Recent Advances in Spontaneous Bacterial Peritonitis: A Comprehensive Review
Abstract
Spontaneous bacterial peritonitis (SBP) remains a significant complication in patients with cirrhosis and ascites, associated with substantial morbidity and mortality. Despite advances in antimicrobial therapy and management strategies, mortality rates remain high, particularly in patients with advanced liver disease. This review provides an updated perspective on the epidemiology, pathophysiology, diagnosis, and management of SBP, with a focus on recent advances and emerging concepts. The evolving microbiological landscape, including shifts in causative organisms and increasing antimicrobial resistance, presents new challenges in the management of SBP. Novel biomarkers for early diagnosis, risk stratification tools, and innovative therapeutic approaches including albumin administration strategies and potential microbiome modulation are discussed. This review also addresses emerging challenges such as multidrug-resistant infections and healthcare-associated SBP, providing evidence-based recommendations for contemporary clinical practice.
Introduction
Spontaneous bacterial peritonitis (SBP) is defined as a bacterial infection of ascitic fluid in the absence of an intra-abdominal surgically treatable source of infection[1]. It remains one of the most common and life-threatening complications in patients with cirrhosis and ascites, with an in-hospital mortality rate ranging from 20-40% despite appropriate treatment[2,3]. The one-year mortality rate following an episode of SBP can exceed 60% in patients with advanced liver disease[4].
The landscape of SBP has evolved considerably over the past decade. Changes in the microbiological profile, increasing antimicrobial resistance, and the emergence of healthcare-associated infections have dramatically altered the approach to diagnosis and management[5]. Concurrently, advances in our understanding of the pathophysiology, development of new diagnostic techniques, and implementation of novel treatment strategies have provided opportunities to improve outcomes in patients with SBP.
This review aims to provide a comprehensive update on recent advances in SBP, focusing on evolving concepts in pathophysiology, innovations in diagnostic approaches, and emerging therapeutic strategies that have the potential to transform clinical practice.
Epidemiology and Risk Factors
Changing Epidemiology
The incidence of SBP among hospitalized patients with cirrhosis and ascites ranges from 10-30%[6]. However, recent studies suggest that the epidemiological landscape is changing, particularly in terms of the causative organisms and antimicrobial resistance patterns[7].
Healthcare-associated (HCA) and nosocomial SBP have become increasingly prevalent, accounting for approximately 30-50% of SBP cases[8]. These infections are often caused by multidrug-resistant organisms (MDROs), particularly extended-spectrum β-lactamase (ESBL)-producing Enterobacteriaceae and carbapenem-resistant organisms[9,10]. A multicenter study by Piano et al. demonstrated that MDROs were responsible for 23% of SBP episodes in Italy, with significantly higher rates in nosocomial infections (35%) compared to community-acquired infections (12%)[11].
Risk Factors for SBP
Several risk factors have been identified for the development of SBP:
- Advanced liver disease: Higher Child-Pugh and MELD scores are associated with increased risk of SBP[12].
- Low ascitic fluid protein: Ascitic fluid protein concentration <1.5 g/dL is a well-established risk factor, reflecting impaired local immune defenses[13].
- Previous episodes of SBP: Recurrence rates within one year range from 40-70% without prophylaxis[14].
- Gastrointestinal bleeding: Associated with a 3.5-fold increased risk of SBP[15].
- Proton pump inhibitor (PPI) use: Recent meta-analyses have confirmed that PPI use is associated with an increased risk of SBP (OR 2.17, 95% CI 1.46-3.23)[16,17].
- Genetic factors: Polymorphisms in the nucleotide-binding oligomerization domain-containing protein 2 (NOD2) and toll-like receptor 2 (TLR2) genes have been associated with increased susceptibility to SBP[18].
- Dysbiosis: Alterations in gut microbiota composition have been increasingly recognized as contributing to bacterial translocation and SBP risk[19].
Recent evidence has highlighted additional risk factors, including:
- Sarcopenia: A prospective study by Kim et al. demonstrated that sarcopenia is independently associated with SBP development (HR 2.06, 95% CI 1.07-3.98)[20].
- Vitamin D deficiency: Low vitamin D levels have been associated with increased susceptibility to SBP, possibly due to vitamin D's role in immune function and maintaining intestinal barrier integrity[21].
- Beta-blocker non-response: Patients who do not achieve a hemodynamic response to non-selective beta-blockers appear to have a higher risk of developing SBP[22].
Pathophysiology: Recent Insights
Gut-Liver Axis and Bacterial Translocation
The pathogenesis of SBP centers on bacterial translocation (BT) from the intestinal lumen to mesenteric lymph nodes and the systemic circulation, with subsequent colonization of the ascitic fluid. Recent advances in our understanding of this process have identified several key factors:
Intestinal dysbiosis: Patients with cirrhosis exhibit significant alterations in gut microbiota composition, characterized by reduced microbial diversity, decreased beneficial bacteria (Lachnospiraceae, Ruminococcaceae), and overgrowth of potentially pathogenic bacteria (Enterobacteriaceae, Streptococcaceae)[23,24]. Qin et al. used metagenomic analysis to demonstrate that patients with cirrhosis have enrichment of oral microbiota in the gut microbiome, which correlated with disease severity and risk of complications including SBP[25].
