Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Nov 6, 2023; 11(31): 7530-7542
Published online Nov 6, 2023. doi: 10.12998/wjcc.v11.i31.7530
Antibiotic resistance in patients with liver cirrhosis: Prevalence and current approach to tackle
Valentina Liakina, Clinic of Gastroenterology, Nephrourology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 01513, Lithuania
Valentina Liakina, Department of Chemistry and Bioengineering, Faculty of Fundamental Sciences, Vilnius Tech, Vilnius 10223, Lithuania
ORCID number: Valentina Liakina (0000-0001-8685-1292).
Author contributions: Liakina V created the study conception and design, performed literature search and analysis, drafting and editing of the manuscript.
Conflict-of-interest statement: The author has nothing to declare.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Valentina Liakina, PhD, Senior Researcher, Clinic of Gastroenterology, Nephrourology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 21, Ciurlionio Street, Vilnius 01513, Lithuania. valentina.liakina@santa.lt
Received: August 31, 2023
Peer-review started: August 31, 2023
First decision: September 25, 2023
Revised: October 2, 2023
Accepted: October 17, 2023
Article in press: October 17, 2023
Published online: November 6, 2023
Processing time: 67 Days and 0.1 Hours

Abstract

Regardless of etiology, complications with bacterial infection in patients with cirrhosis are reported in the range of 25%-46% according to the most recent data. Due to frequent episodes of bacterial infection and repetitive antibiotic treatment, most often with broad-spectrum gram negative coverage, patients with cirrhosis are at increased risk of encountering multidrug resistant bacteria, and this raises concern. In such patients, extended-spectrum beta-lactamase and AmpC-producing Enterobacterales, methicillin- or vancomycin-resistant Staphylococcus aureus, vancomycin-resistant Enterococci, carbapenem-resistant Pseudomonas aeruginosa, and Acinetobacter baumannii, all of which are difficult to treat, are the most common. That is why novel approaches to the prophylaxis and treatment of bacterial infections to avoid antibiotic resistance have recently been developed. At the same time, our knowledge of resistance mechanisms is constantly updated. This review summarizes the current situation regarding the burden of antibiotic resistance, including the prevalence and mechanisms of intrinsic and acquired resistance in bacterial species that most frequently cause complications in patients with liver cirrhosis and recent developments on how to deal with multidrug resistant bacteria.

Key Words: Cirrhosis, Bacterial complications, Antibiotics, Intrinsic resistance, Acquired resistance, Probiotics, Vaccines

Core Tip: This review presents current data on bacterial complications in patients with cirrhosis. A comprehensive analysis of the prevalence of antibiotic resistant isolates with a brief presentation of intrinsic and acquired resistance mechanisms was performed in the most prevalent pathobionts causing infections in cirrhosis. Current approved and developing options to treat bacterial complications to avoid resistance are also discussed.



INTRODUCTION

Numerous clinical studies have revealed that, in patients with liver disease, bacterial complications manifest mainly in the advanced stage, as classified by Child-Pugh[1,2]. Their prevalence does not vary significantly depending on the etiology of cirrhosis[3] and is reported in the range of 25% to 46% according to the most recent data[4,5]; 44.4% of such patients suffer more than one episode[3].

The development of bacterial infection can accelerate the course of liver disease at any stage[6]. Regardless of the severity of liver disease itself, it can decompensate liver function and significantly increase the mortality rate[3,7-9]. The 30-mo survival rate in cases of cirrhosis with bacterial complications has been reported to be around half that of those without complications (34% vs 62%, respectively)[7]. Furthermore, lethal outcomes occurred more frequently when infection was caused by multidrug-resistant bacteria (MDR) than non-MDR bacteria (72% vs 28%, respectively)[10]. According to an international expert proposal, MDR was defined as acquired nonsusceptibility to at least one agent in three or more antimicrobial categories[11].

Due to frequent episodes of bacterial infection and repetitive antibiotic treatment, most often with broad-spectrum Gram negative coverage, patients with cirrhosis are at a higher risk of encountering MDR bacteria[5]. Due to the frequent need for invasive procedures, such as large-volume paracentesis or varices ligations, approximately 37% of patients with cirrhosis are readmitted to the hospital within 30 d[12], while hospitalization itself can be considered a risk factor since it can involve infection with bacteria spread in hospital units, including their MDR representatives[5].

Many research efforts have recently been directed towards developing strategies for the prevention of bacterial complications and demonstrating more efficient methods for the treatment of bacterial infections that avoid contributing to resistance. At the same time, our knowledge of resistance mechanisms is constantly updated. The above considerations led to this summary of the current situation with respect to the burden of antibiotic resistance in patients with liver cirrhosis.

In this review, the prevalence and mechanisms of antibiotic resistance in bacterial species that most commonly cause complications in patients with cirrhosis are discussed, alongside recent developments on how to cope with multidrug-resistant bacterial infections.

THE MOST COMMON BACTERIAL COMPLICATIONS IN PATIENTS WITH CIRRHOSIS
The prevalence of bacterial complications

Lymph fluid leakage into the peritoneal cavity (ascites) and bleeding of the esophageal varices are often diagnosed in patients with advanced cirrhosis. Ascites in combination with increased intestinal permeability and impaired venous blood flow comprise favorable conditions for bacterial growth. What is important to stress is that, in such patients, bacterial infection can manifest without an obvious source. Most often (around one third of patients with cirrhosis) spontaneous bacterial peritonitis (SBP) occurs[2]. When ascites is diagnosed, the 5-year survival rate is 30%-40%[13]. Acute decompensation is strongly associated with bacterial infection (22.3% of cases), especially when accompanied by acute-on-chronic liver failure (44% of cases)[14].

In addition to commonly diagnosed SBP, urinary tract infections (12%-25%) and pneumonia (15%-20%) are often present in patients with cirrhosis[3,4,15]. Less frequently, skin and soft tissue infections (5%-10%) and other miscellaneous infections (approximately 12%) can manifest[2-4]. In approximately 10% of patients, a life-threatening condition develops, such as spontaneous bacteremia[16,17]. The reported 30-day mortality rate of such patients is extremely high: 45%-53%[10,17]. Although rarely, spontaneous bacterial empyema (SBE) – an infection of preexisting hepatic hydrothorax fluid – most frequently caused by Ecsherichia coli or Klebsiella pneumoniae can occur[18].

The common sources of infection agents

Although infection agents are most commonly community-acquired (in 30% to 50% of cases)[4], due to the frequent need for various invasive procedures, such as peritoneocentesis, endoscopic ligations of the varices, intrahepatic transjugular shunt, percutaneous treatment, etc. – patients are at a higher risk of acquiring healthcare-associated or nosocomial infections. According to a study by Fernández et al[19], 35% of patients with cirrhosis were diagnosed with nosocomial infections vs 5% of patients without cirrhosis. This finding has also been confirmed by others, suggesting that healthcare-associated infections develop in approximately one third of such patients[4,20].

Commensal gut bacteria can also be the cause of bacterial complications. Inflammatory-like abnormalities of the intestinal mucosa were found in two-thirds of patients with cirrhosis, and this effect becomes more pronounced with disease progression[21]. Patients with cirrhosis are also characterized by prolonged intestinal transit and bacterial overgrowth in the small intestine[22]. These abnormalities are aggravated by impaired immunity (so- called cirrhosis associated immune dysfunction), which is characterized by reduced leukocyte count due to hypersplenism, reduced production of innate immunity proteins such as complement, and general exhaustion of immune cells despite activation of their pro-inflammatory state[4,6,23]. Due to the synergistic effect of the aforementioned conditions, the intestinal barrier is compromised, and this facilitates the translocation of intestinal bacteria into the mesenteric lymph nodes, causing SBP, endotoxemia, or bacteremia[24,25]. The activation of pro-inflammatory cytokines and the tumor necrosis factor alpha facilitates secondary infections and contributes to sepsis-related organ failure[26]. Additionally, portosystemic collaterals slow down the clearance of bacteria and their metabolites from circulation[4,10].

Gram-negative intestinal bacteria are more likely to translocate than Gram-positive (approximately 60% vs approximately 40%)[3,22,27]. The most common Gram-negative pathobionts that cause infections in patients with cirrhosis belong to the Enterobacterales family (Escherichia coli, Proteus spp., Klebsiella pneumoniae), while Gram-positive pathobionts belong to the Enterococcaceae or Staphylococcaceae families (Staphylococcus aureus, Enterococci)[4,10]. Fungal infections are less common (approximately 4%)[20,27].

