An, Q.; Li, C.; Chen, Y.; Deng, Y.; Yang, T.; Luo, Y. Repurposed drug candidates for antituberculosis therapy. Eur. J. Med. Chem. 2020, 192, 112175. [Google Scholar] [CrossRef]
  27. Thangamani, S.; Younis, W.; Seleem, M.N. Repurposing Clinical Molecule Ebselen to Combat Drug Resistant Pathogens. PLoS ONE 2015, 10, e0133877. [Google Scholar] [CrossRef]
  28. Folliero, V.; Dell’Annunziata, F.; Santella, B.; Roscetto, E.; Zannella, C.; Capuano, N.; Perrella, A.; De Filippis, A.; Boccia, G.; Catania, M.R.; et al. Repurposing Selamectin as an Antimicrobial Drug against Hospital-Acquired Staphylococcus aureus Infections. Microorganisms 2023, 11, 2242. [Google Scholar] [CrossRef]
  29. Thangamani, S.; Younis, W.; Seleem, M.N. Repurposing celecoxib as a topical antimicrobial agent. Front. Microbiol. 2015, 6, 750. [Google Scholar] [CrossRef]
  30. Hendrix, A.S.; Spoonmore, T.J.; Wilde, A.D.; Putnam, N.E.; Hammer, N.D.; Snyder, D.J.; Guelcher, S.A.; Skaar, E.P.; Cassat, J.E. Repurposing the Nonsteroidal Anti-inflammatory Drug Diflunisal as an Osteoprotective, Antivirulence Therapy for Staphylococcus aureus Osteomyelitis. Antimicrob. Agents Chemother. 2016, 60, 5322–5330. [Google Scholar] [CrossRef]
  31. Chifiriuc, M.C.; Filip, R.; Constantin, M.; Pircalabioru, G.G.; Bleotu, C.; Burlibasa, L.; Ionica, E.; Corcionivoschi, N.; Mihaescu, G. Common themes in antimicrobial and anticancer drug resistance. Front. Microbiol. 2022, 13, 960693. [Google Scholar] [CrossRef]
  32. Yeo, W.S.; Arya, R.; Kim, K.K.; Jeong, H.; Cho, K.H.; Bae, T. The FDA-approved anti-cancer drugs, streptozotocin and floxuridine, reduce the virulence of Staphylococcus aureus. Sci. Rep. 2018, 8, 2521. [Google Scholar] [CrossRef] [PubMed]
  33. Linzner, N.; Loi, V.V.; Antelmann, H. The Catalase KatA Contributes to Microaerophilic H2O2 Priming to Acquire an Improved Oxidative Stress Resistance in Staphylococcus aureus. Antioxidants 2022, 11, 1793. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, C.I.; Liu, G.Y.; Song, Y.; Yin, F.; Hensler, M.E.; Jeng, W.Y.; Nizet, V.; Wang, A.H.; Oldfield, E. A cholesterol biosynthesis inhibitor blocks Staphylococcus aureus virulence. Science 2008, 319, 1391–1394. [Google Scholar] [CrossRef]
  35. Chung, I.Y.; Jang, H.J.; Yoo, Y.J.; Hur, J.; Oh, H.Y.; Kim, S.H.; Cho, Y.H. Artemisinin displays bactericidal activity via copper-mediated DNA damage. Virulence 2022, 13, 149–159. [Google Scholar] [CrossRef] [PubMed]
  36. Lu, J.; Vlamis-Gardikas, A.; Kandasamy, K.; Zhao, R.; Gustafsson, T.N.; Engstrand, L.; Hoffner, S.; Engman, L.; Holmgren, A. Inhibition of bacterial thioredoxin reductase: An antibiotic mechanism targeting bacteria lacking glutathione. FASEB J. 2013, 27, 1394–1403. [Google Scholar] [CrossRef]
  37. Harbut, M.B.; Vilcheze, C.; Luo, X.; Hensler, M.E.; Guo, H.; Yang, B.; Chatterjee, A.K.; Nizet, V.; Jacobs, W.R., Jr.; Schultz, P.G.; et al. Auranofin exerts broad-spectrum bactericidal activities by targeting thiol-redox homeostasis. Proc. Natl. Acad. Sci. USA 2015, 112, 4453–4458. [Google Scholar] [CrossRef]