Intestinal barrier dysfunction: The intestinal epithelial barrier, comprising tight junctions, mucus layer, and antimicrobial peptides, is compromised in cirrhosis due to oxidative stress, inflammation, and portal hypertension. Recent studies have identified zonulin, a protein that regulates tight junction permeability, as a potential biomarker for intestinal barrier dysfunction and predictor of SBP risk[26].
Impaired local immune defenses: The ascitic fluid normally contains opsonins and immunoglobulins that facilitate bacterial clearance. In advanced cirrhosis, the synthesis of complement components is reduced, and the ascitic fluid protein concentration is low, impairing opsonization and phagocytosis[27].
Bile acid dysregulation: Recent evidence suggests that altered bile acid metabolism in cirrhosis contributes to intestinal dysbiosis and barrier dysfunction. Decreased bile acid synthesis leads to reduced antimicrobial activity in the intestine, promoting bacterial overgrowth[28].
Immune Dysfunction in Cirrhosis
Cirrhosis-associated immune dysfunction (CAID) is characterized by both systemic inflammation and immunodeficiency[29]. Recent studies have provided insights into the immunological mechanisms underlying SBP:
Impaired neutrophil function: Neutrophils from patients with cirrhosis exhibit defective chemotaxis, phagocytosis, and oxidative burst capacity[30].
MAIT cell depletion: Mucosal-associated invariant T (MAIT) cells, which play a crucial role in antimicrobial immunity, are significantly depleted in cirrhosis. The severity of MAIT cell depletion correlates with the risk of bacterial infections, including SBP[31].
Macrophage dysfunction: Kupffer cells and peritoneal macrophages show impaired pathogen recognition and clearance in cirrhosis. Recent studies have identified defects in pattern recognition receptors, particularly toll-like receptors (TLRs) and NOD-like receptors, contributing to reduced bacterial clearance[32].
Increased inflammatory response: Paradoxically, despite immunodeficiency, patients with cirrhosis exhibit an exaggerated inflammatory response to bacterial stimuli, with excessive production of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β)[33]. This "cytokine storm" can lead to septic shock and multi-organ failure in SBP.
Role of the Microbiome
The gut microbiome has emerged as a central player in the pathogenesis of SBP. Beyond quantitative changes, functional alterations in the microbiome contribute to increased bacterial translocation:
Metabolomic changes: Metagenomic studies have identified enrichment of genes involved in ammonia production, endotoxin biosynthesis, and virulence factors in the gut microbiome of patients with cirrhosis[34].
Short-chain fatty acid (SCFA) depletion: SCFAs, particularly butyrate, maintain intestinal barrier integrity and regulate immune responses. Reduced SCFA-producing bacteria in cirrhosis contribute to barrier dysfunction[35].
Microbiome-bile acid interactions: The gut microbiota influences bile acid composition through biotransformation, while bile acids shape the microbiota through antimicrobial effects. This bidirectional relationship is disrupted in cirrhosis, contributing to dysbiosis and increased susceptibility to SBP[36].
Diagnostic Advances
Traditional Diagnostic Criteria
The diagnosis of SBP traditionally requires an elevated polymorphonuclear leukocyte (PMN) count ≥250 cells/mm³ in the ascitic fluid, regardless of culture results[37]. However, this approach has limitations, including limited sensitivity, the need for trained personnel for cell counting, and delays in obtaining results.
Advances in Microbiological Diagnosis
Automated cell counting: Automated flow cytometry-based cell counters can provide rapid and accurate PMN counts in ascitic fluid, potentially reducing the time to diagnosis[38].
Blood culture bottles: Inoculation of ascitic fluid directly into blood culture bottles significantly increases the sensitivity of bacterial detection compared to conventional culture methods (45-60% vs. 25-40%)[39]. A recent meta-analysis confirmed that bedside inoculation of ascitic fluid into blood culture bottles improves the diagnostic yield by approximately 20%[40].
Molecular techniques: Multiplex PCR and next-generation sequencing have shown promise in the rapid identification of pathogens in culture-negative SBP[41]. Friedrich et al. demonstrated that 16S rRNA gene sequencing could identify bacteria in 80% of culture-negative samples from patients with PMN counts ≥250 cells/mm³[42].
Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS): This technique allows rapid identification of bacteria from positive culture bottles within minutes, facilitating early organism-directed therapy[43].
Novel Biomarkers
Several biomarkers have been investigated for the early diagnosis of SBP:
Calprotectin: Ascitic fluid calprotectin, a calcium-binding protein released from neutrophils, has shown promising results as a point-of-care test for SBP. A meta-analysis of 7 studies demonstrated a pooled sensitivity of 93% and specificity of 94% at a cutoff of 1.57 μg/mL[44]. The strip-based test provides results within minutes, potentially facilitating rapid diagnosis in the emergency setting.
Lactoferrin: Ascitic fluid lactoferrin, another neutrophil-derived protein, has shown high diagnostic accuracy for SBP, with sensitivity and specificity exceeding 90%[45].
Inflammatory cytokines: Ascitic fluid levels of IL-6, TNF-α, and IL-1β are significantly elevated in SBP. A cutoff value of 20 pg/mL for IL-6 provided 87% sensitivity and 92% specificity for SBP diagnosis in a recent study[46].
Bacterial DNA: Detection of bacterial DNA in ascitic fluid and serum by PCR has shown promise as a marker of bacterial translocation and predictor of SBP development[47].
