Next Article in Journal
A Narrative Review on the Approach to Antimicrobial Use in Ventilated Patients with Multidrug Resistant Organisms in Respiratory Samples—To Treat or Not to Treat? That Is the Question
Next Article in Special Issue
Fighting Back against Antimicrobial Resistance with Comprehensive Policy and Education: A Narrative Review
Previous Article in Journal
Neurodegenerative Disease Treatment Drug PBT2 Breaks Intrinsic Polymyxin Resistance in Gram-Positive Bacteria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review: Antimicrobial Therapy for Human Pythiosis

by
Sadeep Medhasi
1,
Ariya Chindamporn
2 and
Navaporn Worasilchai
3,*
1
Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
2
Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
3
Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Immunomodulation of Natural Products Research Group, Chulalongkorn University, Bangkok 10330, Thailand
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(4), 450; https://doi.org/10.3390/antibiotics11040450
Submission received: 14 January 2022 / Revised: 16 March 2022 / Accepted: 22 March 2022 / Published: 26 March 2022

Abstract

:
Human pythiosis is associated with poor prognosis with significant mortality caused by Pythium insidiosum. Antimicrobials’ in vitro and in vivo results against P. insidiosum are inconsistent. Although antimicrobials are clinically useful, they are not likely to achieve therapeutic success alone without surgery and immunotherapy. New therapeutic options are therefore needed. This non-exhaustive review discusses the rationale antimicrobial therapy, minimum inhibitory concentrations, and efficacy of antibacterial and antifungal agents against P. insidiosum. This review further provides insight into the immunomodulating effects of antimicrobials that can enhance the immune response to infections. Current data support using antimicrobial combination therapy for the pharmacotherapeutic management of human pythiosis. Also, the success or failure of antimicrobial treatment in human pythiosis might depend on the immunomodulatory effects of drugs. The repurposing of existing drugs is a safe strategy for anti-P. insidiosum drug discovery. To improve patient outcomes in pythiosis, we suggest further research and a deeper understanding of P. insidiosum virulence factors, host immune response, and host immune system modification by antimicrobials.

1. Introduction

Human pythiosis is an infectious disease with high morbidity and mortality [1]. Pythium insidiosum, a fungus-like aquatic oomycete microorganism, is a causative agent of pythiosis. The motile flagellate zoospore plays a significant role in initiating an infection. The zoospores of P. insidiosum adhere to the skin cut or wound sites and encyst on the surface of the injured tissue(s). The encysted spore develops a germination tube (hypha) that uses chemotaxis to find the host and infiltrate human blood vessels [1,2]. Pythiosis risk is higher in tropical and subtropical regions, including Southeast Asia, eastern coastal Australia, and South America [3].
Human pythiosis is associated with a poor prognosis due to the difficulties in diagnosing the infection and the lack of effective therapeutic agents against this disease [4]. The clinical features of human pythiosis are classified into four forms: (i) vascular pythiosis characterized by arteritis, thrombosis, gangrene, aneurysm, or limb claudication; (ii) ocular pythiosis characterized by corneal ulcers, decreased visual acuity, conjunctival redness, eyelid swelling, or multiple, linear, tentacle-like infiltrates and dot-like or pinhead-shaped infiltrates in the surrounding cornea; (iii) cutaneous and subcutaneous pythiosis characterized by a granulomatous and ulcerating lesion in the face or limbs, cellulitis, soft tissue abscess, or lymphadenopathy; and (iv) disseminated pythiosis characterized by the infection of internal organs [1,4,5]. The risk factors for vascular pythiosis include thalassemia, hemoglobinopathy, paroxysmal nocturnal hemoglobinuria, aplastic anemia, and leukemia because P. insidiosum has a higher affinity for iron [6,7].
When an infection is diagnosed as P. insidiosum, the therapeutic options include surgery, pharmacotherapy, and immunotherapy (Figure 1) [8]. Surgical intervention is the mainstay treatment for managing human pythiosis, but such treatment substantially increases the financial burden on patients, postsurgical complications, and uncontrolled infection [9]. Immunotherapy is a promising approach for human pythiosis treatment where antigens of P. insidiosum from in vitro cultures are injected into the patient [10,11]. The mechanism behind P. insidiosum antigen (PIA) immunotherapy in human pythiosis includes a switching from the host’s T helper-2 (Th2) to T helper-1 (Th1) mediated immune response in the host; the Th1 response producing higher levels of interferon-γ (IFN-γ) and interleukin 2 (IL-2) [10,12,13]. Even though a good prognosis in PIA-treated patients can be implied by Th2 to Th1 switching, the efficacy of P. insidiosum antigen is inconclusive when used as immunotherapy in human pythiosis [7,12,14]. In vitro studies have demonstrated the anti-P. insidiosum effect of antifungals even though the P. insidiosum lacks the antifungal drug-target: ergosterol biosynthetic pathway [15]. However, a significant concern with antifungals is the contradictory results in susceptibility to P. insidiosum in in vitro and clinical use [15,16].
This review focuses on evidence supporting and disputing the effectiveness of antimicrobials to expand the pharmacotherapeutic role of antimicrobials in the management of human pythiosis. We do not explicitly discuss the biology of P. insidiosum, the pathogenesis of the P. insidiosum infection in humans, or the management of human pythiosis with immunotherapy and surgical intervention. Finally, we conclude with general remarks on future strategic options for managing human pythiosis.

2. Principles of Antimicrobial Therapy

Antimicrobial therapy should achieve a clinical response by eliminating the invading microorganism(s) while minimizing cost, adverse effects, and antimicrobial resistance [17,18]. When selecting appropriate antimicrobial therapy, both pharmacokinetic and pharmacodynamic properties of the drug(s) must be considered to ensure that effective agents are administered in sufficient doses for therapeutic success [19]. For species such as P. insidiosum, identifying potential targets for antimicrobials is necessary for managing pythiosis. The microbial cell wall is a critical target for antimicrobials, and the cell wall of P. insidiosum is primarily composed of β-glucan and cellulose [20]. However, the cell wall of P. insidiosum lowers the penetration of drug molecules and prevents drug access to targets inside the cell wall [21]. The gene expression of cytochrome oxidase 2 (COX2) in Thai P. insidiosum strains was 2.5-fold higher at 37 °C compared to the expression at 27 °C [22]. In addition, the elicitin protein, ELI025, was highly up-regulated in P. insidiosum hyphae at 37 °C compared to hyphae grown at 28 °C and facilitated the evasion of the host antibody response [23]. COX2 and ELIO25 can be candidate targets for controlling P. insidiosum infection.
Several antifungal and antibacterial drugs have been examined for their susceptibility profile against P. insidiosum in an in vitro study (Figure 2). They have been tried to manage human pythiosis but have been successful only in a few cases [4]. P. insidiosum keratitis was successfully managed in a 20-year-old Japanese man following triple antibiotic therapy (minocycline ointment four times a day, chloramphenicol eye drops hourly, and linezolid 1200 mg orally twice a day) [24]. Recently, a P. insidiosum keratitis patient was successfully managed with topical 0.2% linezolid and topical 1% azithromycin, administered hourly [25]. Antimicrobial susceptibility testing (AST) is a procedure to determine the concentration of an antimicrobial that inhibits microbial growth in vitro by establishing minimum inhibitory concentration (MIC), which is the lowest concentration of an antimicrobial that inhibits visible growth of a microorganism [26,27]. Table 1 summarizes the methods used to determine the MIC of antimicrobial drugs against P. insidiosum discussed in our review.

3. Why Do Antimicrobial Treatments Fail?

Factors contributing to the antimicrobial treatment failure include antimicrobial agent’s pharmacokinetic and pharmacodynamic issues related to the antimicrobial agent, lack of pathogen control, development of infection complications, drug-resistant pathogens, conflicting AST results, disparities between in vitro and in vivo efficacy, host immune response, and wrong choice of antimicrobial drug (Figure 3) [33,34]. Pharmacokinetics variability can be defined as differences in plasma antimicrobial exposure, impacting treatment success [35]. Antimicrobials, like beta-lactams and aminoglycosides, achieve suboptimal plasma concentrations in critically ill patients due to increased volume of distribution and increased renal and hepatic clearance [36,37]. As another example, linezolid’s pharmacokinetic variability results in adverse effects and ineffective therapy because of the narrow therapeutic window of linezolid [38].
P. insidiosum produces six enzymes (ERG3, ERG5, ERG11, ERG20, ERG24, and ERG26) included in the sterol biosynthetic pathways [16]. However, more than 40 enzymes are involved in the sterol biosynthetic pathways; thus, drugs targeting sterol pathways exhibit limited efficacy against P. insidiosum. These drugs cannot be exploited for rationalized and successful management of pythiosis [39]. Different strategies could be considered to prevent the antimicrobial treatment failure in pythiosis, namely: delivering adequate concentration of antimicrobial drug at the site of infection [40], increased periods of exposure of P. insidiosum to the antimicrobial drug [41], redesigning drug to penetrate the outer membrane of P. insidiosum and avoid being pumped out of the membrane [42], and modulate host immunity [43].