  38. Thangamani, S.; Mohammad, H.; Abushahba, M.F.; Sobreira, T.J.; Hedrick, V.E.; Paul, L.N.; Seleem, M.N. Antibacterial activity and mechanism of action of auranofin against multi-drug resistant bacterial pathogens. Sci. Rep. 2016, 6, 22571. [Google Scholar] [CrossRef]
  39. Hu, Y.; Wen, Z.; Liu, S.; Cai, Y.; Guo, J.; Xu, Y.; Lin, D.; Zhu, J.; Li, D.; Chen, X. Ibrutinib suppresses intracellular Mycobacterium tuberculosis growth by inducing macrophage autophagy. J. Infect. 2020, 80, e19–e26. [Google Scholar] [CrossRef]
  40. Zheng, Y.; Zhong, T.; Wu, H.; Li, N.; Fang, Z.; Cao, L.; Yin, X.; He, Q.; Ge, R.; Sun, X. Crizotinib Shows Antibacterial Activity against Gram-Positive Bacteria by Reducing ATP Production and Targeting the CTP Synthase PyrG. Microbiol. Spectr. 2022, 10, e00884-22. [Google Scholar] [CrossRef]
  41. Chang, J.; Kim, J.; Lee, W. Raloxifene prevents intracellular invasion of pathogenic bacteria through modulation of cell metabolic pathways. J. Antimicrob. Chemother. 2022, 77, 1617–1624. [Google Scholar] [CrossRef]
  42. Flores, R.; Dohrmann, S.; Schaal, C.; Hakkim, A.; Nizet, V.; Corriden, R. The Selective Estrogen Receptor Modulator Raloxifene Inhibits Neutrophil Extracellular Trap Formation. Front. Immunol. 2016, 7, 566. [Google Scholar] [CrossRef]
  43. Bravo-Santano, N.; Ellis, J.K.; Mateos, L.M.; Calle, Y.; Keun, H.C.; Behrends, V.; Letek, M. Intracellular Staphylococcus aureus Modulates Host Central Carbon Metabolism To Activate Autophagy. mSphere 2018, 3, e00374-18. [Google Scholar] [CrossRef] [PubMed]
  44. Villar-Hernandez, R.; Ghodousi, A.; Konstantynovska, O.; Duarte, R.; Lange, C.; Raviglione, M. Tuberculosis: Current challenges and beyond. Breathe 2023, 19, 220166. [Google Scholar] [CrossRef] [PubMed]
  45. Zumla, A.; Maeurer, M.; Host-Directed Therapies, N.; Chakaya, J.; Hoelscher, M.; Ntoumi, F.; Rustomjee, R.; Vilaplana, C.; Yeboah-Manu, D.; Rasolof, V.; et al. Towards host-directed therapies for tuberculosis. Nat. Rev. Drug Discov. 2015, 14, 511–512. [Google Scholar] [CrossRef] [PubMed]
  46. Torfs, E.; Piller, T.; Cos, P.; Cappoen, D. Opportunities for Overcoming Mycobacterium tuberculosis Drug Resistance: Emerging Mycobacterial Targets and Host-Directed Therapy. Int. J. Mol. Sci. 2019, 20, 2868. [Google Scholar] [CrossRef]
  47. Kumar, A.; Alam, A.; Grover, S.; Pandey, S.; Tripathi, D.; Kumari, M.; Rani, M.; Singh, A.; Akhter, Y.; Ehtesham, N.Z.; et al. Peptidyl-prolyl isomerase-B is involved in Mycobacterium tuberculosis biofilm formation and a generic target for drug repurposing-based intervention. NPJ Biofilms Microbiomes 2019, 5, 3. [Google Scholar] [CrossRef] [PubMed]
  48. Pires, D.; Valente, S.; Calado, M.; Mandal, M.; Azevedo-Pereira, J.M.; Anes, E. Repurposing Saquinavir for Host-Directed Therapy to Control Mycobacterium tuberculosis Infection. Front. Immunol. 2021, 12, 647728. [Google Scholar] [CrossRef] [PubMed]