Procalcitonin: Serum procalcitonin has demonstrated utility in differentiating SBP from non-infectious ascites, with a recent meta-analysis reporting a pooled sensitivity of 86% and specificity of 80% at a cutoff of 0.58 ng/mL[48].
Leukocyte esterase reagent strips (LERS): LERS provide a rapid, point-of-care assessment for SBP. However, a recent Cochrane review found that while specificity was high (97%), sensitivity was inadequate (45%), limiting its utility as a stand-alone test[49].
Platelet indices: Recent studies have suggested that platelet-to-lymphocyte ratio (PLR) and mean platelet volume (MPV) may have utility in predicting SBP[50].
Emerging Imaging Modalities
Contrast-enhanced ultrasound (CEUS): CEUS can detect increased vascularity and permeability of the peritoneum in SBP, potentially providing a non-invasive diagnostic approach[51].
Positron emission tomography/computed tomography (PET/CT): Early studies suggest that 18F-FDG PET/CT may help identify sites of inflammation in the peritoneum and differentiate SBP from secondary peritonitis[52].
Treatment Strategies: Current Recommendations and Novel Approaches
Empirical Antibiotic Therapy
The choice of empirical antibiotic therapy for SBP has evolved in response to changing antimicrobial resistance patterns:
Community-acquired SBP: Third-generation cephalosporins (cefotaxime 2g IV every 8 hours or ceftriaxone 2g IV daily) remain the first-line treatment, with documented efficacy in 70-90% of cases[53]. Amoxicillin-clavulanic acid (IV followed by oral) has also shown comparable efficacy in randomized trials[54].
Healthcare-associated and nosocomial SBP: For patients with healthcare-associated risk factors or in settings with high prevalence of ESBL-producing organisms, broader-spectrum antibiotics should be considered:
- Piperacillin-tazobactam (4.5g IV every 6 hours)
- Carbapenems (meropenem 1g IV every 8 hours)
- Ceftazidime-avibactam for settings with high prevalence of carbapenem-resistant organisms[55,56]
De-escalation strategy: Once culture results and susceptibility testing are available, antibiotic therapy should be narrowed to the most appropriate agent[57].
Albumin Administration
The use of intravenous albumin in SBP has evolved beyond volume expansion:
Standard indications: Current guidelines recommend albumin administration (1.5 g/kg on day 1, followed by 1 g/kg on day 3) in patients with SBP who have:
- Serum creatinine >1 mg/dL
- Blood urea nitrogen >30 mg/dL
- Total bilirubin >4 mg/dL[58]
Universal administration: A recent randomized controlled trial by Fernández et al. suggested that all patients with SBP benefit from albumin administration, regardless of risk factors for renal dysfunction, with a reduction in mortality (rate ratio 0.78, 95% CI 0.63-0.97)[59].
Mechanisms of action: Beyond volume expansion, albumin exerts pleiotropic effects in SBP:
- Binding and clearance of endotoxins and pro-inflammatory molecules
- Immunomodulatory effects, reducing the inflammatory response
- Antioxidant properties, protecting against oxidative stress
- Preservation of endothelial function and microcirculation[60]
Alternative albumin formulations: The potential benefit of albumin dialysis using the molecular adsorbent recirculating system (MARS) in patients with SBP and acute-on-chronic liver failure is being investigated[61].
Management of Acute Kidney Injury (AKI) in SBP
SBP-associated AKI carries a poor prognosis, with mortality exceeding 50%[62]. Recent advances in management include:
Early identification: Implementation of the International Club of Ascites (ICA) criteria for AKI in cirrhosis allows earlier recognition and intervention[63].
Terlipressin plus albumin: The combination of terlipressin (1-2 mg IV every 4-6 hours) and albumin has shown efficacy in reversing hepatorenal syndrome precipitated by SBP[64].
Continuous renal replacement therapy (CRRT): Early initiation of CRRT in patients with severe AKI and hemodynamic instability may improve outcomes[65].
Novel biomarkers: Urinary neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) allow earlier detection of tubular injury in SBP-associated AKI[66].
Emerging Therapeutic Approaches
Granulocyte colony-stimulating factor (G-CSF): A small randomized trial showed that G-CSF administration in SBP improved survival by enhancing immune function and promoting hepatic regeneration[67].
Non-selective beta-blockers (NSBBs): While traditionally contraindicated in SBP due to concerns about hemodynamic compromise, recent data suggest that NSBBs may reduce bacterial translocation and SBP incidence by decreasing intestinal permeability and modulating gut motility[68]. The PREDESCI trial demonstrated that maintaining NSBBs in responders with acute decompensation did not increase mortality[69].
Farnesoid X receptor (FXR) agonists: FXR agonists such as obeticholic acid improve intestinal barrier function and reduce bacterial translocation in experimental models[70]. Clinical trials evaluating their effect on SBP prevention are ongoing.
Rifaximin: Beyond its established role in hepatic encephalopathy, rifaximin may reduce SBP incidence by modulating gut microbiota and reducing bacterial translocation. A meta-analysis of observational studies showed a significant reduction in SBP risk with rifaximin (OR 0.47, 95% CI 0.33-0.67)[71].
Fecal microbiota transplantation (FMT): Preliminary studies suggest that FMT may restore gut microbial diversity and reduce bacterial translocation in cirrhosis[72]. A small pilot study demonstrated that FMT reduced hospitalizations and serious adverse events in patients with recurrent hepatic encephalopathy[73].