ANTIBIOTIC RESISTANCE PATHWAYS AND MECHANISMS
Two antibiotic resistance pathways in bacteria

Because many antibiotics are naturally occurring compounds that bacteria are exposed to throughout their evolution, bacteria have evolved a series of structures and metabolic processes that allow them to survive in antibiotic-enriched environments. Many known antibiotics were mainly discovered as natural metabolites synthesized by Gram-positive bacteria or fungi. For example, Amycolatopsis orientalis synthesizes vancomycin; Streptomyces spp. – tetracycline; Micromonospora purpurea – gentamicin; Amycolatopsis rifamycinica – rifamycins; while carbapenems were developed from thienamycin – the naturally derived metabolite of Streptomyces cattleya[28]. Seventeen years after penicillin was isolated from the Penicillium spp. fungus, cephalosporin was discovered in Acremonium spp.[29].

Intrinsic or inherited antibiotic resistance pathways are usually encoded in the bacterial chromosome and passed on to their offspring (Table 1, Figure 1A)[30-33].

Figure 1
Figure 1 Antibiotic resistance pathways in bacteria. A: Intrinsic resistance mechanisms. 1: Decrease in bacterial wall permeability: suppression of porin expression, 2: Increase in efflux: activation of efflux pumps, 3: Alteration of antibiotic targets, 4: Antibiotic inactivation by enzymatic modifications, 5: Antibiotic hydrolysis; B: Acquired resistance mechanisms. 1: Decrease in bacterial wall permeability: mutations of porin-coding genes, 2: Increase in efflux: enhance efflux pump gene expression by acquiring additional genes via external DNA, 3: Alteration of antibiotic targets: mutations of genes that encode antibiotic targets, 4: Acquisition of additional ARGs via external DNA (plasmids and others), 5: Antibiotic hydrolysis: broadening of the substrate specificity of hydrolyses.
Table 1 Examples of intrinsic (natural) antibiotic resistance pathways in bacteria.
Resistance mechanism
Molecule/process involved
Resistance to antibiotic
Primary host
Genome-encoded
Plasmid-encoded
Decrease in bacterial wall permeabilityReduced porin expressionDifferent antibioticsGram-negative bacteriaoprD geneNo
Increase in effluxTripartite RND pump familyDifferent antibioticsGram-negative bacteriaYesYes
Antibiotic inactivation by chemical group transferModification AminoglycosidesGram-negative and Gram-positive bacteriaYesYes
OxidationTetracyclines Many bacteriaTet(X) genesYes
Antibiotic hydrolysis AmpC beta-lactamasesBroad-spectrum beta-lactamsGram-negative bacteria: EnterobacteriaceaeYesYes
CarbapenemasesA variety of beta-lactamsEnterobacteriaceaeYesYes
Modification of antibiotic targetModification of peptidoglycan precursorsGlycopeptides: vancomycinGram-positive cocci: EnterococcaceaevanC, vanD operonsvanA gene claster
Alteration of LPSPolymyxins: colistinGram-negative: EnterobacteriaceaeYesYes
Exchange of conventional PBP to PBP2aBeta-lactamsStaphylococcaceaemecA geneYes

Under antibiotic pressure, a bacterium can enhance its resistance by mutations in existing genes. For example, in Gram-negative bacteria, resistance to multiple antibiotics has been developed by loss or mutation of the oprD gene, which encodes an outer membrane porin (Table 2, Figure 1B)[30-33]. This can be called acquired resistance[30,31]. Antibiotic resistance genes (ARGs) can also be carried out by plasmids. In this form, they can easily be transmitted between bacteria, whether of the same species or between different species. In general, plasmids, bacteriophages, and extracellular deoxyribonucleic acid (DNA) are the three main pathways of horizontal gene transfer in bacteria through the processes of conjugation, transduction, and natural transformation, respectively[34]. Conjugation is cell-to-cell contact through sexual pili, allowing bacteria to share plasmids or other mobile DNA elements, including ARGs. Bacteria can share genetic information through the transduction process, which is mediated by bacteriophages. Naturally transformable bacteria are able to take up short fragments of naked DNA from their environment.

Table 2 Examples of acquired antibiotic resistance in bacteria.
Metabolic process
Molecule/reaction involved
Resistance to antibiotic
Host
Genome encoded
Plasmid encoded
Decrease in bacterial wall permeabilityModified porinsMany antibioticsGram-negative bacteriaLoss or mutated oprD mutations in porin operon
Increase in effluxRND pump familyMany antibioticsGram-negative bacteria; S. aureusMutations in mtr region, mexEF-oprNRND with NDM-1
Efflux pumpFluoroquinolonesS. aureusAmplification of norA gene
Enzymatic antibiotic inactivation OxidationTetracyclinesMany bacteriaMobile Tet(X)
Antibiotic hydrolysisESBLsBroad-spectrum beta-lactamsGram-negative bacteriaYesYes
Modification of antibiotic targetRNA polymerase RpoBRifampicinS. aureusMutated RpoB gene
DNA gyrase GyrA and topoisomerase IV ParCQuinolones FluoroquinolonesS. aureus; K. pneumoniaeQRDRYes
DHPS and DHFRTrimethoprim–sulfamethoxazoleGram-negative bacteriaMutated dfrA geneYes
Altered rRNAMacrolides: erythromycin, Oxazolidinones: linezolidStaphylococcus spp.; Streptococcus spp.Mutated rRNA operonYes
Modification of peptidoglycan precursorsVancomycinEnterococcaceae, StaphyloccaceaeMutated van genes, vanR/van S controlsYes
Enoyl-ACP reductaseTriclosanP. aeruginosaAlternative fabV gene
Addition of moieties to LPSColistinEnterobacter spp.Mutated TSC genesmcr
Transpeptidase moiety of PBPMethicillin and other beta-lactams Staphylococcus spp.mecA geneSCCmec
Dihydropteroate synthaseSulphonamidesMutated sul1/2 gene
Mechanisms and influencing factors of antibiotic resistance

Resistance to antibiotics can be achieved by decreasing the permeability of the bacterial wall, controlling the intracellular antibiotic concentration to a harmless level by accelerating the efflux pumps or by enzymatic modification of the antibiotic or its target[30,31]. In this article, a brief overview of intrinsic and acquired antibiotic resistance pathways is presented (Tables 1 and 2), while a detailed description of these mechanisms is well explored in several reviews[30-33].

The mechanisms of antibiotic resistance themselves cannot be significantly influenced by humans. What can be influenced is the selection of antibiotic resistant species under human-driven antibiotic pressures – either in the environment (such as through agricultural activity) or in the human organism (during antibiotic treatment). The use of oral or injected antibiotics for the treatment of any disease is associated with direct selection pressure on the commensal microbiota of any location in the body, primarily the digestive tract. Just one year after the introduction of semisynthetic penicillin, methicillin-resistant S. aureus (MRSA) emerged due to the selection of strains with the mecA gene, which is responsible for methicillin resistance[30]. After introducing vancomycin for the treatment of MRSA, vancomycin-resistant S. aureus (VRSA) was detected shortly thereafter[30].

Bacteria are not only capable of modifying an antibiotic target, but can also use target bypass strategies, as in the case of MRSA. Here, the conventional penicillin binding protein (PBP; an enzyme involved in peptidoglycan biosynthesis) changes to exogenous PBP – PBP2a that is homologous to the original target, but with a lower affinity for β-lactams[33].

Gram-positive bacteria in general are more susceptible to different antibiotics, but can acquire resistance through mutations in their genome or horizontal gene transfer[30]. For example, Boekhoud et al[35] have reported a Clostridioides difficile isolate resistant to metronidazole due to the plasmid pCD-METRO.

Many bacteria carry multiple MDR efflux pump genes in their chromosomes and can acquire them as plasmids. A tripartite resistance nodulation division pump has been found to be carried by a plasmid, with genes encoding the antibiotic-targeting enzyme New Delhi metalo-beta-lactamase 1 (Table 2)[30]. Although overproduction of the MDR efflux pump is a less effective way to diminish the toxic effect of antibiotics compared to enzymatic alteration of antibiotics or their target, bacteria can achieve resistance to a whole class of antimicrobial molecules through multiple biochemical pathways involving different resistance mechanisms[36].

Resistance to antibiotics is not always associated with ARGs

To survive in an antibiotic-enriched environment, bacteria can engage a mechanism of switching between metabolic stages – from the normal, susceptible cell type to the tolerant or persister state[37]. The ability of a bacterium to survive high concentrations of bactericidal agents to which it is fully susceptible is called antibiotic persistence[30]. Unlike resistant strains, which can grow under antibiotic pressure, persisters do not grow. Instead, they switch to a stringent response stage, minimizing metabolic processes that include blockage of transcription and DNA replication[37]. Although in different proportions (from 0.001% to 1%), persister cells have been identified in almost all examined bacterial species, including Gram-negative and Gram-positive bacteria[37]. Antibiotic persistence is thought to be involved in developing resistance to antibiotics and maintaining chronic bacterial infections. Chronic infection is assumed to be unresolved by a host’s immune system due to a population of persisters[30].