  49. de Chastellier, C.; Thilo, L. Cholesterol depletion in Mycobacterium avium-infected macrophages overcomes the block in phagosome maturation and leads to the reversible sequestration of viable mycobacteria in phagolysosome-derived autophagic vacuoles. Cell Microbiol. 2006, 8, 242–256. [Google Scholar] [CrossRef]
  50. Lobato, L.S.; Rosa, P.S.; da Silva Ferreira, J.; da Silva Neumann, A.; Gomes da Silva, M.; Caitano do Nascimento, D.; Teixeira Soares, C.; Barbosa Pedrini, S.C.; Leal de Oliveira, D.S.; Peres Monteiro, C.; et al. Statins Increase Rifampin Mycobactericidal Effect. Antimicrob. Agents Chemother. 2014, 58, 5766–5774. [Google Scholar] [CrossRef]
  51. Skerry, C.; Pinn, M.L.; Bruiners, N.; Pine, R.; Gennaro, M.L.; Karakousis, P.C. Simvastatin increases the in vivo activity of the first-line tuberculosis regimen. J. Antimicrob. Chemother. 2014, 69, 2453–2457. [Google Scholar] [CrossRef]
  52. Gupta, S.; Tyagi, S.; Almeida, D.V.; Maiga, M.C.; Ammerman, N.C.; Bishai, W.R. Acceleration of tuberculosis treatment by adjunctive therapy with verapamil as an efflux inhibitor. Am. J. Respir. Crit. Care Med. 2013, 188, 600–607. [Google Scholar] [CrossRef] [PubMed]
  53. Vilaplana, C.; Marzo, E.; Tapia, G.; Diaz, J.; Garcia, V.; Cardona, P.J. Ibuprofen therapy resulted in significantly decreased tissue bacillary loads and increased survival in a new murine experimental model of active tuberculosis. J. Infect. Dis. 2013, 208, 199–202. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, P.T.; Stenger, S.; Li, H.; Wenzel, L.; Tan, B.H.; Krutzik, S.R.; Ochoa, M.T.; Schauber, J.; Wu, K.; Meinken, C.; et al. Toll-Like Receptor Triggering of a Vitamin D–Mediated Human Antimicrobial Response. Science 2006, 311, 1770–1773. [Google Scholar] [CrossRef]
  55. Boland, R.; Heemskerk, M.T.; Forn-Cuní, G.; Korbee, C.J.; Walburg, K.V.; Esselink, J.J.; Carvalho dos Santos, C.; de Waal, A.M.; van der Hoeven, D.C.M.; van der Sar, E.; et al. Repurposing Tamoxifen as Potential Host-Directed Therapeutic for Tuberculosis. mBio 2022, 14, e0302422. [Google Scholar] [CrossRef] [PubMed]
  56. Heemskerk, M.T.; Korbee, C.J.; Esselink, J.J.; Dos Santos, C.C.; van Veen, S.; Gordijn, I.F.; Vrieling, F.; Walburg, K.V.; Engele, C.G.; Dijkman, K.; et al. Repurposing diphenylbutylpiperidine-class antipsychotic drugs for host-directed therapy of Mycobacterium tuberculosis and Salmonella enterica infections. Sci. Rep. 2021, 11, 19634. [Google Scholar] [CrossRef] [PubMed]
  57. Napier, R.J.; Rafi, W.; Cheruvu, M.; Powell, K.R.; Zaunbrecher, M.A.; Bornmann, W.; Salgame, P.; Shinnick, T.M.; Kalman, D. Imatinib-sensitive tyrosine kinases regulate mycobacterial pathogenesis and represent therapeutic targets against tuberculosis. Cell Host Microbe 2011, 10, 475–485. [Google Scholar] [CrossRef]
  58. Kuijl, C.; Savage, N.D.; Marsman, M.; Tuin, A.W.; Janssen, L.; Egan, D.A.; Ketema, M.; van den Nieuwendijk, R.; van den Eeden, S.J.; Geluk, A.; et al. Intracellular bacterial growth is controlled by a kinase network around PKB/AKT1. Nature 2007, 450, 725–730. [Google Scholar] [CrossRef]