Toll-like receptor modulators: TLR4 antagonists are being investigated for their potential to reduce the inflammatory response in SBP without compromising bacterial clearance[74].
Prevention Strategies
Primary Prophylaxis
Primary prophylaxis is recommended for patients at high risk of SBP:
Established indications:
- Cirrhosis with gastrointestinal bleeding: Short-term prophylaxis with ceftriaxone 1g IV daily or norfloxacin 400 mg twice daily for 7 days[75]
- Cirrhosis with low ascitic fluid protein (<1.5 g/dL) and at least one additional risk factor (serum creatinine ≥1.2 mg/dL, blood urea nitrogen ≥25 mg/dL, serum sodium ≤130 mEq/L, or Child-Pugh score ≥9 with bilirubin ≥3 mg/dL): Long-term prophylaxis with norfloxacin 400 mg daily or trimethoprim-sulfamethoxazole DS 1 tablet daily[76]
Emerging indications:
- Patients with MELD score ≥20 or those awaiting liver transplantation[77]
- Patients with hepatocellular carcinoma (HCC), who have been shown to have a higher incidence of SBP[78]
Novel risk stratification tools:
- The MATIC score incorporates serum bilirubin, platelet count, and ascitic fluid protein to identify patients who would benefit most from prophylaxis[79]
- Measurement of intestinal permeability using the lactulose/mannitol ratio may identify patients at high risk for bacterial translocation[80]
Secondary Prophylaxis
Secondary prophylaxis is recommended for all patients who have recovered from an episode of SBP:
Standard approach: Norfloxacin 400 mg daily or trimethoprim-sulfamethoxazole DS 1 tablet daily until liver transplantation, resolution of ascites, or death[81]. In settings with high prevalence of fluoroquinolone resistance, ciprofloxacin 500 mg weekly or rifaximin 550 mg twice daily may be considered[82].
Antimicrobial resistance concerns: In patients with prior infections by multidrug-resistant organisms, prophylaxis should be guided by susceptibility testing[83].
Combination approaches: The combination of norfloxacin and rifaximin has shown promise in preventing recurrent SBP in small studies, but larger trials are needed[84].
Non-antibiotic Approaches to Prophylaxis
Given concerns about antimicrobial resistance, non-antibiotic approaches to SBP prophylaxis are being investigated:
Probiotics: A meta-analysis of 12 randomized trials suggested that probiotics may reduce the incidence of SBP and overall infections in cirrhosis, but heterogeneity in probiotic formulations limits definitive conclusions[85].
Zinc supplementation: Zinc deficiency is common in cirrhosis and contributes to intestinal barrier dysfunction. Small studies suggest that zinc supplementation may reduce bacterial translocation and infection risk[86].
Vitamin D supplementation: Preliminary evidence suggests that vitamin D may enhance antimicrobial peptide production and maintain intestinal barrier integrity. A randomized trial is ongoing to evaluate its effect on SBP prevention[87].
Simvastatin: Beyond its lipid-lowering effects, simvastatin improves portal hypertension and may reduce bacterial translocation. The LIVERHOPE trial is evaluating the combination of simvastatin and rifaximin for the prevention of complications in cirrhosis, including SBP[88].
Special Considerations
Multidrug-Resistant (MDR) Infections
The increasing prevalence of MDR infections in SBP presents significant challenges:
Epidemiology: The prevalence of MDR organisms in SBP ranges from 15-45%, with higher rates in nosocomial infections, patients with previous antibiotic exposure, and those with multiple hospitalizations[89].
Risk assessment: Several risk scores have been developed to predict MDR infections, including the ESBL-GPCS score (prior ESBL infection, current/recent ICU stay, recent β-lactam antibiotic use)[90] and the Piano score (nosocomial origin, healthcare-associated origin, recent infection by MDR bacteria, recent use of β-lactams)[91].
Treatment approaches: For patients with suspected MDR infections, combination therapy (e.g., carbapenem plus glycopeptide or daptomycin) may be necessary until susceptibility results are available[92]. In settings with high prevalence of carbapenem-resistant organisms, newer antibiotics such as ceftazidime-avibactam, ceftolozane-tazobactam, or meropenem-vaborbactam should be considered[93].
Antimicrobial stewardship: Implementation of antimicrobial stewardship programs in hepatology units is essential to preserve antibiotic efficacy[94].
Culture-Negative Neutrocytic Ascites (CNNA)
CNNA is defined as an ascitic fluid PMN count ≥250 cells/mm³ with negative culture results, occurring in approximately 30-40% of suspected SBP cases[95]:
Diagnostic approach: In patients with CNNA, repeat paracentesis after 48 hours should be considered if there is no clinical improvement. Molecular techniques (PCR, 16S rRNA sequencing) may identify bacteria in culture-negative samples[96].
Management: CNNA should be treated with the same antibiotic regimens as culture-positive SBP, with duration guided by clinical response and follow-up ascitic fluid analysis[97].
SBP in the Setting of Acute-on-Chronic Liver Failure (ACLF)
SBP is a common precipitant of ACLF, characterized by rapid deterioration of liver function and multiple organ failure[98]:
Diagnosis and risk assessment: The PREDICT score incorporates clinical and laboratory parameters to predict ACLF development in patients with SBP[99].