Another known mechanism, originally discovered in bacteria as a plasmid maintenance tool[38], is the toxin-antitoxin system (TA), which potentially contributes to both the transition to the persister stage and the development of antibiotic resistance[39]. A toxin is usually a protein that is capable of inhibiting or modifying essential cell processes – such as mRNA transcription and translation, DNA replication, or cell wall functioning – in response to threatening conditions. An antitoxin (usually noncoding RNA) inhibits the cognate toxin or degrades its mRNA when there is no need for a stringent response. The antitoxin is encoded in the same TA module as the toxin. Bacteria can possess several different TA modules encoded in their genome or plasmids, and they are activated under specific stresses, such as the appearance of a bactericidal agent in the environment. Conjugative plasmids carrying ARGs and TAs are considered to have the most efficient pathway for the dissemination of antibiotic resistance between bacteria[39]. A recent comprehensive genetic study of the most prevalent MDR bacteria, Escherichia coli and Klebsiella pneumoniae, elucidated the complexity of the resistance acquisition process, in which the adoption of ARG-carrying plasmids is facilitated by mutation in the core metabolic genes[40]. Advanced genomic approaches will undoubtedly shed light on other options for bacteria to survive in a toxin-enriched environment.

RECENT RECOMMENDATIONS FOR PROPHYLAXIS AND TREATMENT OF BACTERIAL COMPLICATIONS IN CIRRHOSIS

Antibiotics, having antibacterial and germicidal effects, can not only effectively prevent and treat bacterial infection, but can also decrease the incidence of further decompensation of cirrhosis[41]. The most common recommendation for the treatment of patients with advanced liver disease aggravated by bacterial complications is the use of broad-spectrum antibiotics such as penicillin derivatives (amoxicillin-clavulanate) and cephalosporin derivatives (cefotaxime, ceftriaxone), quinolones (norfloxacin, ofloxacin), or, less commonly, aminoglycosides (neomycin) and trimethoprim/sulfamethoxazole[26].

Third-generation cephalosporins (ceftriaxone, cefotaxime) are recommended to be started empirically (before obtaining culture results) in all patients with suspected SBP or SBE when the polymorphonuclear cell count in the ascites or pleural fluid is > 250/mm3, however, the risk of MDR isolates must be considered, as cephalosporins have become less effective in settings where MDR bacteria are prevalent[42].

Although there is a risk of the emergence of isolates resistant to quinolones, the most recent recommendation for the primary prophylaxis of SBP in patients with ascites is the use of oral norfloxacin[6,42]. Norfloxacin or ciprofloxacin is also recommended for secondary prophylaxis[42]. When ascites is accompanied by gastrointestinal hemorrhage, ceftriaxone, cefotaxime, and piperacillin-tazobactam, along with albumin injections are recommended[42,43]. Third-generation cephalosporins are also the drug of choice for the treatment of bacteremia, pneumonia, and soft tissue infections in conjunction with penicillin derivatives and macrolides[4,42]. In the case of nosocomial infection, treatment with a broader spectrum antibiotic (piperacillin-tazobactam, vancomycin, carbapenems, tetracyclines or trimethoprim/sulfamethoxazole) is recommended since the third-generation cephalosporins, quinolones, and amoxicillin/clavulanic acid are ineffective in these patients[4,15,26,42].

MDR BACTERIA BURDEN IN PATIENTS WITH CIRRHOSIS

Delayed antibiotic treatment is associated with an increased risk of mortality, in particular in patients with septic shock[4]. On the other hand, conventional strategies of antibiotic use carry the danger of selection of resistant commensal bacteria in the gut or elsewhere in the body. Currently, in patients with cirrhosis, approximately 30%-40% of infections are caused by MDR bacteria[27]. Among the main sources of bacterial infection in patients with cirrhosis, healthcare-associated and nosocomial origins are characterized by the highest prevalence of antibiotic-resistant species – 35% and 14%, respectively – while among community-acquired species only 4% appear to be MDR[19].

To avoid resistance, new and existing antibacterial regimens are constantly being developed for the treatment of bacterial infections and prophylaxis. In case of suspected SBP, recent AASL guidelines for the management of ascites strongly recommend narrowing the coverage of antibiotics as soon as the culture results are available, and restricting the primary prophylaxis of SBP with antibiotics to patients with very advanced cirrhosis[42]. Considering the risk of bacterial complications originating from the intestinal microflora, non-absorbable oral antibiotics are chosen more frequently for the prophylaxis of infections and can improve short-term survival in high-risk patients[24,44]. However, resistance can also develop to such antibiotics. For example, after rifaximin prophylaxis of overt hepatic encephalopathy, Staphylococcus spp. resistant to rifaximin was detected in 50% of patients[45]. Rifaximin per os can also modify intestinal microflora: a significant increase in Eubacteriaceae and Propionibacterium and a decrease in the abundance of Veillonellaceae, Roseburia, and Blautia have been reported[46].

In contrast, a study reported that rifaximin treatment led to an increase in the abundance of potentially beneficial taxa and a decrease in Klebsiella spp. resistomes (resistome – a set of genes responsible for antibiotic resistance), as well as antibiotic-resistant Gram-negative bacteria[47].

Use of broad-spectrum antibiotics diminishes the diversity of the gut microbiota, promotes changes in the proportion of bacterial communities, and decreases the total abundance of fungi[47,48]. Bajaj et al[49] reported a significant reduction in Sclerodermataceae, Dothideomycetes, and Saccharomyces boulardii and a substantial increase in Candida in patients with cirrhosis after treatment with broad-spectrum antibiotics. They also detected a decrease in the Basidiomycota/Ascomycete ratio and concluded that broad-spectrum antibiotics can alter the balanced bacterial and fungal communities. This is one of the main risk factors for fungal infections.

In a recent study by Shamsaddini et al[47] among 163 outpatients with cirrhosis and 40 healthy control subjects, the abundance of ARGs in the gut microbiota was found to be higher than in the control samples and worsened with the severity of the cirrhosis. Furthermore, the abundance of ARGs predicts a poor prognosis and increases the mortality rate. In the patients enrolled, resistome-associated pathobionts belonging to Enterobacterales, Streptococcus spp., Enterococcus spp. and Acinetobacter spp. had already been detected at admission. The resistome pattern included genes resistant to β-lactamases, macrolides, quinolones, aminoglycosides, tetracyclines, fosfomycin, and rifamycin. During hospitalization, three patients became infected with methicillin-resistant Staphylococcus aureus, three with Candida spp. and one with Streptococcus viridans. In this study, treatment with rifaximin only minimally affected the prevalence of resistome[47].

The extensive use of broad-spectrum antibiotics in healthcare settings has established conditions under which MDR bacteria have spread. The most common of these are extended-spectrum beta-lactamase producing Enterobacterales; non-fermentative Gram-negative bacilli (i.e., Pseudomonas aeruginosa, Stenotrophomonas maltophilia, and Acinetobacter baumannii); carbapenemase-producing Enterobacterales; methicillin-resistant (MRSA) or VRSA Staphylococcus aureus; vancomycin-susceptible or resistant Enterococci (VSE or VRE); and extensively drug-resistant (XDR) Mycobacterium tuberculosis[26]. XDR was defined as non-susceptibility to at least one agent in all but two or fewer antimicrobial categories (i.e. bacterial isolates remain susceptible to only one or two categories)[11].

In frequently hospitalized patients with cirrhosis, extended spectrum beta lactamase and AmpC-producing Enterobacterales (that is, Escherichia coli, Klebsiella pneumoniae), methicillin- or vancomycin-resistant Staphylococcus aureus, and vancomycin-resistant Enterococci, all of which are difficult to treat, are the most common[4]. XDR bacteria such as carbapenemase-producing Enterobacterales, carbapenem-resistant Pseudomonas aeruginosa, and Acinetobacter baumannii are also found as causative agents of bacterial complications in such patients[4].

In a study by Milovanovic et al[10] involving 85 patients with cirrhosis and bacteremia, the most common pathogen was Enterococcus spp. (32%), followed by methicillin-susceptible Staphylococcus aureus (15%) and Escherichia coli (14%), and 37% of all isolated bacteria appeared to be multidrug resistant. Specifically, 73% of MDR bacteria were resistant to ampicillin, 70% to amoxicillin, and 51% to amoxicillinclavulanic acid. Resistance to third- and fourth-generation cephalosporins was detected in 44% to 49% of MDR bacteria. This group exhibited unexpectedly high resistance to amikacin (54%) and trimethoprim/sulfamethoxazole (51%). Resistance to meropenem was 20%, and 15% for imipenem, resistance to glycopeptide vancomycin was 15.4%, and 45% for teicoplanin. MDR bacteria was an independent predictor of mortality (OR = 6.2), and the proportion of MDR isolates increased with disease progression[10]. The authors emphasized that, even in developed countries with well-established healthcare systems, MDR bacteria significantly increased the mortality rate in patients with cirrhosis[10].