4. Immune Response and Antimicrobial Therapy

The innate immune system protects the host from various toxins and infectious agents, including bacteria, fungi, viruses, and parasites via phagocytosis and intracellular killing, recruitment of other inflammatory cells, and presentation of antigens [44]. The innate immune system is highly complex and comprises physical and anatomical barriers, effector cells, antimicrobial peptides, soluble mediators, and cell receptors [45]. However, pathogens can breach the early innate immune mechanisms. In these circumstances, a strategy to modify the function of immune cells can lead to the elimination of the pathogenic intruder [46]. Interestingly, host immunity is often overlooked in the process of pathogen clearance. A favorable innate immune response can considerably reduce the need for more prolonged antimicrobial therapy in infections [47].
Once P. insidiosum enters and adheres to the host tissues, the soluble exoantigens from P. insidiosum trigger the Th2 response and lock the host immune system into a Th2 subset. Further, P. insidiosum protects itself from the host immune system by concealing inside the eosinophilic material formed by the eosinophil degranulation, which helps protect the P. insidiosum from being fully presented to the host’s immune system [10]. Toll-like receptors (TLRs) play a central role in the innate immune system by recognizing pathogen-associated molecular patterns and triggering downstream signaling pathways that activate the innate immune response [48]. Wongprompitak et al. demonstrated that both zoospores and hyphae of P. insidiosum induced a TLR2-mediated innate immune response with a subsequent increase in the levels of the pro-inflammatory cytokines IL-6 and IL-8 [49].
To combat the pathogen and prevent its spread, it is rational to administer antimicrobial drugs that interact with the host’s innate immune system to provide profound indirect effects and enhance pathogen clearance. Antimicrobial drugs have been shown to modify the immune responses to infection, guiding improved treatment strategies in human pythiosis (Table 2 and Figure 3).

5. Antibacterial Drugs against P. insidiosum

Previous in vitro screening of antibacterial drugs has identified tetracycline, minocycline, tigecycline, azithromycin, clarithromycin, erythromycin, gentamicin, streptomycin, paromomycin, neomycin, linezolid, nitrofurantoin, quinupristin-dalfopristin, chloramphenicol, clindamycin, and mupirocin, which demonstrated inhibitory activity against P. insidiosum [29,30,31,63,64,65]. Among the arsenal of antibiotics, the best-studied antibiotics in human pythiosis are tetracyclines, macrolides, and oxazolidinones. This section discusses different classes of antibacterial drugs to manage human pythiosis.

5.1. Tetracyclines

Tetracycline antibiotics such as tetracycline, tigecycline, and minocycline inhibit bacterial protein synthesis by binding with the bacterial 30S ribosomal subunit [66]. Tetracyclines can inhibit mammalian collagenase activity and assist wound healing [67]. Further, tetracyclines potentiate the innate immune response and augment the resolution of inflammation [50].
Based on the in vivo studies in rabbits, minocycline in combination with immunotherapy may be an effective therapeutic medical treatment of pythiosis to heal injuries [68]. Worasilchai et al. evaluated the in vitro susceptibility of human, environmental, and animal P. insidiosum isolates to eight antibiotic classes and demonstrated that tetracyclines and macrolides inhibited the in vitro growth of P. insidiosum isolates at concentrations 10 to 100 times lower than those observed for previously studied antifungal drugs [28]. Also, the combination of tetracyclines and macrolides resulted in a synergistic effect that reduced MICs against P. insidiosum isolates. Loreto et al. also reported a similar in vitro susceptibility of P. insidiosum isolates to tetracyclines and their superior potency compared to amphotericin B, echinocandins, and triazole antifungals [29].

5.2. Macrolides

Macrolides are the group of antibiotics that inhibit bacterial protein synthesis by binding with the bacterial 50S ribosomal subunit. Common macrolides include erythromycin, clarithromycin, and azithromycin [69]. Among the macrolides, azithromycin, in particular, is highly accumulated in phagocytes and is targeted to the sites of infection [70]. Azithromycin reduces the production of IL-12, resulting in enhanced Th2 response [51]. Th2 cells are involved in wound healing and tissue repair [71,72]. The immunomodulatory activities of macrolides are evident with both pro-inflammatory and anti-inflammatory effects. For example, erythromycin can suppress pro-inflammatory cytokine production, such as IL-6, IL-8, and tumor necrosis factor-α (TNF-α) [73].
Jesus et al. investigated the antimicrobial activity of azithromycin alone and in combination with minocycline against P. insidiosum in a rabbit model [74]. The results revealed a strong in vivo activity of azithromycin (20 mg/kg/day twice daily) alone and combination with minocycline (10 mg/kg/day twice daily) against subcutaneous lesions. In an in vitro susceptibility study, the MICs of azithromycin and clarithromycin were less than 4 μg/mL for P. insidiosum isolates [29].

5.3. Oxazolidinones

Oxazolidinones such as linezolid inhibit bacterial protein synthesis by binding with the 50S subunit of the ribosome [75]. The suppression of the synthesis of pro-inflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, IL-8, IFN-γ, and TNF-α by linezolid has highlighted an exciting role of linezolid in immunomodulatory effects [52,53,54]. Linezolid may significantly reduce the inflammatory damage induced by the excessive release of pro-inflammatory cytokines during critical infections [76].
In a rabbit model of P. insidiosum keratitis, topical linezolid demonstrated superior efficacy and safety compared to azithromycin and tigecycline after prolonged treatment for more than 3–4 weeks [77].

5.4. Lincosamides, Streptogramins, and Phenicols

Lincosamides, streptogramins, and phenicols inhibit bacterial protein synthesis by interacting with the 50S subunit of bacterial ribosomes [78]. Among lincosamides, clindamycin possesses immunomodulatory activity by suppressing the release of inflammatory cytokines such as TNF-α and IL-1β and enhancing the phagocytosis of microorganisms by host cells [55,56]. Quinupristin and dalfopristin, used in a fixed combination, belong to a class of streptogramins [78]. Quinupristin-dalfopristin decreased the concentration of pro-inflammatory cell wall components (lipoteichoic acid and teichoic acid) and TNF activity in cerebrospinal fluid compared to the ceftriaxone-treated rabbits [57]. A previous report showed that chloramphenicol, a member of the phenicols group, elevated the IL-10 levels, a potent anti-inflammatory cytokine [58].
Lincosamides, streptogramins, and phenicols have shown the ability to inhibit the growth of P. insidiosum isolates. The microdilution-based MIC ranges (with geometric means) of lincosamides, streptogramins, and phenicols against P. insidiosum were reported to be 2 to >4 μg/mL, 1 to >2 μg/mL, and 8 to >16 μg/mL, respectively [30].

5.5. Aminoglycosides

Aminoglycosides such as gentamicin, streptomycin, paromomycin, and neomycin bind to the bacterial ribosome and inhibit protein synthesis [79]. Streptomycin stimulated the in vitro growth of one of the Thai P. insidiosum isolates [80]. Aminoglycoside antibiotics inhibited the in vitro growth of P. insidiosum; however, they may not be clinically relevant due to the high MIC values [31]. Therefore, aminoglycosides for clinical use in managing human pythiosis are questionable.

5.6. Miscellaneous Antibacterial Drugs

Nitrofurantoin is used to treat urinary tract infections and works by attacking bacterial ribosomal proteins non-specifically, causing complete inhibition of protein synthesis [81]. P. insidiosum mycelial growth was inhibited with nitrofurantoin (MIC range of 64 to >64 μg/mL) in an in vitro susceptibility test [30].
Mupirocin inhibits bacterial protein and RNA synthesis by reversibly inhibiting isoleucyl-transfer RNA [82]. A study evaluating the in vitro susceptibility of Brazilian P. insidiosum strains showed that mupirocin could inhibit the growth of P. insidiosum isolates at MIC lower than 4 μg/mL [29].

6. Antifungal Drugs against P. insidiosum

Studies have focused on several antifungal medications, such as polyenes, azoles, allylamines, and echinocandins, for the adjunctive therapy in managing human pythiosis [4]. Despite the evidence of anti-P. insidiosum effects, it has been highly challenging to achieve consistently effective antifungal treatment in human pythiosis.

6.1. Polyenes

Amphotericin B is a polyene antifungal that binds to ergosterol in the fungal cell membrane, which alters cell membrane permeability leading to the loss of intracellular components [83]. Two Australian cases with subcutaneous pythiosis responded well to amphotericin B treatment [84]. However, the evidence of the effectiveness of amphotericin B against other forms of human pythiosis and substantial activity against P. insidiosum is lacking [14,85,86,87].
Studies have shown amphotericin B’s immunomodulatory properties, which activate the host’s innate immunity [88]. Nitric oxide (NO) is an endogenous regulator of inflammation and an antibacterial agent, and it plays a crucial role in wound repair [89,90]. Amphotericin B can augment the IL-1β-induced inducible nitric-oxide synthase (iNOS) expression and NO production [59]. In addition, amphotericin B is reported to induce oxidative stress and improve antifungal efficacy [91,92].