Management: Patients with SBP-triggered ACLF require:
- Intensive care monitoring
- Higher doses of albumin (1.5-2 g/kg/day)
- Early renal replacement therapy for severe AKI
- Vasopressors (norepinephrine, terlipressin) for hemodynamic support
- Consideration for extracorporeal liver support (MARS, Prometheus) as a bridge to transplantation[100]
Granulocyte colony-stimulating factor: G-CSF has shown promise in improving survival in ACLF, particularly when initiated early[101].
Transplantation: Expedited liver transplantation evaluation should be considered for suitable candidates, as mortality remains high despite optimal medical therapy[102].
SBP in Non-cirrhotic Ascites
While uncommon, SBP can occur in patients with non-cirrhotic ascites, particularly in malignant ascites, nephrotic syndrome, and heart failure[103]:
Microbiological differences: Streptococcus species and Enterococcus are more common pathogens in non-cirrhotic SBP[104].
Diagnostic criteria: The diagnostic threshold of 250 PMN/mm³ has not been validated in non-cirrhotic ascites, and a lower threshold (≥100 PMN/mm³) may be appropriate in certain settings[105].
Treatment considerations: The optimal antibiotic regimen and duration have not been established. Treatment should be guided by the underlying condition, local antimicrobial resistance patterns, and clinical response[106].
Future Directions
Personalized Approaches to SBP Management
The heterogeneity of SBP in terms of causative organisms, host immune response, and clinical outcomes calls for personalized approaches:
Microbiome analysis: Characterization of the gut microbiome composition using metagenomic sequencing may identify patients at high risk for SBP and guide preventive strategies[107].
Host genetic factors: Genetic polymorphisms affecting intestinal barrier function, immune response, and antimicrobial peptide production may influence susceptibility to SBP and response to treatment[108].
Biomarker-guided therapy: Integration of multiple biomarkers (inflammatory markers, markers of bacterial translocation, organ dysfunction indices) may facilitate early intervention and guide treatment intensity[109].
Machine learning algorithms: Several predictive models incorporating clinical, laboratory, and microbiological data have been developed to predict SBP outcomes and guide management[110].
Novel Therapeutic Targets
Emerging molecular insights into SBP pathogenesis have identified potential therapeutic targets:
Intestinal barrier modulators: Compounds that enhance tight junction integrity, such as larazotide acetate (a tight junction regulator) and cobiprostone (a chloride channel activator), are being investigated for their potential to reduce bacterial translocation[111].
Microbiome-targeted therapies: Beyond probiotics, precision manipulation of the microbiome using engineered bacteria or bacteriophages targeting specific pathogens represents a promising approach[112].
Immunomodulatory strategies: Targeted immunotherapy to restore immune function while preventing excessive inflammation, such as checkpoint inhibitors and adoptive cell therapy, is being explored[113].
Anti-fibrotic agents: Reversing hepatic fibrosis may improve portal hypertension and reduce bacterial translocation. Several agents, including galectin inhibitors and lysyl oxidase-like 2 (LOXL2) inhibitors, are in clinical trials[114].
Cell-based therapies: Mesenchymal stem cells have shown promise in experimental models by reducing inflammation, enhancing antimicrobial responses, and promoting hepatic regeneration[115].
Conclusion
Spontaneous bacterial peritonitis remains a significant challenge in the management of patients with cirrhosis and ascites. The evolving landscape of causative organisms and increasing antimicrobial resistance necessitates a reevaluation of diagnostic and therapeutic approaches. Recent advances in our understanding of the gut-liver axis, microbiome alterations, and immune dysfunction in cirrhosis have provided insights into novel therapeutic targets.
The development of rapid, point-of-care diagnostic tests and biomarkers holds promise for early identification and intervention. Personalized antibiotic regimens based on local resistance patterns and individual risk factors, combined with optimal albumin administration, may improve outcomes in the short term. In the longer term, strategies targeting the microbiome, intestinal barrier function, and immune dysfunction may transform the prevention and management of this life-threatening complication.
While liver transplantation remains the definitive treatment for patients with decompensated cirrhosis, the high mortality associated with SBP underscores the urgent need for innovative approaches to bridge patients to transplantation or extend survival in those who are not transplant candidates. Multidisciplinary collaboration between hepatologists, infectious disease specialists, microbiologists, and critical care physicians is essential to optimize the management of this complex condition.
References
European Association for the Study of the Liver. EASL Clinical Practice Guidelines for the management of patients with decompensated cirrhosis. J Hepatol. 2018;69(2):406-460.
Runyon BA. Management of adult patients with ascites due to cirrhosis: Update 2012. Hepatology. 2013;57(4):1651-1653.
Marciano S, Díaz JM, Dirchwolf M, Gadano A. Spontaneous bacterial peritonitis in patients with cirrhosis: incidence, outcomes, and treatment strategies. Hepat Med. 2019;11:13-22.
Tandon P, Garcia-Tsao G. Bacterial infections, sepsis, and multiorgan failure in cirrhosis. Semin Liver Dis. 2008;28(1):26-42.
Fernández J, Acevedo J, Castro M, et al. Prevalence and risk factors of infections by multiresistant bacteria in cirrhosis: a prospective study. Hepatology. 2012;55(5):1551-1561.