A worldwide study of 1302 hospitalized patients with cirrhosis from 46 centers in Asia, Europe, and North and South America also found that up to 34% of bacterial infections were caused by MDR species[2], with the highest prevalence in patients from India[2]. The most common species were extended-spectrum beta-lactamase-producing Enterobacterales, MRSA, vancomycin-resistant Enterococci, Pseudomonas aeruginosa, and Acinetobacter baumannii[2].

In a prospective study conducted almost 10 years ago by Fernández et al[19] among 233 patients with cirrhosis, only 18% of bacterial infections (most commonly SBP, urinary tract infections and pneumonia) were caused by MDR species. Again, the most common were extended-spectrum beta-lactamase-producing Enterobacterales followed by Pseudomonas aeruginosa, methicillin-resistant Staphylococcus aureus and Enterococcus faecium. MDR bacteria caused septic shock and mortality twice as often as non-MDR bacteria. In particular, when the authors repeated this study 4 years later, they obtained the same results[19].

During recent decades, the prevalence of infections due to MDR and XDR bacteria among patients with cirrhosis has increased[20,50]. The CANONIC Study group reported an increase in MDR infections in patients with cirrhosis in Europe –from 29% in 2011 to 38% in 2017 and 2018[50]. Long-term quinolones for the prophylaxis of SBP have been found to be strongly associated with continuously increasing rates of MDR pathobionts (approaching 40%), and carry an inherent risk of SBP breakthrough[51]. An increase in the proportion of Gram-positive MDR bacterial infections has also been observed[4]. The widespread use of third-generation cephalosporins has contributed to the emergence of nosocomial and healthcare-associated enterococcal infections resistant to cephalosporins[4].

CURRENTLY APPROVED ANTIBIOTIC-FREE PROPHYLAXIS OF BACTERIAL COMPLICATIONS

Because bacteria are so powerfully equipped with survival tools to help them when faced with antibiotics, the best way to avoid the selection of antibiotic-resistant bacteria is by reducing antibiotic use. This is difficult to implement if the infection has already manifested, while antibiotic prophylaxis should be restricted to the subgroup of patients who are at a very high risk of infection based on the availability of evidence[5,6,42,52]. For example, previously recommended for the treatment of hepatic encephalopathy[53], non-absorbable rifaximin has been proposed for SBP prophylaxis as an alternative to norfloxacin to avoid selection of isolates resistant to quinolones. Rifaximin has been shown to reduce the risk of SBP in patients with hepatic encephalopathy, but prophylaxis has not yet been confirmed[4].

By reducing portal hypertension, non-selective beta-blockers contribute to diminishing the risk of variceal bleeding and ascites formation[6]. Beta-blockers have also been shown to reduce intestinal bacteria translocation and therefore lower the risk of SBP, but they have no influence on urinary or respiratory tract infections[4].

To prevent refractory septic shock in patients with acute decompensation stage cirrhosis, a low-dose steroids prescription has been shown to be appropriate to monitor adrenal insufficiency that often appears in such patients[4].

Human albumin is not only the main regulator of blood oncotic pressure but is also characterized by scavenging and immunomodulatory activity. Albumin treatment has been shown to restore immune dysfunction in cirrhosis[4]. Along with antibiotics, albumin injections are recommended for SBP treatment, especially for patients with kidney dysfunction[6,42]. Furthermore, since albumin binds to some antibiotics, which keeps them in circulation longer, hypoalbuminemia promotes the clearance of antibiotics from plasma[26]. For example, ceftriaxone is 83%-96% bound to albumin, flucloxacillin 95%, cephalothin 55%-75%, aztreonam 60%, carbapenems 85%-95% (ertapenem) and 96%-99%(faropenem) and vancomycin 30%-60%[54]. Therefore, even when faced non-MDR bacteria characterized by a low minimal inhibitory concentration, hypoalbuminemia can prevent patients from maintaining an effective antibiotic concentration in the blood over time and thus can decrease the therapeutic effect of antibiotics[26]. In this case, a higher-than-routinely-prescribed dose of antibiotics is required.

However, the therapeutic effect of time-dependent antibiotics, such as those belonging to the ß-lactams family, strongly depends on the clearance rate. Due to the disruption of blood flow through the liver caused by fibrosis and portosystemic shunts, the drugs metabolized in the liver remain in the bloodstream for a longer period of time, leading to higher concentrations of antibiotics, which persist for longer. For this reason, in addition to hypoalbuminemia, blood flow and kidney impairment should also be taken into account when prescribing a particular antibiotic course to a patient[54]. Since empirical dosing regimens of medications are usually derived from studies of healthy populations, optimizing the antibacterial dosage for patients with cirrhosis is mandatory, considering hypoalbuminemia, hepatic blood flow impairment, the presence of portosystemic shunts, and renal dysfunction, according to Zoratti et al[26].

FECAL MICROBIOTA TRANSPLANTATION (FMT) TO REDUCE THE BURDEN OF MDR BACTERIA

Currently, fecal microbiota transplantation (FMT) has been approved for the treatment of recurrent Clostridioides difficile infection that does not respond to metronidazole or vancomycin. In two recent clinical trials, it was found that FMT reduced the abundance of antibiotic resistance genes in the microbiota of patients with cirrhosis[55]. After one single administration of an FMT capsule, a lower abundance of vancomycin, beta-lactamase, and rifamycin ARGs was detected after 4 wk, compared to baseline and placebo levels[55]. These findings suggest that FMT offers a promising therapy that may reduce the population of multidrug resistant organisms. However, it is too early to implement FMT therapy to reduce the risk of MDR infections because several important issues remain unsolved.

First, FMT safety issues have not yet been fully addressed. Since patients with cirrhosis have compromised intestinal barriers, it is necessary to ensure that only the “healthy” microbiota is transplanted. Therefore, a methodology is needed to select donors. It would be desirable to know which composition of the donor microbiota performs the most effective therapeutic function and then standardize the supply of donor material. A methodology for screening the donor microbiota for potential pathobionts and their metabolites should be established to avoid transplantation of MDR bacteria and other dangerous pathobionts to the recipient. Even if this were to be the case, there would be a risk of transplanting potentially pathogenic microbes for which screening techniques are not yet available, or some undefined molecules presented in donor feces which could trigger an adverse immune response. For example, in a study by Bajaj et al[55], MDR Escherichia coli was transplanted from one donor to two recipients during the FMT procedure.

Second, it remains unclear what administration route (enemas, capsules, etc.) and dosage of FMT is necessary to achieve a therapeutic response, how many treatment courses should be applied and how long it is necessary to wait between courses[55].

PREBIOTICS AND PROBIOTICS CAN REDUCE THE BURDEN OF MDR BACTERIA

In many studies, although usually without strong evidence-based argumentation due to methodological flaws, it has been proven that high-fiber foods such as whole grains and various vegetables maintain the intestine microbiota in healthy shape due to the abundance of prebiotics in their composition[56]. Prebiotics promote an increase in the proportion of beneficial gut bacteria such as Bifidobacterium longum. At the same time, fermentation of prebiotics by the gut microbiota produces short-chain fatty acids such as acetate, butyrate, and propionate. In particular, butyrate has an anti-inflammatory effect and promotes the reestablishment of the intestinal barrier[57]. A randomized perspective study of healthy adults has shown that a diet enriched with prebiotics improves intestinal microbial diversity and relieves the pro-inflammatory immune response[58]. Short-chain fatty acids also have a positive effect on liver lipid metabolism. Treatment with oligosaccharides such as inulin-type fructans considerably improves liver steatosis in patients with cirrhosis[59].

The beneficial effects of probiotics on the gut microbiota have been widely known for a considerable period of time. This is why probiotic-rich foods such as kefir, kimchi, kombucha, pickles, buttermilk, cottage cheese, tempeh, sauerkraut, and miso soup are widely recommended as components of a healthy diet[57]. Pharmaceutical forms of probiotics, such as Lactobacillus plantarum, Lactobacillus brevis, Leuconostoc mesenteroides, Lactobacillus bulgaricus, and Streptococcus thermophilus, are often prescribed in combination with prebiotics to restore a bacterial community after antibiotic treatment. However, because of the different composition of such preparations, not only at the species level but also at the strain level, their efficacy can vary considerably. This circumstance makes it very difficult to assess and compare the results of clinical trials on such products, therefore, the confidence level of evidence of such trials is still low[60].