6.2. Allylamines and Azoles

The primary mode of action of allylamines, such as terbinafine, is the inhibition of the enzyme squalene monooxygenase. Therefore, these drugs inhibit the fungal synthesis of ergosterol [93]. Azoles, such as miconazole, ketoconazole, fluconazole, itraconazole, posaconazole, and voriconazole, exhibit antifungal activity by inhibiting the 14α-lanosterol demethylase, a key enzyme in ergosterol biosynthesis, in fungi [94,95]. Studies have suggested that the enhanced microbiocidal activity of monocytes, macrophages, and neutrophils against intracellular Candida albicans is enhanced when combined with azoles [61,62]. However, terbinafine has been reported to stimulate pro-inflammatory cytokines [60].
Susaengrat et al. reported favorable responses to voriconazole and itraconazole in Thai vascular pythiosis patients [96]. Synergistic effects have been demonstrated for terbinafine and fluconazole against P. insidiosum isolates in vitro [97]. A synergistic combination of itraconazole and terbinafine was effective during the in vitro susceptibility testing of a P. insidiosum isolate from the 2-year-old patient with a deeply invasive facial infection [98]. The growth of P. insidiosum isolates was inhibited by terbinafine, and the efficacy of terbinafine increased against P. insidiosum isolates when combined with cetrimide, an antiseptic [63]. Pediatricians used a combination of itraconazole and terbinafine to manage a child with vascular pythiosis [99]. In vitro susceptibility testing of P. insidiosum showed a MICs from 0.5 to 128 μg/mL for terbinafine, 2 to 32 μg/mL for miconazole, 4 to 64 μg/mL for ketoconazole, 1 to >128 μg/mL for itraconazole, 2 to >16 μg/mL for voriconazole, greater than 1 to >32 μg/mL for fluconazole, and >8 μg/mL for posaconazole based on the strains of P. insidiosum [9,14].

6.3. Echinocandins

Echinocandins, such as caspofungin, anidulafungin, and micafungin, act by inhibiting beta-(1,3)-D-glucan synthase, an enzyme that is necessary for the synthesis of beta-(1,3)-D-glucan, which is an essential component of the fungal cell wall [99]. Studies have documented the immunomodulatory effects of echinocandins with increased fungal beta-(1,3)-D-glucan exposure and caspofungin-induced neutrophil-mediated fungal damage and anidulafungin- and micafungin-induced phagocyte-mediated fungal damage [100,101].
Synergistic anti-P. insidiosum effects were observed with caspofungin and terbinafine in vitro [97]. The MICs of caspofungin and anidulafungin against human P. insidiosum isolates ranged from 2 to 8 μg/mL [32]. However, when used alone, echinocandins showed poor in vitro and in vivo activity against P. insidiosum [29,102]. Caspofungin demonstrated less fungistatic activity against P. insidiosum [103].

6.4. Miscellaneous Antifungal Drugs

Amorolfine, a morpholine derivative, inhibits fungal ergosterol biosynthesis and leads to changes in the membrane permeability, which in turn causes fungal growth inhibition and cell death [104]. Only recently, amorolfine hydrochloride exhibited in vitro inhibitory activity against P. insidiosum [105]. The MICs of amorolfine hydrochloride tested against P. insidiosum isolates were 16 to 64 mg/L. Further, amorolfine hydrochloride produced alterations in P. insidiosum hyphae, with changes in the surface of hyphae, intracellular organelles, the cell wall, and plasma membrane of P. insidiosum.

7. Repurposing Antimicrobials against P. insidiosum

Due to the limited success of pharmacological interventions against P. insidiosum in humans, identifying novel therapeutic strategies is required to treat P. insidiosum infection in humans. Drug repurposing is a process for identifying new therapeutic indications different from the scope of the initial pharmacological indication [106]. For example, antibiotics such as macrolides, tetracyclines, and fluoroquinolones have been used in the clinical management of coronavirus disease 2019 (COVID-19) [107]. Using the drug repurposing strategy, existing FDA-approved antimicrobials can forgo early phases of drug development in managing human pythiosis [108]. Disulfiram irreversibly inhibits aldehyde dehydrogenase (ALDH1A1) and is an alcohol-deterrent medication that causes a severe adverse reaction when patients use alcohol. Disulfiram effectively treats individuals dependent on alcohol but highly motivated to discontinue alcohol use [109]. Krajaejun et al. evaluated disulfiram for its anti-P. insidiosum activity using agar- and broth-based methods and revealed that P. insidiosum strains were susceptible to disulfiram with MICs ranging from 8 to 32 mg/Liter [110]. Further, disulfiram was found to bind and inactivate aldehyde dehydrogenase and urease of P. insidiosum.
Researchers utilize computational and experimental approaches to identify the promising candidates in the drug repurposing process [111]. The computational system uses various databases and computational tools, such as Gene Signature Database (GeneSigDB), Gene Set Enrichment Analysis (GSEA), The Pharmacogenetics and Pharmacogenomics Knowledge Base (PharmGKB), DrugBank, ChemBank, Genecard, Online Mendelian Inheritance in Man (OMIM), PubMed, e-Drug3D, DrugPredict, Promiscuous, Mantra2.0, Protein Data Bank (PDB), DRAR-CPI, repoDB, Repurpose DB, DeSigN, Cmap, and DPDR-CPI, etc. [106,112]. Computational techniques employed for drug repurposing include (i) profile-based drug repositioning, (ii) network-based drug repositioning, and (iii) data-based drug repositioning [113]. Experimental-based approaches validate the computer-generated hits for preclinical drug evaluation [112]. An experimental technique for drug repurposing involves protein target-based and cell/organism-based screens in in vitro and in vivo assays [114].
Using combination regimens of antibacterial plus antifungal or antibacterial plus antibacterial to achieve synergistic activity is one of the drug repurposing strategies against P. insidiosum [115]. Synergism between antibacterial and antifungal against P. insidiosum was observed for in vitro minocycline with amphotericin B, itraconazole, and micafungin and clarithromycin with micafungin [65]. Susaengrat et al. reported two cases of relapsed vascular pythiosis patients who were successfully clinically managed with a combination of antibacterial plus antifungal [96]. However, isolate-specific combinations for treatment must be implemented because of the varying effectiveness of any given drug combination for different isolates of P. insidiosum [116]. Studies have found the enhanced killing effects of multiple classes of antibiotics when combined with NO [117,118]. We expect that NO-containing antibiotics might improve the therapeutic outcomes in patients with pythiosis.

8. Conclusions and Future Perspectives

Evidence supports using the antimicrobials reviewed in our article as a new therapeutic option in treating human pythiosis. In vitro studies have demonstrated the tetracyclines, macrolides, oxazolidinones, lincosamides, streptogramins, phenicols, aminoglycosides, polyenes, allylamines, azoles, and echinocandins reviewed in our papers inhibit the growth of P. insidiosum and have the potential implications for further research on their use in the management of human pythiosis. However, prolonged use of antimicrobials and prolonged treatment with antimicrobials is not warranted due to the side effects and threat of antimicrobial resistance. A practical pharmacological intervention guideline for human pythiosis remains to be discovered and is necessary to assist practitioner and patient decisions, lower treatment costs, and optimize patient outcomes. Despite the disease affecting the most vulnerable populations with higher mortality rates, pythiosis is not included in the Sanford Guide, which provides evidence-based recommendations for treating infectious diseases [119].
In the future, human pythiosis could be managed with antimicrobials owing to their anti-inflammatory and immunomodulatory activities. Clinicians can optimize drug combinations based on the anti-P. insidiosum susceptibility testing for the management of pythiosis. Studies have shown the growth inhibitory effects of antimicrobials against P. insidiosum; nevertheless, studies regarding the mechanism of action of the antimicrobials against P. insidiosum are vital for clinical approval. Researchers must consider the pharmacodynamics principle involved in selecting the antimicrobials to assess the anti-P. insidiosum activity.
Microbial virulence factors are molecules produced by microorganisms and may cause disease in the host (e.g., toxins, enzymes, exopolysaccharides, lipopolysaccharides, lipoproteins, etc.) [22]. The potential virulence factors of P. insidiosum include glucan 1,3-beta-glucosidase, heat shock protein (Hsp) 70, and enolase [23]. Keeratijarut et al. reported genetic, immunological, and biochemical characteristics of Exo-1,3-β-glucanase (Exo1) in P. insidiosum and found up-regulated exo1 expression at 37 °C compared to 28 °C, thus suggesting a drug target against P. insidiosum [120]. A new therapeutic approach with anti-virulence therapy combined with antimicrobials might prevent the pathogenesis of P. insidiosum and limit host damage. Metabolites have been isolated from Pseudomonas stutzeri and Klebsiella pneumoniaei, and these organisms have shown anti-P. insidiosum activity [15,121]. Therefore, the role of potential microbial metabolites in the treatment of pythiosis must be subjected to intense research in the future.
With the evidence of the effectiveness of some antimicrobials in the management of human pythiosis, we suggest using new drug delivery systems to release the drug to the target site in the body and minimize the off-target accumulation of the drug. Antibiotics can be reformulated using nanotechnology-derived delivery systems to improve the targeting and specificity at the infected areas [122]. Due to the genetic variability among individuals, not all individuals with pythiosis exhibit similar therapeutics responses to antimicrobials [123]. Therefore, it is essential to incorporate the pharmacogenomics assay into the clinics to personalize antimicrobial treatment in pythiosis.