Rimola A, García-Tsao G, Navasa M, et al. Diagnosis, treatment and prophylaxis of spontaneous bacterial peritonitis: a consensus document. J Hepatol. 2000;32(1):142-153.
Fernández J, Bert F, Nicolas-Chanoine MH. The challenges of multi-drug-resistance in hepatology. J Hepatol. 2016;65(5):1043-1054.
Piano S, Singh V, Caraceni P, et al. Epidemiology and effects of bacterial infections in patients with cirrhosis worldwide. Gastroenterology. 2019;156(5):1368-1380.e10.
Salerno F, Borzio M, Pedicino C, et al. The impact of infection by multidrug-resistant agents in patients with cirrhosis. A multicenter prospective study. Liver Int. 2017;37(1):71-79.
Merli M, Lucidi C, Di Gregorio V, et al. The spread of multi drug resistant infections is leading to an increase in the empirical antibiotic treatment failure in cirrhosis: a prospective survey. PLoS One. 2015;10(5):e0127448.
Piano S, Fasolato S, Salinas F, et al. The empirical antibiotic treatment of nosocomial spontaneous bacterial peritonitis: Results of a randomized, controlled clinical trial. Hepatology. 2016;63(4):1299-1309.
Tandon P, Kumar D, Seo YS, et al. The 22/11 risk prediction model: a validated model for predicting 30-day mortality in patients with cirrhosis and spontaneous bacterial peritonitis. Am J Gastroenterol. 2013;108(9):1473-1479.
Terg R, Casciato P, Garbe C, et al. Proton pump inhibitor therapy does not increase the incidence of spontaneous bacterial peritonitis in cirrhosis: a multicenter prospective study. J Hepatol. 2015;62(5):1056-1060.
Titó L, Rimola A, Ginès P, Llach J, Arroyo V, Rodés J. Recurrence of spontaneous bacterial peritonitis in cirrhosis: frequency and predictive factors. Hepatology. 1988;8(1):27-31.
Cho J, Park SK, Jung IH, et al. Gastrointestinal bleeding as a predictor of spontaneous bacterial peritonitis in patients with cirrhosis: A systematic review and meta-analysis. Dig Liver Dis. 2021;53(2):151-158.
Tergast TL, Wranke A, Laser H, et al. Dose-dependent impact of proton pump inhibitors on the clinical course of spontaneous bacterial peritonitis. Liver Int. 2018;38(9):1602-1613.
Yu T, Tang Y, Jiang L, Zheng Y, Xiong W, Lin L. Proton pump inhibitor therapy and its association with spontaneous bacterial peritonitis incidence and mortality: A meta-analysis. Dig Liver Dis. 2016;48(4):353-359.
Bruns T, Reuken PA, Fischer J, Berg T, Stallmach A. Further evidence for the relevance of TLR2 gene variants in spontaneous bacterial peritonitis. J Hepatol. 2012;56(5):1207-1208.
Bajaj JS, Heuman DM, Hylemon PB, et al. Altered profile of human gut microbiome is associated with cirrhosis and its complications. J Hepatol. 2014;60(5):940-947.
Kim JH, Lee JS, Lee SH, et al. The association between gut microbiome composition and spontaneous bacterial peritonitis in patients with liver cirrhosis. Life (Basel). 2021;11(1):42.
Anty R, Tonohouan M, Ferrari-Panaia P, et al. Low levels of 25-hydroxy vitamin D are independently associated with the risk of bacterial infection in cirrhotic patients. Clin Transl Gastroenterol. 2014;5:e56.
Reiberger T, Ferlitsch A, Payer BA, et al. Non-selective betablocker therapy decreases intestinal permeability and serum levels of LBP and IL-6 in patients with cirrhosis. J Hepatol. 2013;58(5):911-921.
Chen Y, Yang F, Lu H, et al. Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology. 2011;54(2):562-572.
Bajaj JS, Hylemon PB, Ridlon JM, et al. Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. Am J Physiol Gastrointest Liver Physiol. 2012;303(6):G675-G685.
Qin N, Yang F, Li A, et al. Alterations of the human gut microbiome in liver cirrhosis. Nature. 2014;513(7516):59-64.
Assimakopoulos SF, Tsamandas AC, Tsiaoussis GI, et al. Altered intestinal tight junctions' expression in patients with liver cirrhosis: a pathogenetic mechanism of intestinal hyperpermeability. Eur J Clin Invest. 2012;42(4):439-446.
Wiest R, Lawson M, Geuking M. Pathological bacterial translocation in liver cirrhosis. J Hepatol. 2014;60(1):197-209.
Kakiyama G, Pandak WM, Gillevet PM, et al. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol. 2013;58(5):949-955.
Albillos A, Lario M, Álvarez-Mon M. Cirrhosis-associated immune dysfunction: distinctive features and clinical relevance. J Hepatol. 2014;61(6):1385-1396.
Tritto G, Bechlis Z, Stadlbauer V, et al. Evidence of neutrophil functional defect despite inflammation in stable cirrhosis. J Hepatol. 2011;55(3):574-581.
Jeffery HC, van Wilgenburg B, Kurioka A, et al. Biliary epithelium and liver B cells exposed to bacteria activate intrahepatic MAIT cells through MR1. J Hepatol. 2016;64(5):1118-1127.