It is assumed that probiotics reduce the ability of pathological bacteria to interact with the intestinal epithelium, and thus prevent the translocation of bacteria and their products through the intestinal barrier. Synbiotic treatment (a pharmaceutical form of combining prebiotics and probiotics) has been shown to ensure intact intestinal permeability by maintaining immunoglobulin A (IgA) production, promoting the integrity of gut lining cells with fully functioning tight junctions, promoting Lactobacillus spp. growth and inhibiting the overgrowth of potentially pathogenic Escherichia coli and Staphylococcal species[61]. The above-mentioned effect of synbiotics has a general beneficial effect on liver and intestinal function, and therefore can contribute to reducing the damage caused by MDR bacteria. The meta-analysis conducted by Cai et al[62], involving 826 patients with cirrhosis in total, revealed that probiotics had better efficacy than FMT in the prevention of hepatic encephalopathy, decreasing serum levels of ammonia, endotoxemia levels, and hospitalization rates. However, a randomized clinical trial with 116 participants enrolled concluded that they were not effective against the intestinal microbiota resistome[63]. While in vitro and animal studies demonstrate the promising effects of Lactobacilli and Bifidobacterium on the decrease of pathogenic bacteria in the intestine and urinary tract, including Helicobacter pylori, Salmonella and some MDR bacteria such as Escherichia coli and Klebsiella pneumoniae[64,65]. Through the secretion of antibacterial chemicals, including lactic acid and hydrogen peroxide, probiotics in combination with antibiotics have been shown to enhance the therapeutic effects of the latter[66,67].

OTHER PERSPECTIVES ON COPING WITH MDR BACTERIA

Nanoparticles may serve as an imperative tool for fighting antibiotic resistance because they bypass toxin-resistant bacteria systems. In particular, silver and gold nanoparticles in combination with metal oxide nanoparticles demonstrate promising, although nonspecific, bactericidal effects on Gram-positive and Gram-negative bacteria in vitro. This includes different MDR and XDR bacteria such as Mycobacterium tuberculosis, Acinetobacter baumannii, Pseudomonas aeruginosa, MRSA, Escherichia coli, Enterococcus faecalis, Streptococcus pyogenes, Salmonella typhi and Klebsiella pneumoniae, bearing NDM-1, VRE, and VRSA[66].

Currently, there are no single published study or ongoing clinical trial on nanoparticle treatment or its use in the prophylaxis of bacterial complications in cirrhosis. In the experimental study by Yang et al[68], a biliary stent coated with silver nanoparticles was not only characterized by antibacterial activity but also served longer without obstruction. Metallic and metal oxide nanoparticles have been externally studied for their use in combating nosocomial bacteria, which are highly resistant to antibiotics in clinical settings[69].

Because of their small dimensions, nanoparticles anchor and easily penetrate the bacterial wall, alter the permeability of the cell membrane, promote free radical production, destroy bacterial DNA and operate in other ways to disrupt the metabolism of a bacterium, ultimately causing cell death[66]. However, due to many toxicological issues that remain unsolved, the treatment of bacterial infections with nanoparticles remains a theoretical prospect[70,71]. Although, if designed with specific surface functions to enhance their therapeutic effect and diminish toxicity, nanoparticles can be effective at least externally for antibiotic-resistant oral and wound biofilms treatment[71].

Host defense peptides, the cationic amphipathic peptides like difensins and cathelicidins, and antimicrobial peptides such as polylysine are currently under investigation as potential agents against MDR bacteria. These peptides are naturally synthesized in many organisms, including humans, in response to infection. Having low cellular toxicity, they are active against gram-negative and gram-positive bacteria, including MDR representatives. Furthermore, they are effective against fungi and biofilms, which are difficult to treat with antibiotics[66].

Another promising strategy for combating MDR bacteria is bacteriophage therapy. Bacteriophages are a natural component of the intestinal microflora. They infect bacteria, causing cell lysis when in their virulent state, or they can switch to a temperate state via incorporation of their genome into the bacterial chromosome or by forming a self-replicating plasmid[12,72]. The therapeutic potential of bacteriophages is due to their affinity for specific bacteria species. Therefore, its action can be accurately predicted to target a specific bacterium. However, the clinical implementation of bacteriophage therapy faces a number of challenges. First, bacteriophages have been shown to trigger a pro-inflammatory immune response[73]. Second, due to their narrow host range, bacteriophages have limited therapeutic utility in the event of encountering an infection caused by a number of different bacteria, although this can be overcome through the use of phage cocktails. Finally, bacteriophages can carry genes that are harmful to human cells. Hence, the preparation of a safe bacteriophage cocktail for clinical applications is a considerable challenge[66,72]. Additionally, phages can be ineffective[74].

However, progress in biotechnologies allows for the production of recombinant phage products, genomic-editing tools can be used to construct genetically manipulated phages, or engineered phages with the required traits in mind[74]. For example, phage lysins have been found to be solely capable of lysing bacterial cells. In experimental studies, the use of phage lysins in combination with antibiotics appeared to be more effective in eradicating bacterial infections than antibiotics alone[72].

New natural antimicrobials are also constantly sought that are effective against MDR bacteria[75].

As vaccines do not develop resistance, the invention of vaccines against MDR bacteria is also considered an option to reduce the burden of MDR bacteria[76]. Especially effective vaccines would be greatly appreciated against such notorious bugs as Staphylococcus aureus (MRSA and VRSA), MDR and XDR isolates of Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterococcus faecium, Clostridioides difficile.

Especially challenging is developing vaccines against representatives of the commensal flora, as they are in constant contact with the human immune system. Given that the physiology of an ex vivo bacterium may differ from its behavior in vivo, this makes the task even more complicated. Furthermore, some bacteria, such as Staphylococcus aureus, an ubiquitous and in general inoffensive colonizer of human skin and nasal passage, can escape immune attack and remain viable even if it is ingested by neutrophils or macrophages[77]. To date, all candidate vaccines against this pathogen have been discontinued at or before Phase 3 clinical development due to ineffectiveness or side effects[78-80].

The worldwide spread of carbapenem resistant and even colistin resistant Klebsiella pneumoniae isolates is stimulating the development of a vaccine against this pathogen, but recombinant and other vaccine candidates have been tested in experimental models and remain in the preclinical or early clinical phases so far[81,82]. The same is true regarding a vaccine against extraintestinal pathogenic Escherichia coli[79,80].

Acinetobacter baumannii and Clostridioides difficile are ubiquitous in the environment but are not part of normal human flora. Therefore, a vaccine against these pathogens is technically feasible, but no vaccine has been approved for clinical use to date[79,80].

Therapeutic monoclonal antibodies against commensal flora such as Staphylococcus aureus, Pseudomonas aeruginosa, and others are also being investigated. The function of such antibodies is to neutralize bacterial toxins and thus reduce the pathological effects of certain bacteria[83]. Molecules such as virulence factors secreted by staphylococci (enterotoxin serotype B, alpha-hemolysin, etc.) are promising candidates for the development of therapeutic antibodies, but none have yet been approved for clinical use[83]. For example, a chimeric murine/human monoclonal antibody Pagibaximab has been developed to prevent staphylococcal sepsis in low-birthweight infants and showed > 90% efficacy against the most prevalent CoNS isolates in preclinical studies, however, appeared ineffective in the randomized, double-blind, multicenter, placebo-controlled clinical trial (ClinicalTrials.gov Identifier: NCT00646399).

A method of developing a vaccine that does not target a specific bacterium but enhances mucosal immunity in general has shown promise. Uromune, a sublingual vaccine for the prophylaxis of recurrent urinary tract infections, has shown tangible benefits over antibiotics in clinical trials and is already on the market[84,85]. Taking into account the experience gained in vaccine development and the use of modern biotechnological methods, more effective vaccines can be expected to become available in the near future[86].

CONCLUSION

Recent data on the development of MDR bacterial complications in cirrhotic patients prompt the use of antibiotic-free prophylaxis strategies. Some, such as the combination of probiotics/prebiotics and human albumin injections, appear promising. Effective vaccines and therapeutic antibodies can also be expected in the near future.

Currently, there is no effective alternative for the treatment of bacterial infections, and antibiotics remain the first choice. However, there is room to improve existing regimens by applying a personalized approach. The selection of MDR bacteria can occur not only as a result of the activity of ARGs, but also as a result of inadequate therapeutic concentrations of antibiotics in the body of a patient. This may occur due to hypoalbuminemia, hepatic blood flow disturbances, portosystemic shunts, renal impairment, or a combination of the above factors. Thus, individualized regimens and monitoring of antibiotic concentration according to patient conditions can significantly improve the effectiveness of antibiotic treatment and can reduce the selection of MDR bacteria during the treatment of patients with cirrhosis.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Lithuania

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Fan YC, China; Gao S, China S-Editor: Liu JH L-Editor: A P-Editor: Zhao S