Author Contributions

S.M. and N.W.; conceptualization, N.W. and A.C.; funding acquisition, S.M.; writing–original draft preparation, N.W. and A.C.; writing–review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially funded by the National Research Council of Thailand and Health Systems Research Institute (No. 64-087). The funder had no role in the study design, collection, and interpretation of the data or the decision to submit the work for publication.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank the members of the Mycology, Epidemiology, and Education Research Group (MERG), Sureerat Watcharasuwanseree from Udon Thani hospital, and the pythiosis patient for supporting the clinical part and clinical photograph of human pythiosis.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Permpalung, N.; Worasilchai, N.; Chindamporn, A. Human Pythiosis: Emergence of Fungal-Like Organism. Mycopathologia 2020, 185, 801–812. [Google Scholar] [CrossRef] [PubMed]
  2. Gaastra, W.; Lipman, L.J.; De Cock, A.W.; Exel, T.K.; Pegge, R.B.; Scheurwater, J.; Vilela, R.; Mendoza, L. Pythium insidiosum: An overview. Vet. Microbiol. 2010, 146, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mar Htun, Z.; Laikul, A.; Pathomsakulwong, W.; Yurayart, C.; Lohnoo, T.; Yingyong, W.; Kumsang, Y.; Payattikul, P.; Sae-Chew, P.; Rujirawat, T.; et al. Identification and Biotyping of Pythium insidiosum Isolated from Urban and Rural Areas of Thailand by Multiplex PCR, DNA Barcode, and Proteomic Analyses. J. Fungi 2021, 7, 242. [Google Scholar] [CrossRef] [PubMed]
  4. Chitasombat, M.N.; Jongkhajornpong, P.; Lekhanont, K.; Krajaejun, T. Recent update in diagnosis and treatment of human pythiosis. PeerJ 2020, 8, e8555. [Google Scholar] [CrossRef] [Green Version]
  5. Krajaejun, T.; Imkhieo, S.; Intaramat, A.; Ratanabanangkoon, K. Development of an immunochromatographic test for rapid serodiagnosis of human pythiosis. Clin. Vaccine Immunol. 2009, 16, 506–509. [Google Scholar] [CrossRef] [Green Version]
  6. Zanette, R.A.; Bitencourt, P.E.R.; Alves, S.H.; Fighera, R.A.; Flores, M.M.; Wolkmer, P.; Hecktheuer, P.A.; Thomas, L.R.; Pereira, P.L.; Loreto, E.S.; et al. Insights into the pathophysiology of iron metabolism in Pythium insidiosum infections. Vet. Microbiol. 2013, 162, 826–830. [Google Scholar] [CrossRef]
  7. Chitasombat, M.N.; Larbcharoensub, N.; Chindamporn, A.; Krajaejun, T. Clinicopathological features and outcomes of pythiosis. Int. J. Infect. Dis. 2018, 71, 33–41. [Google Scholar] [CrossRef] [Green Version]
  8. Mendoza, L.; Ajello, L.; McGinnis, M.R. Infections Caused by the Oomycetous Pathogen Pythium insidiosum. J. Med. Mycol. 1996, 6, 151–164. [Google Scholar]
  9. Yolanda, H.; Krajaejun, T. Review of methods and antimicrobial agents for susceptibility testing against Pythium insidiosum. Heliyon 2020, 6, e03737. [Google Scholar] [CrossRef]
  10. Mendoza, L.; Newton, J.C. Immunology and immunotherapy of the infections caused by Pythium insidiosum. Med. Mycol. 2005, 43, 477–486. [Google Scholar] [CrossRef] [Green Version]
  11. Wanachiwanawin, W.; Mendoza, L.; Visuthisakchai, S.; Mutsikapan, P.; Sathapatayavongs, B.; Chaiprasert, A.; Suwanagool, P.; Manuskiatti, W.; Ruangsetakit, C.; Ajello, L. Efficacy of immunotherapy using antigens of Pythium insidiosum in the treatment of vascular pythiosis in humans. Vaccine 2004, 22, 3613–3621. [Google Scholar] [CrossRef] [PubMed]
  12. Yolanda, H.; Krajaejun, T. History and Perspective of Immunotherapy for Pythiosis. Vaccines 2021, 9, 1080. [Google Scholar] [CrossRef] [PubMed]
  13. Tondolo, J.S.M.; Ledur, P.C.; Loreto, E.S.; Verdi, C.M.; Bitencourt, P.E.R.; de Jesus, F.P.K.; Rocha, J.P.; Alves, S.H.; Sassaki, G.L.; Santurio, J.M. Extraction, characterization and biological activity of a (1,3)(1,6)-beta-d-glucan from the pathogenic oomycete Pythium insidiosum. Carbohydr. Polym. 2017, 157, 719–727. [Google Scholar] [CrossRef] [PubMed]
  14. Permpalung, N.; Worasilchai, N.; Plongla, R.; Upala, S.; Sanguankeo, A.; Paitoonpong, L.; Mendoza, L.; Chindamporn, A. Treatment outcomes of surgery, antifungal therapy and immunotherapy in ocular and vascular human pythiosis: A retrospective study of 18 patients. J. Antimicrob. Chemother. 2015, 70, 1885–1892. [Google Scholar] [CrossRef] [Green Version]
  15. Wittayapipath, K.; Yenjai, C.; Prariyachatigul, C.; Hamal, P. Evaluation of antifungal effect and toxicity of xanthyletin and two bacterial metabolites against Thai isolates of Pythium insidiosum. Sci. Rep. 2020, 10, 4495. [Google Scholar] [CrossRef]
  16. Lerksuthirat, T.; Sangcakul, A.; Lohnoo, T.; Yingyong, W.; Rujirawat, T.; Krajaejun, T. Evolution of the Sterol Biosynthetic Pathway of Pythium insidiosum and Related Oomycetes Contributes to Antifungal Drug Resistance. Antimicrob. Agents Chemother. 2017, 61, e02352-16. [Google Scholar] [CrossRef] [Green Version]
  17. Thom, K.A.; Schweizer, M.L.; Osih, R.B.; McGregor, J.C.; Furuno, J.P.; Perencevich, E.N.; Harris, A.D. Impact of empiric antimicrobial therapy on outcomes in patients with Escherichia coli and Klebsiella pneumoniae bacteremia: A cohort study. BMC Infect. Dis. 2008, 8, 116. [Google Scholar] [CrossRef] [Green Version]
  18. Rawson, T.M.; Wilson, R.C.; O’Hare, D.; Herrero, P.; Kambugu, A.; Lamorde, M.; Ellington, M.; Georgiou, P.; Cass, A.; Hope, W.W.; et al. Optimizing antimicrobial use: Challenges, advances and opportunities. Nat. Rev. Microbiol. 2021, 19, 747–758. [Google Scholar] [CrossRef]
  19. Jarrell, A.S.; Kruer, R.M.; Johnson, D.; Lipsett, P.A. Antimicrobial Pharmacokinetics and Pharmacodynamics. Surg. Infect. (Larchmt) 2015, 16, 375–379. [Google Scholar] [CrossRef]
  20. Melida, H.; Sandoval-Sierra, J.V.; Dieguez-Uribeondo, J.; Bulone, V. Analyses of extracellular carbohydrates in oomycetes unveil the existence of three different cell wall types. Eukaryot. Cell 2013, 12, 194–203. [Google Scholar] [CrossRef] [Green Version]
  21. Pires, L.; Bosco Sde, M.; Baptista, M.S.; Kurachi, C. Photodynamic therapy in Pythium insidiosum—An in vitro study of the correlation of sensitizer localization and cell death. PLoS ONE 2014, 9, e85431. [Google Scholar] [CrossRef] [PubMed]
  22. Kammarnjassadakul, P.; Rangsipanuratn, W.; Sriprapun, M.; Ngamsakulrungruj, P.; Palaga, T.; Sritunyalucksana, K.; Chindamporn, A. Cytochrome Oxidase 2 (COX2), β-Tubulin (TUB) and Chitin Synthase Subunit 2 (CHS2) Expression in Pythium insidiosum Thai Strains. Walailak J. Sci. Technol. 2021, 18, 9433–9438. [Google Scholar] [CrossRef]
  23. Lerksuthirat, T.; Lohnoo, T.; Inkomlue, R.; Rujirawat, T.; Yingyong, W.; Khositnithikul, R.; Phaonakrop, N.; Roytrakul, S.; Sullivan, T.D.; Krajaejun, T. The elicitin-like glycoprotein, ELI025, is secreted by the pathogenic oomycete Pythium insidiosum and evades host antibody responses. PLoS ONE 2015, 10, e0118547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Maeno, S.; Oie, Y.; Sunada, A.; Tanibuchi, H.; Hagiwara, S.; Makimura, K.; Nishida, K. Successful medical management of Pythium insidiosum keratitis using a combination of minocycline, linezolid, and chloramphenicol. Am. J. Ophthalmol. Case Rep. 2019, 15, 100498. [Google Scholar] [CrossRef]
  25. Gurnani, B.; Narayana, S.; Christy, J.; Rajkumar, P.; Kaur, K.; Gubert, J. Successful management of pediatric pythium insidiosum keratitis with cyanoacrylate glue, linezolid, and azithromycin: Rare case report. Eur. J. Ophthalmol. 2021, 11206721211006564. [Google Scholar] [CrossRef] [PubMed]
  26. Mercer, D.K.; Torres, M.D.T.; Duay, S.S.; Lovie, E.; Simpson, L.; von Kockritz-Blickwede, M.; de la Fuente-Nunez, C.; O’Neil, D.A.; Angeles-Boza, A.M. Antimicrobial Susceptibility Testing of Antimicrobial Peptides to Better Predict Efficacy. Front. Cell Infect. Microbiol. 2020, 10, 326. [Google Scholar] [CrossRef]