  59. Ouyang, Q.; Zhang, K.; Lin, D.; Feng, C.G.; Cai, Y.; Chen, X. Bazedoxifene Suppresses Intracellular Mycobacterium tuberculosis Growth by Enhancing Autophagy. mSphere 2020, 5, e00124-20. [Google Scholar] [CrossRef]
  60. Campbell, G.R.; Spector, S.A. Vitamin D inhibits human immunodeficiency virus type 1 and Mycobacterium tuberculosis infection in macrophages through the induction of autophagy. PLoS Pathog. 2012, 8, e1002689. [Google Scholar] [CrossRef]
  61. Yuk, J.M.; Shin, D.M.; Lee, H.M.; Yang, C.S.; Jin, H.S.; Kim, K.K.; Lee, Z.W.; Lee, S.H.; Kim, J.M.; Jo, E.K. Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin. Cell Host Microbe 2009, 6, 231–243. [Google Scholar] [CrossRef]
  62. Stover, C.K.; Warrener, P.; VanDevanter, D.R.; Sherman, D.R.; Arain, T.M.; Langhorne, M.H.; Anderson, S.W.; Towell, J.A.; Yuan, Y.; McMurray, D.N.; et al. A small-molecule nitroimidazopyran drug candidate for the treatment of tuberculosis. Nature 2000, 405, 962–966. [Google Scholar] [CrossRef] [PubMed]
  63. Restrepo, B.I. Metformin: Candidate host-directed therapy for tuberculosis in diabetes and non-diabetes patients. Tuberculosis 2016, 101S, S69–S72. [Google Scholar] [CrossRef] [PubMed]
  64. Magee, M.J.; Salindri, A.D.; Kornfeld, H.; Singhal, A. Reduced prevalence of latent tuberculosis infection in diabetes patients using metformin and statins. Eur. Respir. J. 2019, 53, 1801695. [Google Scholar] [CrossRef]
  65. Datta, M.; Via, L.E.; Kamoun, W.S.; Liu, C.; Chen, W.; Seano, G.; Weiner, D.M.; Schimel, D.; England, K.; Martin, J.D.; et al. Anti-vascular endothelial growth factor treatment normalizes tuberculosis granuloma vasculature and improves small molecule delivery. Proc. Natl. Acad. Sci. USA 2015, 112, 1827–1832. [Google Scholar] [CrossRef]
  66. Ameen, S.M.; Drancourt, M. In vitro susceptibility of Mycobacterium tuberculosis to trimethoprim and sulfonamides in France. Antimicrob. Agents Chemother. 2013, 57, 6370–6371. [Google Scholar] [CrossRef]
  67. Singh, A.; Somvanshi, P.; Grover, A. Drug repurposing against arabinosyl transferase (EmbC) of Mycobacterium tuberculosis: Essential dynamics and free energy minima based binding mechanics analysis. Gene 2019, 693, 114–126. [Google Scholar] [CrossRef]
  68. Battah, B.; Chemi, G.; Butini, S.; Campiani, G.; Brogi, S.; Delogu, G.; Gemma, S. A Repurposing Approach for Uncovering the Anti-Tubercular Activity of FDA-Approved Drugs with Potential Multi-Targeting Profiles. Molecules 2019, 24, 4373. [Google Scholar] [CrossRef] [PubMed]
  69. Lim, L.E.; Vilcheze, C.; Ng, C.; Jacobs, W.R., Jr.; Ramon-Garcia, S.; Thompson, C.J. Anthelmintic avermectins kill Mycobacterium tuberculosis, including multidrug-resistant clinical strains. Antimicrob. Agents Chemother. 2013, 57, 1040–1046. [Google Scholar] [CrossRef]
  70. Ranjbar, S.; Haridas, V.; Nambu, A.; Jasenosky, L.D.; Sadhukhan, S.; Ebert, T.S.; Hornung, V.; Cassell, G.H.; Falvo, J.V.; Goldfeld, A.E. Cytoplasmic RNA Sensor Pathways and Nitazoxanide Broadly Inhibit Intracellular Mycobacterium tuberculosis Growth. iScience 2019, 22, 299–313. [Google Scholar] [CrossRef]