References
1.  Caly WR, Strauss E. A prospective study of bacterial infections in patients with cirrhosis. J Hepatol. 1993;18:353-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 347]  [Cited by in F6Publishing: 324]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
2.  Piano S, Singh V, Caraceni P, Maiwall R, Alessandria C, Fernandez J, Soares EC, Kim DJ, Kim SE, Marino M, Vorobioff J, Barea RCR, Merli M, Elkrief L, Vargas V, Krag A, Singh SP, Lesmana LA, Toledo C, Marciano S, Verhelst X, Wong F, Intagliata N, Rabinowich L, Colombato L, Kim SG, Gerbes A, Durand F, Roblero JP, Bhamidimarri KR, Boyer TD, Maevskaya M, Fassio E, Kim HS, Hwang JS, Gines P, Gadano A, Sarin SK, Angeli P; International Club of Ascites Global Study Group. Epidemiology and Effects of Bacterial Infections in Patients With Cirrhosis Worldwide. Gastroenterology. 2019;156:1368-1380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 262]  [Article Influence: 52.4]  [Reference Citation Analysis (0)]
3.  Papp M, Norman GL, Vitalis Z, Tornai I, Altorjay I, Foldi I, Udvardy M, Shums Z, Dinya T, Orosz P, Lombay B Jr, Par G, Par A, Veres G, Csak T, Osztovits J, Szalay F, Lakatos PL. Presence of anti-microbial antibodies in liver cirrhosis--a tell-tale sign of compromised immunity? PLoS One. 2010;5:e12957.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
4.  Piano S, Tonon M, Angeli P. Changes in the epidemiology and management of bacterial infections in cirrhosis. Clin Mol Hepatol. 2021;27:437-445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 30]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
5.  Mitra M, Mancuso A, Politi F, Maringhini A. Bacterial infections in cirrhosis: A narrative review and key points for clinical practice. ITJM. 2020;14:126-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
6.  European Association for the Study of the Liver. EASL Clinical Practice Guidelines for the management of patients with decompensated cirrhosis. J Hepatol. 2018;69:406-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1177]  [Cited by in F6Publishing: 1515]  [Article Influence: 252.5]  [Reference Citation Analysis (2)]
7.  Dionigi E, Garcovich M, Borzio M, Leandro G, Majumdar A, Tsami A, Arvaniti V, Roccarina D, Pinzani M, Burroughs AK, O'Beirne J, Tsochatzis EA. Bacterial Infections Change Natural History of Cirrhosis Irrespective of Liver Disease Severity. Am J Gastroenterol. 2017;112:588-596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 74]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
8.  Borzio M, Salerno F, Piantoni L, Cazzaniga M, Angeli P, Bissoli F, Boccia S, Colloredo-Mels G, Corigliano P, Fornaciari G, Marenco G, Pistarà R, Salvagnini M, Sangiovanni A. Bacterial infection in patients with advanced cirrhosis: a multicentre prospective study. Dig Liver Dis. 2001;33:41-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 277]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
9.  Tandon P, Garcia-Tsao G. Bacterial infections, sepsis, and multiorgan failure in cirrhosis. Semin Liver Dis. 2008;28:26-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in F6Publishing: 357]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
10.  Milovanovic T, Pantic I, Velickovic J, Oluic B, Vlaisavljevic Z, Dragasevic S, Stojkovic Lalosevic M, Dumic I. Bacteremia in patients with liver cirrhosis in the era of increasing antimicrobial resistance: single-center epidemiology. J Infect Dev Ctries. 2021;15:1883-1890.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Magiorakos AP, Srinivasan A, Carey RB, Carmeli Y, Falagas ME, Giske CG, Harbarth S, Hindler JF, Kahlmeter G, Olsson-Liljequist B, Paterson DL, Rice LB, Stelling J, Struelens MJ, Vatopoulos A, Weber JT, Monnet DL. Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria: an international expert proposal for interim standard definitions for acquired resistance. Clin Microbiol Infect. 2012;18:268-281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6072]  [Cited by in F6Publishing: 7690]  [Article Influence: 591.5]  [Reference Citation Analysis (0)]
12.  Edwards LA, Goldenberg SD, Shawcross DL. Meeting the Challenge of Antimicrobial Resistance in Cirrhosis: The Invisible Threat That Lies Within. Gastroenterology. 2021;161:413-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
13.  Bonacini M. Diagnosis and management of cirrhosis in coinfected patients. J Acquir Immune Defic Syndr. 2007;45 Suppl 2:S38-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
14.  Trebicka J, Fernandez J, Papp M, Caraceni P, Laleman W, Gambino C, Giovo I, Uschner FE, Jansen C, Jimenez C, Mookerjee R, Gustot T, Albillos A, Bañares R, Jarcuska P, Steib C, Reiberger T, Acevedo J, Gatti P, Shawcross DL, Zeuzem S, Zipprich A, Piano S, Berg T, Bruns T, Danielsen KV, Coenraad M, Merli M, Stauber R, Zoller H, Ramos JP, Solé C, Soriano G, de Gottardi A, Gronbaek H, Saliba F, Trautwein C, Kani HT, Francque S, Ryder S, Nahon P, Romero-Gomez M, Van Vlierberghe H, Francoz C, Manns M, Garcia-Lopez E, Tufoni M, Amoros A, Pavesi M, Sanchez C, Praktiknjo M, Curto A, Pitarch C, Putignano A, Moreno E, Bernal W, Aguilar F, Clària J, Ponzo P, Vitalis Z, Zaccherini G, Balogh B, Gerbes A, Vargas V, Alessandria C, Bernardi M, Ginès P, Moreau R, Angeli P, Jalan R, Arroyo V; PREDICT STUDY group of the EASL-CLIF CONSORTIUM. PREDICT identifies precipitating events associated with the clinical course of acutely decompensated cirrhosis. J Hepatol. 2021;74:1097-1108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 132]  [Article Influence: 44.0]  [Reference Citation Analysis (0)]
15.  Vorobioff JD, Contreras F, Tanno F, Hernández L, Bessone F, Colombato L, Adi J, Fassio E, Felgueres M, Fernández G, Gaite L, Gibelli D, Darrichon HG, Lafage M, Lombardo D, López S, Mateo A, Mendizábal M, Pecoraro J, Ruf A, Ruiz P, Severini J, Stieben T, Sixto M, Zárate F, Barraza SB, Sierra ID, Pacheco VR, Roblero JP, Rojas JO, González PR, Rodríguez DSM, Sierralta A, Manchego AU, Valdes E, Yaquich P, Wolff R, Valdivia FB, Gallegos RC, Galloso R, Marcelo JS, Montes P, Tenorio L, Veramendi I, Alava E, Armijos X, Benalcazar G, Carrera E, Pazmiño GF, Díaz EM, Garassini M, Marrero RP, Infante M, Suárez DP, Gutiérrez JC, Reyes CMV, Serrano YM, Hernández RH, Martínez OM, González TP, Andara MT, Hernández MS, Gerona S, García I, Tijera F, López EP, Torres K, Garzón M. A Latin American survey on demographic aspects of hospitalized, decompensated cirrhotic patients and the resources for their management. Ann Hepatol. 2020;19:396-403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
16.  Yang Q, Jiang XZ, Zhu YF, Lv FF. Clinical risk factors and predictive tool of bacteremia in patients with cirrhosis. J Int Med Res. 2020;48:300060520919220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
17.  Thulstrup AM, Sørensen HT, Schønheyder HC, Møller JK, Tage-Jensen U. Population-based study of the risk and short-term prognosis for bacteremia in patients with liver cirrhosis. Clin Infect Dis. 2000;31:1357-1361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 92]  [Article Influence: 3.8]  [Reference Citation Analysis (1)]
18.  Rusk A, Daniels C. Spontaneous bacterial empyema: a dangerous complication of hepatic hydrothorax. Chest. 2020;158:Suppl. A1235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
19.  Fernández J, Acevedo J, Castro M, Garcia O, de Lope CR, Roca D, Pavesi M, Sola E, Moreira L, Silva A, Seva-Pereira T, Corradi F, Mensa J, Ginès P, Arroyo V. Prevalence and risk factors of infections by multiresistant bacteria in cirrhosis: a prospective study. Hepatology. 2012;55:1551-1561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 399]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
20.  Onorato L, Monari C, Capuano S, Grimaldi P, Coppola N. Prevalence and Therapeutic Management of Infections by Multi-Drug-Resistant Organisms (MDROs) in Patients with Liver Cirrhosis: A Narrative Review. Antibiotics (Basel). 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
21.  De Palma GD, Rega M, Masone S, Persico F, Siciliano S, Patrone F, Matantuono L, Persico G. Mucosal abnormalities of the small bowel in patients with cirrhosis and portal hypertension: a capsule endoscopy study. Gastrointest Endosc. 2005;62:529-534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 123]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
22.  Wiest R, Garcia-Tsao G. Bacterial translocation (BT) in cirrhosis. Hepatology. 2005;41:422-433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 492]  [Cited by in F6Publishing: 484]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
23.  Albillos A, Lario M, Álvarez-Mon M. Cirrhosis-associated immune dysfunction: distinctive features and clinical relevance. J Hepatol. 2014;61:1385-1396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 691]  [Cited by in F6Publishing: 749]  [Article Influence: 74.9]  [Reference Citation Analysis (1)]
24.  Saitoh O, Sugi K, Lojima K, Matsumoto H, Nakagawa K, Kayazawa M, Tanaka S, Teranishi T, Hirata I, Katsu Ki KI. Increased prevalence of intestinal inflammation in patients with liver cirrhosis. World J Gastroenterol. 1999;5:391-396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