  27. Leekha, S.; Terrell, C.L.; Edson, R.S. General principles of antimicrobial therapy. Mayo. Clin. Proc. 2011, 86, 156–167. [Google Scholar] [CrossRef] [Green Version]
  28. Worasilchai, N.; Chindamporn, A.; Plongla, R.; Torvorapanit, P.; Manothummetha, K.; Chuleerarux, N.; Permpalung, N. In Vitro Susceptibility of Thai Pythium insidiosum Isolates to Antibacterial Agents. Antimicrob. Agents Chemother. 2020, 64, e02099-19. [Google Scholar] [CrossRef]
  29. Loreto, E.S.; Tondolo, J.S.; Pilotto, M.B.; Alves, S.H.; Santurio, J.M. New insights into the in vitro susceptibility of Pythium insidiosum. Antimicrob. Agents Chemother. 2014, 58, 7534–7537. [Google Scholar] [CrossRef] [Green Version]
  30. Loreto, E.S.; Tondolo, J.S.M.; Santurio, J.M.; Alves, S.H. Screening of antibacterial drugs for antimicrobial activity against Pythium insidiosum. Med. Mycol. 2019, 57, 523–525. [Google Scholar] [CrossRef]
  31. Mahl, D.L.; de Jesus, F.P.; Loreto, E.; Zanette, R.A.; Ferreiro, L.; Pilotto, M.B.; Alves, S.H.; Santurio, J.M. In vitro susceptibility of Pythium insidiosum isolates to aminoglycoside antibiotics and tigecycline. Antimicrob. Agents Chemother. 2012, 56, 4021–4023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Worasilchai, N.; Permpalung, N.; Chongsathidkiet, P.; Leelahavanichkul, A.; Mendoza, A.L.; Palaga, T.; Reantragoon, R.; Finkelman, M.; Sutcharitchan, P.; Chindamporn, A. Monitoring Anti-Pythium insidiosum IgG Antibodies and (1-->3)-beta-d-Glucan in Vascular Pythiosis. J. Clin. Microbiol. 2018, 56, e00610-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Richter, A.; Fessler, A.T.; Bottner, A.; Koper, L.M.; Wallmann, J.; Schwarz, S. Reasons for antimicrobial treatment failures and predictive value of in-vitro susceptibility testing in veterinary practice: An overview. Vet. Microbiol. 2020, 245, 108694. [Google Scholar] [CrossRef] [PubMed]
  34. Bassetti, M.; Montero, J.G.; Paiva, J.A. When antibiotic treatment fails. Intens Care Med. 2018, 44, 73–75. [Google Scholar] [CrossRef] [Green Version]
  35. Cicchese, J.M.; Dartois, V.; Kirschner, D.E.; Linderman, J.J. Both Pharmacokinetic Variability and Granuloma Heterogeneity Impact the Ability of the First-Line Antibiotics to Sterilize Tuberculosis Granulomas. Front. Pharmacol. 2020, 11, 333. [Google Scholar] [CrossRef] [Green Version]
  36. Shah, S.; Barton, G.; Fischer, A. Pharmacokinetic considerations and dosing strategies of antibiotics in the critically ill patient. J. Intensive Care Soc. 2015, 16, 147–153. [Google Scholar] [CrossRef] [Green Version]
  37. Veiga, R.P.; Paiva, J.A. Pharmacokinetics-pharmacodynamics issues relevant for the clinical use of beta-lactam antibiotics in critically ill patients. Crit. Care 2018, 22, 233. [Google Scholar] [CrossRef] [Green Version]
  38. Alghamdi, W.A.; Al-Shaer, M.H.; Klinker, K.P.; Peloquin, C.A. Variable linezolid exposure and response and the role of therapeutic drug monitoring: Case series. Clin. Case Rep. 2020, 8, 1126–1129. [Google Scholar] [CrossRef] [Green Version]
  39. Fugi, M.A.; Gunasekera, K.; Ochsenreiter, T.; Guan, X.; Wenk, M.R.; Maser, P. Genome profiling of sterol synthesis shows convergent evolution in parasites and guides chemotherapeutic attack. J. Lipid Res. 2014, 55, 929–938. [Google Scholar] [CrossRef] [Green Version]
  40. Levison, M.E.; Levison, J.H. Pharmacokinetics and pharmacodynamics of antibacterial agents. Infect. Dis. Clin. N. Am. 2009, 23, 791–815. [Google Scholar] [CrossRef] [Green Version]
  41. Jacobs, M.R. Optimisation of antimicrobial therapy using pharmacokinetic and pharmacodynamic parameters. Clin. Microbiol. Infect. 2001, 7, 589–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhang, R.; Qin, X.; Kong, F.; Chen, P.; Pan, G. Improving cellular uptake of therapeutic entities through interaction with components of cell membrane. Drug Deliv. 2019, 26, 328–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Lee, C.R.; Cho, I.H.; Jeong, B.C.; Lee, S.H. Strategies to minimize antibiotic resistance. Int. J. Environ. Res. Public. Health 2013, 10, 4274–4305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Actor, J.K. Innate Immunity. In Elsevier’s Integrated Review Immunology and Microbiology, 2nd ed.; Elsevier/Mosby: Philadelphia, PA, USA, 2012; pp. 43–51. [Google Scholar]
  45. Aristizábal, B.; González, A. Innate immune system. In Autoimmunity: From Bench to Bedside; Anaya, J.M., Shoenfeld, Y., Rojas-Villarraga, A., Levy, R.A., Cervera, R., Eds.; El Rosario University Press: Bogota, Colombia, 2013. [Google Scholar]
  46. Bergman, P.; Raqib, R.; Rekha, R.S.; Agerberth, B.; Gudmundsson, G.H. Host Directed Therapy against Infection by Boosting Innate Immunity. Front. Immunol. 2020, 11, 1209. [Google Scholar] [CrossRef] [PubMed]
  47. Gjini, E.; Brito, P.H. Integrating Antimicrobial Therapy with Host Immunity to Fight Drug-Resistant Infections: Classical vs. Adaptive Treatment. PLoS Comput. Biol. 2016, 12, e1004857. [Google Scholar] [CrossRef] [Green Version]
  48. Zheng, W.J.; Xu, Q.; Zhang, Y.Y.; E, X.F.; Gao, W.; Zhang, M.G.; Zhai, W.J.; Rajkumar, R.S.; Liu, Z.J. Toll-like receptor-mediated innate immunity against herpesviridae infection: A current perspective on viral infection signaling pathways. Virol. J. 2020, 17, 192. [Google Scholar] [CrossRef] [PubMed]
  49. Wongprompitak, P.; Pleewan, N.; Tantibhedhyangkul, W.; Chaiprasert, A.; Prabhasawat, P.; Inthasin, N.; Ekpo, P. Involvement of Toll-like receptor 2 on human corneal epithelium during an infection of Pythium insidiosum. Asian Pac. J. Allergy 2020, 38, 129–138. [Google Scholar] [CrossRef]
  50. Garrido-Mesa, J.; Rodriguez-Nogales, A.; Algieri, F.; Vezza, T.; Hidalgo-Garcia, L.; Garrido-Barros, M.; Utrilla, M.P.; Garcia, F.; Chueca, N.; Rodriguez-Cabezas, M.E.; et al. Immunomodulatory tetracyclines shape the intestinal inflammatory response inducing mucosal healing and resolution. Br. J. Pharmacol. 2018, 175, 4353–4370. [Google Scholar] [CrossRef]
  51. Yamauchi, K.; Shibata, Y.; Kimura, T.; Abe, S.; Inoue, S.; Osaka, D.; Sato, M.; Igarashi, A.; Kubota, I. Azithromycin suppresses interleukin-12p40 expression in lipopolysaccharide and interferon-gamma stimulated macrophages. Int. J. Biol. Sci. 2009, 5, 667–678. [Google Scholar] [CrossRef] [Green Version]
  52. Takahashi, G.; Sato, N.; Yaegashi, Y.; Kojika, M.; Matsumoto, N.; Kikkawa, T.; Shozushima, T.; Akitomi, S.; Aoki, K.; Ito, N.; et al. Effect of linezolid on cytokine production capacity and plasma endotoxin levels in response to lipopolysaccharide stimulation of whole blood. J. Infect. Chemother. 2010, 16, 94–99. [Google Scholar] [CrossRef]
  53. Pichereau, S.; Moran, J.J.; Hayney, M.S.; Shukla, S.K.; Sakoulas, G.; Rose, W.E. Concentration-dependent effects of antimicrobials on Staphylococcus aureus toxin-mediated cytokine production from peripheral blood mononuclear cells. J. Antimicrob. Chemother. 2012, 67, 123–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Lambers, C.; Burian, B.; Binder, P.; Ankersmit, H.J.; Wagner, C.; Muller, M.; Zeitlinger, M. Early immunomodulatory effects of linezolid in a human whole blood endotoxin model. Int. J. Clin. Pharmacol. Ther. 2010, 48, 419–424. [Google Scholar] [CrossRef]
  55. Hirata, N.; Hiramatsu, K.; Kishi, K.; Yamasaki, T.; Ichimiya, T.; Nasu, M. Pretreatment of mice with clindamycin improves survival of endotoxic shock by modulating the release of inflammatory cytokines. Antimicrob. Agents Chemother. 2001, 45, 2638–2642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Veringa, E.M.; Lambe, D.W., Jr.; Ferguson, D.A., Jr.; Verhoef, J. Enhancement of opsonophagocytosis of Bacteroides spp. by clindamycin in subinhibitory concentrations. J. Antimicrob. Chemother. 1989, 23, 577–587. [Google Scholar] [CrossRef] [PubMed]