  71. Salie, S.; Labuschagne, A.; Walters, A.; Geyer, S.; Jardine, A.; Jacobs, M.; Hsu, N.J. In vitro and in vivo toxicity evaluation of non-neuroleptic phenothiazines, antitubercular drug candidates. Regul. Toxicol. Pharmacol. 2019, 109, 104508. [Google Scholar] [CrossRef]
  72. Kumar, P.; Kaushik, A.; Lloyd, E.P.; Li, S.G.; Mattoo, R.; Ammerman, N.C.; Bell, D.T.; Perryman, A.L.; Zandi, T.A.; Ekins, S.; et al. Non-classical transpeptidases yield insight into new antibacterials. Nat. Chem. Biol. 2017, 13, 54–61. [Google Scholar] [CrossRef] [PubMed]
  73. Kaushik, A.; Makkar, N.; Pandey, P.; Parrish, N.; Singh, U.; Lamichhane, G. Carbapenems and Rifampin Exhibit Synergy against Mycobacterium tuberculosis and Mycobacterium abscessus. Antimicrob. Agents Chemother. 2015, 59, 6561–6567. [Google Scholar] [CrossRef] [PubMed]
  74. Choi, W.H. Novel Pharmacological Activity of Artesunate and Artemisinin: Their Potential as Anti-Tubercular Agents. J. Clin. Med. 2017, 6, 30. [Google Scholar] [CrossRef] [PubMed]
  75. Pethe, K.; Bifani, P.; Jang, J.; Kang, S.; Park, S.; Ahn, S.; Jiricek, J.; Jung, J.; Jeon, H.K.; Cechetto, J.; et al. Discovery of Q203, a potent clinical candidate for the treatment of tuberculosis. Nat. Med. 2013, 19, 1157–1160. [Google Scholar] [CrossRef] [PubMed]
  76. Sharma, K.; Ahmed, F.; Sharma, T.; Grover, A.; Agarwal, M.; Grover, S. Potential Repurposed Drug Candidates for Tuberculosis Treatment: Progress and Update of Drugs Identified in Over a Decade. ACS Omega 2023, 8, 17362–17380. [Google Scholar] [CrossRef]
  77. Schuchat, A.; Swaminathan, B.; Broome, C.V. Epidemiology of Human Listeriosis. Clin. Microbiol. Rev. 1991, 4, 169–183. [Google Scholar] [CrossRef]
  78. Cong, Z.; Xiong, Y.; Lyu, L.; Fu, B.; Guo, D.; Sha, Z.; Yang, B.; Wu, H. The relationship between Listeria infections and host immune responses: Listeriolysin O as a potential target. Biomed. Pharmacother. 2024, 171, 116129. [Google Scholar] [CrossRef]
  79. Beauregard, K.E.; Lee, K.-D.; Collier, R.J.; Swanson, J.A. pH-dependent Perforation of Macrophage Phagosomes by Listeriolysin O from Listeria monocytogenes. J. Exp. Med. 1997, 186, 1159–1163. [Google Scholar] [CrossRef]
  80. Gonzales-Barron, U.; Cadavez, V.; Guillier, L.; Sanaa, M. A Critical Review of Risk Assessment Models for Listeria monocytogenes in Dairy Products. Foods 2023, 12, 4436. [Google Scholar] [CrossRef]
  81. Lieberman, L.A.; Higgins, D.E. Inhibition of Listeria monocytogenes infection by neurological drugs. Int. J. Antimicrob. Agents 2010, 35, 292–296. [Google Scholar] [CrossRef]
  82. Lieberman, L.A.; Higgins, D.E. A small-molecule screen identifies the antipsychotic drug pimozide as an inhibitor of Listeria monocytogenes infection. Antimicrob. Agents Chemother. 2009, 53, 756–764. [Google Scholar] [CrossRef]
  83. Geronikaki, A.; Kartsev, V.; Petrou, A.; Akrivou, M.G.; Vizirianakis, I.S.; Chatzopoulou, F.M.; Lichitsky, B.; Sirakanyan, S.; Kostic, M.; Smiljkovic, M.; et al. Antibacterial activity of griseofulvin analogues as an example of drug repurposing. Int. J. Antimicrob. Agents 2020, 55, 105884. [Google Scholar] [CrossRef] [PubMed]