25.  Kronsten VT, Tranah TH, Pariante C, Shawcross DL. Gut-derived systemic inflammation as a driver of depression in chronic liver disease. J Hepatol. 2022;76:665-680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 34]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
26.  Zoratti C, Moretti R, Rebuzzi L, Albergati IV, Di Somma A, Decorti G, Di Bella S, Crocè LS, Giuffrè M. Antibiotics and Liver Cirrhosis: What the Physicians Need to Know. Antibiotics (Basel). 2021;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
27.  Piano S, Brocca A, Mareso S, Angeli P. Infections complicating cirrhosis. Liver Int. 2018;38 Suppl 1:126-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 102]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
28.  Wencewicz TA. Crossroads of Antibiotic Resistance and Biosynthesis. J Mol Biol. 2019;431:3370-3399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 75]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
29.  Zaffiri L, Gardner J, Toledo-Pereyra LH. History of antibiotics. From salvarsan to cephalosporins. J Invest Surg. 2012;25:67-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 110]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
30.  Huemer M, Mairpady Shambat S, Brugger SD, Zinkernagel AS. Antibiotic resistance and persistence-Implications for human health and treatment perspectives. EMBO Rep. 2020;21:e51034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 207]  [Article Influence: 51.8]  [Reference Citation Analysis (0)]
31.  Xia J, Gao J, Tang W. Nosocomial infection and its molecular mechanisms of antibiotic resistance. Biosci Trends. 2016;10:14-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 54]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
32.  Blair JM, Webber MA, Baylay AJ, Ogbolu DO, Piddock LJ. Molecular mechanisms of antibiotic resistance. Nat Rev Microbiol. 2015;13:42-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2022]  [Cited by in F6Publishing: 2211]  [Article Influence: 221.1]  [Reference Citation Analysis (0)]
33.  Darby EM, Trampari E, Siasat P, Gaya MS, Alav I, Webber MA, Blair JMA. Molecular mechanisms of antibiotic resistance revisited. Nat Rev Microbiol. 2023;21:280-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 218]  [Article Influence: 218.0]  [Reference Citation Analysis (0)]
34.  Lerminiaux NA, Cameron ADS. Horizontal transfer of antibiotic resistance genes in clinical environments. Can J Microbiol. 2019;65:34-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 246]  [Cited by in F6Publishing: 324]  [Article Influence: 54.0]  [Reference Citation Analysis (0)]
35.  Boekhoud IM, Hornung BVH, Sevilla E, Harmanus C, Bos-Sanders IMJG, Terveer EM, Bolea R, Corver J, Kuijper EJ, Smits WK. Plasmid-mediated metronidazole resistance in Clostridioides difficile. Nat Commun. 2020;11:598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 67]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
36.  Munita JM, Arias CA. Mechanisms of Antibiotic Resistance. Microbiol Spectr. 2016;4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 927]  [Cited by in F6Publishing: 1154]  [Article Influence: 164.9]  [Reference Citation Analysis (0)]
37.  Maisonneuve E, Gerdes K. Molecular mechanisms underlying bacterial persisters. Cell. 2014;157:539-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 458]  [Cited by in F6Publishing: 389]  [Article Influence: 38.9]  [Reference Citation Analysis (0)]
38.  Ogura T, Hiraga S. Mini-F plasmid genes that couple host cell division to plasmid proliferation. Proc Natl Acad Sci U S A. 1983;80:4784-4788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 397]  [Cited by in F6Publishing: 404]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
39.  Yang QE, Walsh TR. Toxin-antitoxin systems and their role in disseminating and maintaining antimicrobial resistance. FEMS Microbiol Rev. 2017;41:343-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 79]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
40.  Dunn SJ, Connor C, McNally A. The evolution and transmission of multi-drug resistant Escherichia coli and Klebsiella pneumoniae: the complexity of clones and plasmids. Curr Opin Microbiol. 2019;51:51-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 71]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
41.  Fernández J, Navasa M, Planas R, Montoliu S, Monfort D, Soriano G, Vila C, Pardo A, Quintero E, Vargas V, Such J, Ginès P, Arroyo V. Primary prophylaxis of spontaneous bacterial peritonitis delays hepatorenal syndrome and improves survival in cirrhosis. Gastroenterology. 2007;133:818-824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 508]  [Cited by in F6Publishing: 430]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
42.  Biggins SW, Angeli P, Garcia-Tsao G, Ginès P, Ling SC, Nadim MK, Wong F, Kim WR. Diagnosis, Evaluation, and Management of Ascites, Spontaneous Bacterial Peritonitis and Hepatorenal Syndrome: 2021 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology. 2021;74:1014-1048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 316]  [Article Influence: 105.3]  [Reference Citation Analysis (0)]
43.  Premkumar M, Anand AC. Overview of Complications in Cirrhosis. J Clin Exp Hepatol. 2022;12:1150-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
44.  Saab S, Hernandez JC, Chi AC, Tong MJ. Oral antibiotic prophylaxis reduces spontaneous bacterial peritonitis occurrence and improves short-term survival in cirrhosis: a meta-analysis. Am J Gastroenterol. 2009;104:993-1001; quiz 1002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 70]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
45.  Chang JY, Kim SE, Kim TH, Woo SY, Ryu MS, Joo YH, Lee KE, Lee J, Lee KH, Moon CM, Jung HK, Shim KN, Jung SA. Emergence of rifampin-resistant staphylococci after rifaximin administration in cirrhotic patients. PLoS One. 2017;12:e0186120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
46.  Bajaj JS, Heuman DM, Sanyal AJ, Hylemon PB, Sterling RK, Stravitz RT, Fuchs M, Ridlon JM, Daita K, Monteith P, Noble NA, White MB, Fisher A, Sikaroodi M, Rangwala H, Gillevet PM. Modulation of the metabiome by rifaximin in patients with cirrhosis and minimal hepatic encephalopathy. PLoS One. 2013;8:e60042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 309]  [Article Influence: 28.1]  [Reference Citation Analysis (0)]
47.  Shamsaddini A, Gillevet PM, Acharya C, Fagan A, Gavis E, Sikaroodi M, McGeorge S, Khoruts A, Albhaisi S, Fuchs M, Sterling RK, Bajaj JS. Impact of Antibiotic Resistance Genes in Gut Microbiome of Patients With Cirrhosis. Gastroenterology. 2021;161:508-521.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
48.  Wang Y, Pan CQ, Xing H. Advances in Gut Microbiota of Viral Hepatitis Cirrhosis. Biomed Res Int. 2019;2019:9726786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
49.  Bajaj JS, Liu EJ, Kheradman R, Fagan A, Heuman DM, White M, Gavis EA, Hylemon P, Sikaroodi M, Gillevet PM. Fungal dysbiosis in cirrhosis. Gut. 2018;67:1146-1154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 89]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
50.  Fernández J, Prado V, Trebicka J, Amoros A, Gustot T, Wiest R, Deulofeu C, Garcia E, Acevedo J, Fuhrmann V, Durand F, Sánchez C, Papp M, Caraceni P, Vargas V, Bañares R, Piano S, Janicko M, Albillos A, Alessandria C, Soriano G, Welzel TM, Laleman W, Gerbes A, De Gottardi A, Merli M, Coenraad M, Saliba F, Pavesi M, Jalan R, Ginès P, Angeli P, Arroyo V; European Foundation for the Study of Chronic Liver Failure (EF-Clif). Multidrug-resistant bacterial infections in patients with decompensated cirrhosis and with acute-on-chronic liver failure in Europe. J Hepatol. 2019;70:398-411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 195]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
51.  Patel VC, Williams R. Antimicrobial resistance in chronic liver disease. Hepatol Int. 2020;14:24-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
52.  Yoshiji H, Nagoshi S, Akahane T, Asaoka Y, Ueno Y, Ogawa K, Kawaguchi T, Kurosaki M, Sakaida I, Shimizu M, Taniai M, Terai S, Nishikawa H, Hiasa Y, Hidaka H, Miwa H, Chayama K, Enomoto N, Shimosegawa T, Takehara T, Koike K. Evidence-based clinical practice guidelines for Liver Cirrhosis 2020. J Gastroenterol. 2021;56:593-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 127]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]
53.  Wu D, Wu SM, Lu J, Zhou YQ, Xu L, Guo CY. Rifaximin versus Nonabsorbable Disaccharides for the Treatment of Hepatic Encephalopathy: A Meta-Analysis. Gastroenterol Res Pract. 2013;2013:236963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 44]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
54.  Ulldemolins M, Roberts JA, Rello J, Paterson DL, Lipman J. The effects of hypoalbuminaemia on optimizing antibacterial dosing in critically ill patients. Clin Pharmacokinet. 2011;50:99-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 266]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