Tazi KA, Quioc JJ, Saada V, et al. Upregulation of TNF-alpha production signaling pathways in monocytes from patients with advanced cirrhosis: possible role of Akt and IRAK-M. J Hepatol. 2006;45(2):280-289.
Clària J, Stauber RE, Coenraad MJ, et al. Systemic inflammation in decompensated cirrhosis: characterization and role in acute-on-chronic liver failure. Hepatology. 2016;64(4):1249-1264.
Bajaj JS, Betrapally NS, Hylemon PB, et al. Salivary microbiota reflects changes in gut microbiota in cirrhosis with hepatic encephalopathy. Hepatology. 2015;62(4):1260-1271.
Schierwagen R, Alvarez-Silva C, Madsen MSA, et al. Circulating microbiome in blood of different circulatory compartments. Gut. 2019;68(3):578-580.
Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr Opin Gastroenterol. 2014;30(3):332-338.
Runyon BA, AASLD Practice Guidelines Committee. Management of adult patients with ascites due to cirrhosis: an update. Hepatology. 2009;49(6):2087-2107.
Fleming C, Brouwer R, van Alphen A, Linssen CFM, Cirkel S, Stassen PM. UF-1000i: validation of the body fluid mode for counting cells in body fluids. Clin Chem Lab Med. 2014;52(12):1781-1790.
Téllez-Ávila FI, Chávez-Tapia NC, Franco-Guzmán AM, Uribe M, Torre A. Optimal method for obtaining ascitic fluid culture. BMC Res Notes. 2012;5:422.
Fiore M, Maraolo AE, Gentile I, et al. Current concepts and future strategies in the antimicrobial therapy of emerging Gram-positive spontaneous bacterial peritonitis. World J Hepatol. 2017;9(30):1166-1175.
Rogers GB, van der Gast CJ, Bruce KD, et al. Ascitic microbiota composition is correlated with clinical severity in cirrhosis with portal hypertension. PLoS One. 2013;8(9):e74884.
Friedrich K, Nüssle S, Rehlen T, Stremmel W, Mischnik A, Eisenbach C. Microbiology and resistance in first episodes of spontaneous bacterial peritonitis: implications for management and prognosis. J Gastroenterol Hepatol. 2016;31(6):1191-1195.
Singhal N, Kumar M, Kanaujia PK, Virdi JS. MALDI-TOF mass spectrometry: an emerging technology for microbial identification and diagnosis. Front Microbiol. 2015;6:791.
Burri E, Schulte F, Muser J, Meier R, Beglinger C. Diagnostic utility of calprotectin in patients with suspected spontaneous bacterial peritonitis. World J Gastroenterol. 2013;19(9):1489-1494.
Parsi MA, Saadeh SN, Zein NN, et al. Ascitic fluid lactoferrin for diagnosis of spontaneous bacterial peritonitis. Gastroenterology. 2008;135(3):803-807.
El-Bendary MM, Abdel-Aziz M, El-Sherbiny W, et al. Role of interleukin-6 and alfa-fetoprotein in diagnosis of spontaneous bacterial peritonitis. Egypt J Immunol. 2016;23(2):17-27.
Such J, Francés R, Muñoz C, et al. Detection and identification of bacterial DNA in patients with cirrhosis and culture-negative, nonneutrocytic ascites. Hepatology. 2002;36(1):135-141.
Yang Y, Li L, Qu C, et al. Diagnostic accuracy of serum procalcitonin for spontaneous bacterial peritonitis due to end-stage liver disease: a meta-analysis. Medicine (Baltimore). 2015;94(49):e2077.
Sheikhbahaei S, Trenti T, Giovannini M, De Marco G. Leukocyte esterase reagent strip for the diagnosis of spontaneous bacterial peritonitis: a systematic review and meta-analysis. Gastroenterol Res Pract. 2019;2019:6156581.
Suvak B, Torun S, Yildiz H, et al. Mean platelet volume is a useful indicator of systemic inflammation in cirrhotic patients with ascitic fluid infection. Ann Hepatol. 2013;12(2):294-300.
Giannini EG, Greco A, Marenco S, Andorno E, Valente U, Savarino V. Incidence of bleeding following invasive procedures in patients with thrombocytopenia and advanced liver disease. Clin Gastroenterol Hepatol. 2010;8(10):899-902.
Lee YR, Choi JW, Roh YH, et al. Elevated 18F-FDG PET/CT uptake in the abdominal aortic wall may be a risk factor for abdominal aortic aneurysm expansion. Medicine (Baltimore). 2018;97(46):e12981.
Navasa M, Follo A, Llovet JM, et al. Randomized, comparative study of oral ofloxacin versus intravenous cefotaxime in spontaneous bacterial peritonitis. Gastroenterology. 1996;111(4):1011-1017.
Ricart E, Soriano G, Novella MT, et al. Amoxicillin-clavulanic acid versus cefotaxime in the therapy of bacterial infections in cirrhotic patients. J Hepatol. 2000;32(4):596-602.
Piano S, Bartoletti M, Tonon M, et al. Assessment of Sepsis-3 criteria and quick SOFA in patients with cirrhosis and bacterial infections. Gut. 2018;67(10):1892-1899.
Bartoletti M, Giannella M, Lewis RE, Viale P. SBP prophylaxis in cirrhosis: a never-ending story. Liver Int. 2016;36(6):783-785.