  84. Ellis, M.J.; Tsai, C.N.; Johnson, J.W.; French, S.; Elhenawy, W.; Porwollik, S.; Andrews-Polymenis, H.; McClelland, M.; Magolan, J.; Coombes, B.K.; et al. A macrophage-based screen identifies antibacterial compounds selective for intracellular Salmonella Typhimurium. Nat. Commun. 2019, 10, 197. [Google Scholar] [CrossRef] [PubMed]
  85. Li, W.; Ren, Q.; Ni, T.; Zhao, Y.; Sang, Z.; Luo, R.; Li, Z.; Li, S. Strategies adopted by Salmonella to survive in host: A review. Arch. Microbiol. 2023, 205, 362. [Google Scholar] [CrossRef] [PubMed]
  86. Catron, D.M.; Lange, Y.; Borensztajn, J.; Sylvester, M.D.; Jones, B.D.; Haldar, K. Salmonella enterica serovar Typhimurium requires nonsterol precursors of the cholesterol biosynthetic pathway for intracellular proliferation. Infect. Immun. 2004, 72, 1036–1042. [Google Scholar] [CrossRef]
  87. Dutta, N.K.; Annadurai, S.; Mazumdar, K.; Dastidar, S.G.; Kristiansen, J.E.; Molnar, J.; Martins, M.; Amaral, L. Potential management of resistant microbial infections with a novel non-antibiotic: The anti-inflammatory drug diclofenac sodium. Int. J. Antimicrob. Agents 2007, 30, 242–249. [Google Scholar] [CrossRef]
  88. Andersson, J.A.; Fitts, E.C.; Kirtley, M.L.; Ponnusamy, D.; Peniche, A.G.; Dann, S.M.; Motin, V.L.; Chauhan, S.; Rosenzweig, J.A.; Sha, J.; et al. New Role for FDA-Approved Drugs in Combating Antibiotic-Resistant Bacteria. Antimicrob. Agents Chemother. 2016, 60, 3717–3729. [Google Scholar] [CrossRef]
  89. Liu, H.; Li, S.; Denga, L.; Shi, Z.; Jiang, C.; Shua, J.; Liu, Y.; Denga, X.; Wanga, J.; Guoa, Z.; et al. Repurposing Loperamide as an Anti-Infection Drug for the Treatment of Intracellular Bacterial Pathogens. Engineering 2024. [Google Scholar] [CrossRef]
  90. Joshi, T.; Sharma, P.; Joshi, T.; Mathpal, S.; Pande, V.; Chandra, S. Repurposing of FDA approved drugs against Salmonella enterica serovar Typhi by targeting dihydrofolate reductase: An in silico study. J. Biomol. Struct. Dyn. 2022, 40, 3731–3744. [Google Scholar] [CrossRef]
  91. Basak, P.; Maitra, P.; Khan, U.; Saha, K.; Bhattacharya, S.S.; Dutta, M.; Bhattacharya, S. Capsaicin Inhibits Shigella flexneri Intracellular Growth by Inducing Autophagy. Front. Pharmacol. 2022, 13, 903438. [Google Scholar] [CrossRef]
  92. Kronenberg, A.; Butikofer, L.; Odutayo, A.; Muhlemann, K.; da Costa, B.R.; Battaglia, M.; Meli, D.N.; Frey, P.; Limacher, A.; Reichenbach, S.; et al. Symptomatic treatment of uncomplicated lower urinary tract infections in the ambulatory setting: Randomised, double blind trial. BMJ 2017, 359, j4784. [Google Scholar] [CrossRef] [PubMed]
  93. Czyz, D.M.; Potluri, L.P.; Jain-Gupta, N.; Riley, S.P.; Martinez, J.J.; Steck, T.L.; Crosson, S.; Shuman, H.A.; Gabay, J.E. Host-directed antimicrobial drugs with broad-spectrum efficacy against intracellular bacterial pathogens. mBio 2014, 5, e01534-14. [Google Scholar] [CrossRef] [PubMed]