55.  Bajaj JS, Shamsaddini A, Fagan A, Sterling RK, Gavis E, Khoruts A, Fuchs M, Lee H, Sikaroodi M, Gillevet PM. Fecal Microbiota Transplant in Cirrhosis Reduces Gut Microbial Antibiotic Resistance Genes: Analysis of Two Trials. Hepatol Commun. 2021;5:258-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 35]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
56.  Wieërs G, Belkhir L, Enaud R, Leclercq S, Philippart de Foy JM, Dequenne I, de Timary P, Cani PD. How Probiotics Affect the Microbiota. Front Cell Infect Microbiol. 2019;9:454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 218]  [Article Influence: 54.5]  [Reference Citation Analysis (0)]
57.  Hughes RL, Holscher HD. Fueling Gut Microbes: A Review of the Interaction between Diet, Exercise, and the Gut Microbiota in Athletes. Adv Nutr. 2021;12:2190-2215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 54]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
58.  Wastyk HC, Fragiadakis GK, Perelman D, Dahan D, Merrill BD, Yu FB, Topf M, Gonzalez CG, Van Treuren W, Han S, Robinson JL, Elias JE, Sonnenburg ED, Gardner CD, Sonnenburg JL. Gut-microbiota-targeted diets modulate human immune status. Cell. 2021;184:4137-4153.e14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 429]  [Article Influence: 143.0]  [Reference Citation Analysis (0)]
59.  Tilg H, Cani PD, Mayer EA. Gut microbiome and liver diseases. Gut. 2016;65:2035-2044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 313]  [Article Influence: 39.1]  [Reference Citation Analysis (0)]
60.  Dronkers TMG, Ouwehand AC, Rijkers GT. Global analysis of clinical trials with probiotics. Heliyon. 2020;6:e04467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
61.  Maldonado Galdeano C, Cazorla SI, Lemme Dumit JM, Vélez E, Perdigón G. Beneficial Effects of Probiotic Consumption on the Immune System. Ann Nutr Metab. 2019;74:115-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 314]  [Article Influence: 62.8]  [Reference Citation Analysis (0)]
62.  Cai XJ, Wang L, Hu CM. Efficacy of different drugs in the treatment of minimal hepatic encephalopathy: A network meta-analysis involving 826 patients based on 10 randomized controlled trials. J Cell Biochem. 2018;119:8336-8345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
63.  Salomão MC, Heluany-Filho MA, Menegueti MG, Kraker ME, Martinez R, Bellissimo-Rodrigues F. A randomized clinical trial on the effectiveness of a symbiotic product to decolonize patients harboring multidrug-resistant Gram-negative bacilli. Rev Soc Bras Med Trop. 2016;49:559-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
64.  Kumar M, Dhaka P, Vijay D, Vergis J, Mohan V, Kumar A, Kurkure NV, Barbuddhe SB, Malik SV, Rawool DB. Antimicrobial effects of Lactobacillus plantarum and Lactobacillus acidophilus against multidrug-resistant enteroaggregative Escherichia coli. Int J Antimicrob Agents. 2016;48:265-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 51]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
65.  Halder D, Mandal S. Insights into the antagonism of Lactobacillus fermentum curd isolate against Gram-positive and Gram-negative pathogenic bacteria. Biosci Biotechnol Res Commun. 2018;11:461-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
66.  Khan AA, Manzoor KN, Sultan A, Saeed M, Rafique M, Noushad S, Talib A, Rentschler S, Deigner HP. Pulling the Brakes on Fast and Furious Multiple Drug-Resistant (MDR) Bacteria. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
67.  Ouwehand AC, Forssten S, Hibberd AA, Lyra A, Stahl B. Probiotic approach to prevent antibiotic resistance. Ann Med. 2016;48:246-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 89]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
68.  Yang F, Ren Z, Chai Q, Cui G, Jiang L, Chen H, Feng Z, Chen X, Ji J, Zhou L, Wang W, Zheng S. A novel biliary stent coated with silver nanoparticles prolongs the unobstructed period and survival via anti-bacterial activity. Sci Rep. 2016;6:21714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
69.  Lee NY, Ko WC, Hsueh PR. Nanoparticles in the Treatment of Infections Caused by Multidrug-Resistant Organisms. Front Pharmacol. 2019;10:1153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 230]  [Article Influence: 46.0]  [Reference Citation Analysis (0)]
70.  Khan I, Saeed K, Khan I. Nanoparticles: Properties, applications and toxicities. Arab J Chem. 2019;12:908-931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2058]  [Cited by in F6Publishing: 2157]  [Article Influence: 431.4]  [Reference Citation Analysis (0)]
71.  Karnwal A, Kumar G, Pant G, Hossain K, Ahmad A, Alshammari MB. Perspectives on Usage of Functional Nanomaterials in Antimicrobial Therapy for Antibiotic-Resistant Bacterial Infections. ACS Omega. 2023;8:13492-13508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 18]  [Reference Citation Analysis (0)]
72.  Duan Y, Young R, Schnabl B. Bacteriophages and their potential for treatment of gastrointestinal diseases. Nat Rev Gastroenterol Hepatol. 2022;19:135-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 42]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
73.  Taati Moghadam M, Amirmozafari N, Shariati A, Hallajzadeh M, Mirkalantari S, Khoshbayan A, Masjedian Jazi F. How Phages Overcome the Challenges of Drug Resistant Bacteria in Clinical Infections. Infect Drug Resist. 2020;13:45-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 76]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
74.  Imran M, Ahmad MN, Dasgupta A, Rana P, Srinivas N, Chopra S. Novel approaches for the treatment of infections due to multidrug-resistant bacterial pathogens. Future Med Chem. 2022;14:1133-1148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
75.  Elmaidomy AH, Shady NH, Abdeljawad KM, Elzamkan MB, Helmy HH, Tarshan EA, Adly AN, Hussien YH, Sayed NG, Zayed A, Abdelmohsen UR. Antimicrobial potentials of natural products against multidrug resistance pathogens: a comprehensive review. RSC Adv. 2022;12:29078-29102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
76.  Kennedy DA, Read AF. Why does drug resistance readily evolve but vaccine resistance does not? Proc Biol Sci. 2017;284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 77]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
77.  Thwaites GE, Gant V. Are bloodstream leukocytes Trojan Horses for the metastasis of Staphylococcus aureus? Nat Rev Microbiol. 2011;9:215-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 218]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
78.  Mirzaei B, Babaei R, Valinejad S. Staphylococcal Vaccine Antigens related to biofilm formation. Hum Vaccin Immunother. 2021;17:293-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
79.  Jansen KU, Anderson AS. The role of vaccines in fighting antimicrobial resistance (AMR). Hum Vaccin Immunother. 2018;14:2142-2149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 44]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
80.  Frost I, Sati H, Garcia-Vello P, Hasso-Agopsowicz M, Lienhardt C, Gigante V, Beyer P. The role of bacterial vaccines in the fight against antimicrobial resistance: an analysis of the preclinical and clinical development pipeline. Lancet Microbe. 2023;4:e113-e125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 48]  [Article Influence: 48.0]  [Reference Citation Analysis (0)]
81.  Feldman MF, Mayer Bridwell AE, Scott NE, Vinogradov E, McKee SR, Chavez SM, Twentyman J, Stallings CL, Rosen DA, Harding CM. A promising bioconjugate vaccine against hypervirulent Klebsiella pneumoniae. Proc Natl Acad Sci U S A. 2019;116:18655-18663.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 96]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
82.  Malachowa N, Kobayashi SD, Porter AR, Freedman B, Hanley PW, Lovaglio J, Saturday GA, Gardner DJ, Scott DP, Griffin A, Cordova K, Long D, Rosenke R, Sturdevant DE, Bruno D, Martens C, Kreiswirth BN, DeLeo FR. Vaccine Protection against Multidrug-Resistant Klebsiella pneumoniae in a Nonhuman Primate Model of Severe Lower Respiratory Tract Infection. mBio. 2019;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
83.  Zhang Y, Su J, Wu D.   Physiology and Pathology of Multidrug-Resistant Bacteria: Antibodies- and Vaccines-Based Pathogen-Specific Targeting. In: Physiology and Pathology of Immunology 2017.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Carrión-López P, Martínez-Ruiz J, Giménez-Bachs JM, Fernández-Anguita PJ, Díaz de Mera-Sánchez Migallón I, Legido-Gómez O, Rico-Marco S, Lorenzo-Sánchez MV, Salinas-Sánchez AS. Cost-Effectiveness of a Sublingual Bacterial Vaccine for the Prophylaxis of Recurrent Urinary Tract Infections. Urol Int. 2022;106:730-736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
85.  Nickel JC, Saz-Leal P, Doiron RC. Could sublingual vaccination be a viable option for the prevention of recurrent urinary tract infection in Canada? A systematic review of the current literature and plans for the future. Can Urol Assoc J. 2020;14:281-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
86.  Micoli F, Bagnoli F, Rappuoli R, Serruto D. The role of vaccines in combatting antimicrobial resistance. Nat Rev Microbiol. 2021;19:287-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 188]  [Article Influence: 62.7]  [Reference Citation Analysis (0)]