  57. Trostdorf, F.; Reinert, R.R.; Schmidt, H.; Nichterlein, T.; Stuertz, K.; Schmitz-Salue, M.; Sadowski, I.; Bruck, W.; Nau, R. Quinupristin/dalfopristin attenuates the inflammatory response and reduces the concentration of neuron-specific enolase in the cerebrospinal fluid of rabbits with experimental Streptococcus pneumoniae meningitis. J. Antimicrob. Chemother. 1999, 43, 87–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Majumdar, S.; Dutta, K.; Manna, S.K.; Basu, A.; Bishayi, B. Possible protective role of chloramphenicol in TSST-1 and coagulase-positive Staphylococcus aureus-induced septic arthritis with altered levels of inflammatory mediators. Inflammation 2011, 34, 269–282. [Google Scholar] [CrossRef]
  59. Suschek, C.V.; Bonmann, E.; Kapsokefalou, A.; Hemmrich, K.; Kleinert, H.; Forstermann, U.; Kroncke, K.D.; Mahotka, C.; Kolb-Bachofen, V. Revisiting an old antimicrobial drug: Amphotericin B induces interleukin-1-converting enzyme as the main factor for inducible nitric-oxide synthase expression in activated endothelia. Mol. Pharmacol. 2002, 62, 936–946. [Google Scholar] [CrossRef] [Green Version]
  60. Mizuno, K.; Fukami, T.; Toyoda, Y.; Nakajima, M.; Yokoi, T. Terbinafine stimulates the pro-inflammatory responses in human monocytic THP-1 cells through an ERK signaling pathway. Life Sci. 2010, 87, 537–544. [Google Scholar] [CrossRef]
  61. Vora, S.; Purimetla, N.; Brummer, E.; Stevens, D.A. Activity of voriconazole, a new triazole, combined with neutrophils or monocytes against Candida albicans: Effect of granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor. Antimicrob. Agents Chemother. 1998, 42, 907–910. [Google Scholar] [CrossRef] [Green Version]
  62. Baltch, A.L.; Smith, R.P.; Franke, M.A.; Ritz, W.J.; Michelsen, P.B.; Bopp, L.H. Effects of cytokines and fluconazole on the activity of human monocytes against Candida albicans. Antimicrob. Agents Chemother. 2001, 45, 96–104. [Google Scholar] [CrossRef] [Green Version]
  63. Itaqui, S.R.; Verdi, C.M.; Tondolo, J.S.; da Luz, T.S.; Alves, S.H.; Santurio, J.M.; Loreto, E.S. In Vitro Synergism between Azithromycin or Terbinafine and Topical Antimicrobial Agents against Pythium insidiosum. Antimicrob. Agents Chemother. 2016, 60, 5023–5025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Jesus, F.P.; Ferreiro, L.; Bizzi, K.S.; Loreto, E.S.; Pilotto, M.B.; Ludwig, A.; Alves, S.H.; Zanette, R.A.; Santurio, J.M. In vitro activity of carvacrol and thymol combined with antifungals or antibacterials against Pythium insidiosum. J. Mycol. Med. 2015, 25, e89–e93. [Google Scholar] [CrossRef] [PubMed]
  65. Jesus, F.P.; Ferreiro, L.; Loreto, E.S.; Pilotto, M.B.; Ludwig, A.; Bizzi, K.; Tondolo, J.S.; Zanette, R.A.; Alves, S.H.; Santurio, J.M. In vitro synergism observed with azithromycin, clarithromycin, minocycline, or tigecycline in association with antifungal agents against Pythium insidiosum. Antimicrob. Agents Chemother. 2014, 58, 5621–5625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Chopra, I.; Roberts, M. Tetracycline antibiotics: Mode of action, applications, molecular biology, and epidemiology of bacterial resistance. Microbiol. Mol. Biol. Rev. 2001, 65, 232–260. [Google Scholar] [CrossRef] [Green Version]
  67. Nakao, C.; Angel, M.; Mateo, S.D.; Komesu, M.C. Effects of Topical Tetracycline in Wound Healing on Experimental Diabetes in Rats. Open Diabetes J. 2009, 2, 53–59. [Google Scholar] [CrossRef] [Green Version]
  68. Zimmermann, C.E.P.; Jesus, F.P.K.; Schlemmer, K.B.; Loreto, E.S.; Tondolo, J.S.M.; Driemeier, D.; Alves, S.H.; Ferreiro, L.; Santurio, J.M. In vivo effect of minocycline alone and in combination with immunotherapy against pythium insidiosum. Vet. Microbiol. 2020, 243, 108616. [Google Scholar] [CrossRef]
  69. Patel, P.H.; Hashmi, M.F. Macrolides; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar]
  70. Cramer, C.L.; Patterson, A.; Alchakaki, A.; Soubani, A.O. Immunomodulatory indications of azithromycin in respiratory disease: A concise review for the clinician. Postgrad. Med. 2017, 129, 493–499. [Google Scholar] [CrossRef]
  71. Allen, J.E.; Wynn, T.A. Evolution of Th2 immunity: A rapid repair response to tissue destructive pathogens. PLoS Pathog. 2011, 7, e1002003. [Google Scholar] [CrossRef] [Green Version]
  72. Chen, F.; Liu, Z.; Wu, W.; Rozo, C.; Bowdridge, S.; Millman, A.; Van Rooijen, N.; Urban, J.F., Jr.; Wynn, T.A.; Gause, W.C. An essential role for TH2-type responses in limiting acute tissue damage during experimental helminth infection. Nat. Med. 2012, 18, 260–266. [Google Scholar] [CrossRef]
  73. Kanoh, S.; Rubin, B.K. Mechanisms of action and clinical application of macrolides as immunomodulatory medications. Clin. Microbiol. Rev. 2010, 23, 590–615. [Google Scholar] [CrossRef] [Green Version]
  74. Jesus, F.P.; Loreto, E.S.; Ferreiro, L.; Alves, S.H.; Driemeier, D.; Souza, S.O.; Franca, R.T.; Lopes, S.T.; Pilotto, M.B.; Ludwig, A.; et al. In Vitro and In Vivo Antimicrobial Activities of Minocycline in Combination with Azithromycin, Clarithromycin, or Tigecycline against Pythium insidiosum. Antimicrob. Agents Chemother. 2016, 60, 87–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Bozdogan, B.; Appelbaum, P.C. Oxazolidinones: Activity, mode of action, and mechanism of resistance. Int. J. Antimicrob. Agents 2004, 23, 113–119. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, J.; Xia, L.; Wang, R.; Cai, Y. Linezolid and Its Immunomodulatory Effect: In Vitro and In Vivo Evidence. Front. Pharmacol. 2019, 10, 1389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ahirwar, L.K.; Kalra, P.; Sharma, S.; Mohamed, A.; Mittal, R.; Das, S.; Bagga, B. Linezolid shows high safety and efficacy in the treatment of Pythium insidiosum keratitis in a rabbit model. Exp. Eye Res. 2021, 202, 108345. [Google Scholar] [CrossRef] [PubMed]
  78. Schwarz, S.; Shen, J.; Kadlec, K.; Wang, Y.; Brenner Michael, G.; Fessler, A.T.; Vester, B. Lincosamides, Streptogramins, Phenicols, and Pleuromutilins: Mode of Action and Mechanisms of Resistance. Cold Spring Harb. Perspect. Med. 2016, 6, a027037. [Google Scholar] [CrossRef] [Green Version]
  79. Krause, K.M.; Serio, A.W.; Kane, T.R.; Connolly, L.E. Aminoglycosides: An Overview. Cold Spring Harb. Perspect. Med. 2016, 6, a027029. [Google Scholar] [CrossRef] [Green Version]
  80. McMeekin, D.; Mendoza, L. In vitro effect of streptomycin on clinical isolates of Pythium insidiosum. Mycologia 2000, 92, 371–373. [Google Scholar] [CrossRef]
  81. McOsker, C.C.; Fitzpatrick, P.M. Nitrofurantoin: Mechanism of action and implications for resistance development in common uropathogens. J. Antimicrob. Chemother. 1994, 33, 23–30. [Google Scholar] [CrossRef]
  82. Parenti, M.A.; Hatfield, S.M.; Leyden, J.J. Mupirocin: A topical antibiotic with a unique structure and mechanism of action. Clin. Pharm. 1987, 6, 761–770. [Google Scholar]
  83. Noor, A.; Preuss, C.V. Amphotericin B; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar]
  84. Triscott, J.A.; Weedon, D.; Cabana, E. Human subcutaneous pythiosis. J. Cutan. Pathol. 1993, 20, 267–271. [Google Scholar] [CrossRef]
  85. Krajaejun, T.; Sathapatayavongs, B.; Pracharktam, R.; Nitiyanant, P.; Leelachaikul, P.; Wanachiwanawin, W.; Chaiprasert, A.; Assanasen, P.; Saipetch, M.; Mootsikapun, P.; et al. Clinical and epidemiological analyses of human pythiosis in Thailand. Clin. Infect. Dis. 2006, 43, 569–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Sekhon, A.S.; Padhye, A.A.; Garg, A.K. In vitro sensitivity of Penicillium marneffei and Pythium insidiosum to various antifungal agents. Eur. J. Epidemiol. 1992, 8, 427–432. [Google Scholar] [CrossRef] [PubMed]