  94. Erkkila, L.; Jauhiainen, M.; Laitinen, K.; Haasio, K.; Tiirola, T.; Saikku, P.; Leinonen, M. Effect of simvastatin, an established lipid-lowering drug, on pulmonary Chlamydia pneumoniae infection in mice. Antimicrob. Agents Chemother. 2005, 49, 3959–3962. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the most common process to repurpose drugs against intracellular pathogens, as well as the most common mechanisms of action.
Figure 1. Schematic representation of the most common process to repurpose drugs against intracellular pathogens, as well as the most common mechanisms of action.
Antibiotics 13 00834 g001
Figure 2. Summary of the scientific results obtained from different sources (PubMed, conferences and colleagues). The search was narrowed to drugs repurposed against bacteria and subsequently to intracellular bacteria. Only about 1% of the articles on drug repurposing focused on intracellular pathogens. Exclusion criteria were based on the off-target effects, toxicity, or lack of tested activity.
Figure 2. Summary of the scientific results obtained from different sources (PubMed, conferences and colleagues). The search was narrowed to drugs repurposed against bacteria and subsequently to intracellular bacteria. Only about 1% of the articles on drug repurposing focused on intracellular pathogens. Exclusion criteria were based on the off-target effects, toxicity, or lack of tested activity.
Antibiotics 13 00834 g002
Table 3. List of repurposed drugs against L. monocytogenes: classification and mechanism of action.
Table 3. List of repurposed drugs against L. monocytogenes: classification and mechanism of action.
DrugTypeMechanism of ActionReference
BepridilAntitumoralInhibits calcium channel inhibitor[67]
GriseofulvinAntifungalDisruptor of microtubules[69]
PimozideAntipsychoticDopamine D2 receptor antagonist[68]
ThioridazineAntipsychoticDopamine D2 receptor antagonist[67]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lorente-Torres, B.; Llano-Verdeja, J.; Castañera, P.; Ferrero, H.Á.; Fernández-Martínez, S.; Javadimarand, F.; Mateos, L.M.; Letek, M.; Mourenza, Á. Innovative Strategies in Drug Repurposing to Tackle Intracellular Bacterial Pathogens. Antibiotics 2024, 13, 834. https://doi.org/10.3390/antibiotics13090834

AMA Style

Lorente-Torres B, Llano-Verdeja J, Castañera P, Ferrero HÁ, Fernández-Martínez S, Javadimarand F, Mateos LM, Letek M, Mourenza Á. Innovative Strategies in Drug Repurposing to Tackle Intracellular Bacterial Pathogens. Antibiotics. 2024; 13(9):834. https://doi.org/10.3390/antibiotics13090834

Chicago/Turabian Style

Lorente-Torres, Blanca, Jesús Llano-Verdeja, Pablo Castañera, Helena Á. Ferrero, Sergio Fernández-Martínez, Farzaneh Javadimarand, Luis M. Mateos, Michal Letek, and Álvaro Mourenza. 2024. "Innovative Strategies in Drug Repurposing to Tackle Intracellular Bacterial Pathogens" Antibiotics 13, no. 9: 834. https://doi.org/10.3390/antibiotics13090834

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