  87. Permpalung, N.; Worasilchai, N.; Manothummetha, K.; Torvorapanit, P.; Ratanawongphaibul, K.; Chuleerarux, N.; Plongla, R.; Chindamporn, A. Clinical outcomes in ocular pythiosis patients treated with a combination therapy protocol in Thailand: A prospective study. Med. Mycol. 2019, 57, 923–928. [Google Scholar] [CrossRef] [PubMed]
  88. Mesa-Arango, A.C.; Scorzoni, L.; Zaragoza, O. It only takes one to do many jobs: Amphotericin B as antifungal and immunomodulatory drug. Front. Microbiol. 2012, 3, 286. [Google Scholar] [CrossRef] [Green Version]
  89. Malone-Povolny, M.J.; Maloney, S.E.; Schoenfisch, M.H. Nitric Oxide Therapy for Diabetic Wound Healing. Adv. Healthc Mater. 2019, 8, e1801210. [Google Scholar] [CrossRef]
  90. Luo, J.D.; Chen, A.F. Nitric oxide: A newly discovered function on wound healing. Acta Pharmacol. Sin. 2005, 26, 259–264. [Google Scholar] [CrossRef] [Green Version]
  91. Vriens, K.; Kumar, P.T.; Struyfs, C.; Cools, T.L.; Spincemaille, P.; Kokalj, T.; Sampaio-Marques, B.; Ludovico, P.; Lammertyn, J.; Cammue, B.P.A.; et al. Increasing the Fungicidal Action of Amphotericin B by Inhibiting the Nitric Oxide-Dependent Tolerance Pathway. Oxid. Med. Cell Longev. 2017, 2017, 4064628. [Google Scholar] [CrossRef] [Green Version]
  92. Mesa-Arango, A.C.; Trevijano-Contador, N.; Roman, E.; Sanchez-Fresneda, R.; Casas, C.; Herrero, E.; Arguelles, J.C.; Pla, J.; Cuenca-Estrella, M.; Zaragoza, O. The production of reactive oxygen species is a universal action mechanism of Amphotericin B against pathogenic yeasts and contributes to the fungicidal effect of this drug. Antimicrob. Agents Chemother. 2014, 58, 6627–6638. [Google Scholar] [CrossRef] [Green Version]
  93. Ryder, N.S. Terbinafine: Mode of action and properties of the squalene epoxidase inhibition. Br. J. Dermatol. 1992, 126, 2–7. [Google Scholar] [CrossRef]
  94. Kanafani, Z.A.; Perfect, J.R. Antimicrobial resistance: Resistance to antifungal agents: Mechanisms and clinical impact. Clin. Infect. Dis. 2008, 46, 120–128. [Google Scholar] [CrossRef] [Green Version]
  95. Ghannoum, M.A.; Rice, L.B. Antifungal agents: Mode of action, mechanisms of resistance, and correlation of these mechanisms with bacterial resistance. Clin. Microbiol. Rev. 1999, 12, 501–517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Susaengrat, N.; Torvorapanit, P.; Plongla, R.; Chuleerarux, N.; Manothummetha, K.; Tuangsirisup, J.; Worasilchai, N.; Chindamporn, A.; Permpalung, N. Adjunctive antibacterial agents as a salvage therapy in relapsed vascular pythiosis patients. Int. J. Infect. Dis. 2019, 88, 27–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Cavalheiro, A.S.; Maboni, G.; de Azevedo, M.I.; Argenta, J.S.; Pereira, D.I.; Spader, T.B.; Alves, S.H.; Santurio, J.M. In Vitro activity of terbinafine combined with caspofungin and azoles against Pythium insidiosum. Antimicrob. Agents Chemother. 2009, 53, 2136–2138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Shenep, J.L.; English, B.K.; Kaufman, L.; Pearson, T.A.; Thompson, J.W.; Kaufman, R.A.; Frisch, G.; Rinaldi, M.G. Successful medical therapy for deeply invasive facial infection due to Pythium insidiosum in a child. Clin. Infect. Dis. 1998, 27, 1388–1393. [Google Scholar] [CrossRef] [Green Version]
  99. Sucher, A.J.; Chahine, E.B.; Balcer, H.E. Echinocandins: The newest class of antifungals. Ann. Pharmacother. 2009, 43, 1647–1657. [Google Scholar] [CrossRef]
  100. Lamaris, G.A.; Lewis, R.E.; Chamilos, G.; May, G.S.; Safdar, A.; Walsh, T.J.; Raad, I.I.; Kontoyiannis, D.P. Caspofungin-mediated beta-glucan unmasking and enhancement of human polymorphonuclear neutrophil activity against Aspergillus and non-Aspergillus hyphae. J. Infect. Dis. 2008, 198, 186–192. [Google Scholar] [CrossRef] [Green Version]
  101. Katragkou, A.; Roilides, E.; Walsh, T.J. Role of Echinocandins in Fungal Biofilm-Related Disease: Vascular Catheter-Related Infections, Immunomodulation, and Mucosal Surfaces. Clin. Infect. Dis. 2015, 61 (Suppl. 6), S622–S629. [Google Scholar] [CrossRef] [Green Version]
  102. Zanette, R.A.; Jesus, F.P.; Pilotto, M.B.; Weiblen, C.; Potter, L.; Ferreiro, L.; Alves, S.H.; Santurio, J.M. Micafungin alone and in combination therapy with deferasirox against Pythium insidiosum. J. Mycol. Med. 2015, 25, 91–94. [Google Scholar] [CrossRef]
  103. Pereira, D.I.; Santurio, J.M.; Alves, S.H.; Argenta, J.S.; Potter, L.; Spanamberg, A.; Ferreiro, L. Caspofungin in vitro and in vivo activity against Brazilian Pythium insidiosum strains isolated from animals. J. Antimicrob. Chemother. 2007, 60, 1168–1171. [Google Scholar] [CrossRef] [Green Version]
  104. Polak, A.M. Preclinical data and mode of action of amorolfine. Clin. Exp. Dermatol. 1992, 17 (Suppl. 1), 8–12. [Google Scholar] [CrossRef]
  105. Ianiski, L.B.; Stibbe, P.C.; Denardi, L.B.; Weiblen, C.; Soares, M.P.; Valente, J.S.S.; Sangioni, L.A.; Pereira, D.I.B.; Santurio, J.M.; Botton, S.A. In vitro anti-Pythium insidiosum activity of amorolfine hydrochloride and azithromycin, alone and in combination. Med. Mycol. 2021, 59, 67–73. [Google Scholar] [CrossRef]
  106. Sahoo, B.M.; Ravi Kumar, B.V.V.; Sruti, J.; Mahapatra, M.K.; Banik, B.K.; Borah, P. Drug Repurposing Strategy (DRS): Emerging Approach to Identify Potential Therapeutics for Treatment of Novel Coronavirus Infection. Front. Mol. Biosci. 2021, 8, 628144. [Google Scholar] [CrossRef] [PubMed]
  107. Yacouba, A.; Olowo-Okere, A.; Yunusa, I. Repurposing of antibiotics for clinical management of COVID-19: A narrative review. Ann. Clin. Microbiol. Antimicrob. 2021, 20, 37. [Google Scholar] [CrossRef] [PubMed]
  108. Kaul, G.; Shukla, M.; Dasgupta, A.; Chopra, S. Update on drug-repurposing: Is it useful for tackling antimicrobial resistance? Future Microbiol. 2019, 14, 829–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Stokes, M.; Abdijadid, S. Disulfiram; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar]
  110. Krajaejun, T.; Lohnoo, T.; Yingyong, W.; Rujirawat, T.; Kumsang, Y.; Jongkhajornpong, P.; Theerawatanasirikul, S.; Kittichotirat, W.; Reamtong, O.; Yolanda, H. The Repurposed Drug Disulfiram Inhibits Urease and Aldehyde Dehydrogenase and Prevents In Vitro Growth of the Oomycete Pythium insidiosum. Antimicrob. Agents Chemother. 2019, 63, e00609-19. [Google Scholar] [CrossRef] [Green Version]
  111. 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]
  112. Li, J.; Zheng, S.; Chen, B.; Butte, A.J.; Swamidass, S.J.; Lu, Z. A survey of current trends in computational drug repositioning. Brief Bioinform. 2016, 17, 2–12. [Google Scholar] [CrossRef] [Green Version]
  113. Ko, Y. Computational Drug Repositioning: Current Progress and Challenges. Appl. Sci. 2020, 10, 5076. [Google Scholar] [CrossRef]
  114. Rudrapal, M.; Khairnar, S.; Jadhav, A. Drug Repurposing (DR): An Emerging Approach in Drug Discovery. In Drug—Hypothesis, Molecular Aspects and Therapeutic Applications; BoD—Books on Demand: London, UK, 2020. [Google Scholar]
  115. Liu, Y.; Tong, Z.; Shi, J.; Li, R.; Upton, M.; Wang, Z. Drug repurposing for next-generation combination therapies against multidrug-resistant bacteria. Theranostics 2021, 11, 4910–4928. [Google Scholar] [CrossRef]
  116. Cheng, Y.S.; Williamson, P.R.; Zheng, W. Improving therapy of severe infections through drug repurposing of synergistic combinations. Curr. Opin. Pharmacol. 2019, 48, 92–98. [Google Scholar] [CrossRef]
  117. Nguyen, T.K.; Selvanayagam, R.; Ho, K.K.K.; Chen, R.; Kutty, S.K.; Rice, S.A.; Kumar, N.; Barraud, N.; Duong, H.T.T.; Boyer, C. Co-delivery of nitric oxide and antibiotic using polymeric nanoparticles. Chem. Sci. 2016, 7, 1016–1027. [Google Scholar] [CrossRef] [Green Version]
  118. Rouillard, K.R.; Novak, O.P.; Pistiolis, A.M.; Yang, L.; Ahonen, M.J.R.; McDonald, R.A.; Schoenfisch, M.H. Exogenous Nitric Oxide Improves Antibiotic Susceptibility in Resistant Bacteria. ACS Infect. Dis. 2021, 7, 23–33. [Google Scholar] [CrossRef] [PubMed]
  119. Available online: https://www.sanfordguide.com/ (accessed on 13 January 2022).
  120. Keeratijarut, A.; Lohnoo, T.; Rujirawat, T.; Yingyong, W.; Kalambaheti, T.; Miller, S.; Phuntumart, V.; Krajaejun, T. The Immunoreactive Exo-1,3-beta-Glucanase from the Pathogenic Oomycete Pythium insidiosum Is Temperature Regulated and Exhibits Glycoside Hydrolase Activity. PLoS ONE 2015, 10, e0135239. [Google Scholar] [CrossRef] [PubMed]
  121. Wittayapipath, K.; Laolit, S.; Yenjai, C.; Chio-Srichan, S.; Pakarasang, M.; Tavichakorntrakool, R.; Prariyachatigul, C. Analysis of xanthyletin and secondary metabolites from Pseudomonas stutzeri ST1302 and Klebsiella pneumoniae ST2501 against Pythium insidiosum. BMC Microbiol. 2019, 19, 78. [Google Scholar] [CrossRef] [PubMed]
  122. Subramaniam, S.; Joyce, P.; Thomas, N.; Prestidge, C.A. Bioinspired drug delivery strategies for repurposing conventional antibiotics against intracellular infections. Adv. Drug Deliv. Rev. 2021, 177, 113948. [Google Scholar] [CrossRef]
  123. Stocco, G.; Lucafo, M.; Decorti, G. Pharmacogenomics of Antibiotics. Int. J. Mol. Sci. 2020, 21, 5975. [Google Scholar] [CrossRef]
Figure 1. Photograph of human pythiosis. A 46-year-old Thai male with thalassemia was diagnosed with vascular pythiosis. CTA showed the occlusion of the right aorta, and ELISA showed the positive IgG against P. insidiosum (with permission). Abbreviations: CTA, computed tomography angiography; ELISA, enzyme-linked immunosorbent assay.
Figure 1. Photograph of human pythiosis. A 46-year-old Thai male with thalassemia was diagnosed with vascular pythiosis. CTA showed the occlusion of the right aorta, and ELISA showed the positive IgG against P. insidiosum (with permission). Abbreviations: CTA, computed tomography angiography; ELISA, enzyme-linked immunosorbent assay.
Antibiotics 11 00450 g001
Figure 2. Mode of MIC value of each antibacterial/antifungal class against P. insidiosum isolates reviewed in previous publications. Mode of MIC value of P. insidiosum isolates against antimicrobial drugs in class different antimicrobial classes: tetracyclines (4 μg/mL) [28,29], macrolides (6 μg/mL) [29], oxazolidinones (8 μg/mL) [29], lincosamides (4 μg/mL) [30], streptogramins (2 μg/mL) [30], phenicols (16 μg/mL) [30], aminoglycosides (64 μg/mL) [31], nitrofurantoin (no data) [30], mupirocin (4 μg/mL) [29], polyenes (64 μg/mL) [29], allylamines and azoles (4 μg/mL) [9], and echinocandins (4 μg/mL) [32].
Figure 2. Mode of MIC value of each antibacterial/antifungal class against P. insidiosum isolates reviewed in previous publications. Mode of MIC value of P. insidiosum isolates against antimicrobial drugs in class different antimicrobial classes: tetracyclines (4 μg/mL) [28,29], macrolides (6 μg/mL) [29], oxazolidinones (8 μg/mL) [29], lincosamides (4 μg/mL) [30], streptogramins (2 μg/mL) [30], phenicols (16 μg/mL) [30], aminoglycosides (64 μg/mL) [31], nitrofurantoin (no data) [30], mupirocin (4 μg/mL) [29], polyenes (64 μg/mL) [29], allylamines and azoles (4 μg/mL) [9], and echinocandins (4 μg/mL) [32].
Antibiotics 11 00450 g002
Figure 3. Antimicrobial treatment in the management of P. insidiosum infection. Antibacterial and antifungal drugs exhibit immunomodulation activity and can improve treatment strategies for human pythiosis. Several mechanisms contribute to antimicrobial failure during the treatment of diseases.
Figure 3. Antimicrobial treatment in the management of P. insidiosum infection. Antibacterial and antifungal drugs exhibit immunomodulation activity and can improve treatment strategies for human pythiosis. Several mechanisms contribute to antimicrobial failure during the treatment of diseases.
Antibiotics 11 00450 g003
Table 1. Summary of methods for determining MICs of antimicrobial drugs against P. insidiosum.
Table 1. Summary of methods for determining MICs of antimicrobial drugs against P. insidiosum.
Antimicrobial ClassDrugMIC Determination Method(s)Reference(s)
TetracyclinesTetracyclineBroth microdilution[28]
TigecyclineBroth microdilution, disk diffusion, and Etest[28,29,31]
MinocyclineBroth microdilution, disk diffusion, and Etest[28,29]
MacrolidesAzithromycinBroth microdilution, disk diffusion, and Etest[28,29]
ClarithromycinBroth microdilution, disk diffusion, and Etest[28,29]
OxazolidinonesLinezolidBroth microdilution, disk diffusion, and Etest[29]
LincosamidesClindamycinBroth dilution[30]
StreptograminsQuinupristin and dalfopristinBroth dilution[30]
PhenicolsChloramphenicol Broth dilution[30]
AminoglycosidesGentamicinBroth microdilution[31]
NeomycinBroth microdilution[31]
ParomomycinBroth microdilution[31]
StreptomycinBroth microdilution[31]
NitrofurantoinBroth dilution [30]
MupirocinBroth microdilution, disk diffusion, and Etest[29]
PolyenesAmphotericin BEtest[29]
AllylaminesTerbinafine Broth dilution and radial growth[9]
AzolesMiconazole Broth microdilution[9]
Ketoconazole Broth microdilution[9]
Fluconazole Broth microdilution and agar diffusion[9]
Itraconazole Broth microdilution, radial growth, and agar diffusion[9]
Posaconazole Broth microdilution and agar diffusion[9]
Voriconazole Broth microdilution, radial growth, and agar diffusion[9]
EchinocandinsCaspofungin Broth dilution[32]
Anidulafungin Broth dilution[32]
Abbreviations: MIC, minimal inhibitory concentration.
Table 2. Immunomodulatory effects of antimicrobials.
Table 2. Immunomodulatory effects of antimicrobials.
Antimicrobial ClassDrugImmunopharmacological EffectReference(s)
TetracyclinesTigecycline, minocycline Potentiate the innate immune response and augment resolution of inflammation[50]
MacrolidesAzithromycinReduce the production of IL-12, resulting in enhanced Th2 response[51]
OxazolidinonesLinezolidSuppress synthesis of proinflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, IL-8, interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α)[52,53,54]
LincosamidesClindamycinSuppress the release of inflammatory cytokines such as TNF-α and IL-1β and enhance the phagocytosis of microorganisms by host cells[55,56]
StreptograminsQuinupristin-dalfopristinDecrease the concentration of pro-inflammatory cell wall components (lipoteichoic acid and teichoic acid) and the activity of TNF[57]
PhenicolsChloramphenicolElevate the anti-inflammatory IL-10 levels[58]
PolyenesAmphotericin BActivate the host’s innate immunity and augment the IL-1β-induced inducible nitric-oxide synthase (iNOS) expression and the production of nitric oxide (NO)[59]
AllylaminesTerbinafineStimulate proinflammatory cytokines[60]
AzolesFluconazole, voriconazoleEnhance microbicidal activity of monocytes, macrophages, and neutrophils[61,62]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Medhasi, S.; Chindamporn, A.; Worasilchai, N. A Review: Antimicrobial Therapy for Human Pythiosis. Antibiotics 2022, 11, 450. https://doi.org/10.3390/antibiotics11040450

AMA Style

Medhasi S, Chindamporn A, Worasilchai N. A Review: Antimicrobial Therapy for Human Pythiosis. Antibiotics. 2022; 11(4):450. https://doi.org/10.3390/antibiotics11040450

Chicago/Turabian Style

Medhasi, Sadeep, Ariya Chindamporn, and Navaporn Worasilchai. 2022. "A Review: Antimicrobial Therapy for Human Pythiosis" Antibiotics 11, no. 4: 450. https://doi.org/10.3390/antibiotics11040450

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