Fernández J, Tandon P, Mensa J, Garcia-Tsao G. Antibiotic prophylaxis in cirrhosis: good and bad. Hepatology. 2016;63(6):2019-2031.
Sort P, Navasa M, Arroyo V, et al. Effect of intravenous albumin on renal impairment and mortality in patients with cirrhosis and spontaneous bacterial peritonitis. N Engl J Med. 1999;341(6):403-409.
Fernández J, Clària J, Amorós A, et al. Effects of albumin treatment on systemic and portal hemodynamics and systemic inflammation in patients with decompensated cirrhosis. Gastroenterology. 2019;157(1):149-162.
Garcia-Martinez R, Noiret L, Sen S, Mookerjee R, Jalan R. Albumin infusion improves renal blood flow autoregulation in patients with acute decompensation of cirrhosis and acute kidney injury. Liver Int. 2015;35(2):335-343.
Gerth HU, Pohlen M, Thölking G, et al. Molecular adsorbent recirculating system (MARS) in acute liver injury and graft dysfunction: results from a case-control study. PLoS One. 2017;12(4):e0175529.
Fagundes C, Barreto R, Guevara M, et al. A modified acute kidney injury classification for diagnosis and risk stratification of impairment of kidney function in cirrhosis. J Hepatol. 2013;59(3):474-481.
Angeli P, Gines P, Wong F, et al. Diagnosis and management of acute kidney injury in patients with cirrhosis: revised consensus recommendations of the International Club of Ascites. J Hepatol. 2015;62(4):968-974.
Boyer TD, Sanyal AJ, Garcia-Tsao G, et al. Predictors of response to terlipressin plus albumin in hepatorenal syndrome (HRS) type 1: relationship of serum creatinine to hemodynamics. J Hepatol. 2011;55(2):315-321.
Zhang Z, Maddukuri G, Jaipaul N, Cai CX. Role of renal replacement therapy in patients with type 1 hepatorenal syndrome receiving combination treatment of vasoconstrictor plus albumin. J Crit Care. 2015;30(5):969-974.
Belcher JM, Sanyal AJ, Peixoto AJ, et al. Kidney biomarkers and differential diagnosis of patients with cirrhosis and acute kidney injury. Hepatology. 2014;60(2):622-632.
Garg V, Garg H, Khan A, et al. Granulocyte colony-stimulating factor mobilizes CD34(+) cells and improves survival of patients with acute-on-chronic liver failure. Gastroenterology. 2012;142(3):505-512.e1.
Mookerjee RP, Pavesi M, Thomsen KL, et al. Treatment with non-selective beta blockers is associated with reduced severity of systemic inflammation and improved survival of patients with acute-on-chronic liver failure. J Hepatol. 2016;64(3):574-582.
Villanueva C, Albillos A, Genescà J, et al. β blockers to prevent decompensation of cirrhosis in patients with clinically significant portal hypertension (PREDESCI): a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2019;393(10181):1597-1608.
Verbeke L, Farre R, Verbinnen B, et al. The FXR agonist obeticholic acid prevents gut barrier dysfunction and bacterial translocation in cholestatic rats. Am J Pathol. 2015;185(2):409-419.
Goel A, Rahim U, Nguyen LH, Stave C, Nguyen MH. Systematic review with meta-analysis: rifaximin for the prophylaxis of spontaneous bacterial peritonitis. Aliment Pharmacol Ther. 2017;46(11-12):1029-1036.
Philips CA, Pande A, Shasthry SM, et al. Healthy donor fecal microbiota transplantation in steroid-ineligible severe alcoholic hepatitis: a pilot study. Clin Gastroenterol Hepatol. 2017;15(4):600-602.
Bajaj JS, Kassam Z, Fagan A, et al. Fecal microbiota transplant from a rational stool donor improves hepatic encephalopathy: a randomized clinical trial. Hepatology. 2017;66(6):1727-1738.
Nath B, Szabo G. Alcohol-induced modulation of signaling pathways in liver parenchymal and nonparenchymal cells: implications for immunity. Semin Liver Dis. 2009;29(2):166-177.
Fernández J, Ruiz del Arbol L, Gómez C, et al. Norfloxacin vs ceftriaxone in the prophylaxis of infections in patients with advanced cirrhosis and hemorrhage. Gastroenterology. 2006;131(4):1049-1056.
Fernández J, Navasa M, Planas R, et al. Primary prophylaxis of spontaneous bacterial peritonitis delays hepatorenal syndrome and improves survival in cirrhosis. Gastroenterology. 2007;133(3):818-824.
Moreau R, Elkrief L, Bureau C, et al. Effects of long-term norfloxacin therapy in patients with advanced cirrhosis. Gastroenterology. 2018;155(6):1816-1827.e9.
Poca M, Alvarado-Tapias E, Concepcíon M, et al. Predictive model of mortality in patients with spontaneous bacterial peritonitis. Aliment Pharmacol Ther. 2016;44(6):629-637.
Terg R, Gadano A, Cartier M, et al. Serum creatinine and bilirubin predict renal failure and mortality in patients with spontaneous bacterial peritonitis: a retrospective study. Liver Int. 2009;29(3):415-419.
Assimakopoulos SF, Tsamandas AC, Tsiaoussis GI, et al. Intestinal mucosal proliferation, apoptosis and oxidative stress in patients with liver cirrhosis. Ann Hepatol. 2013;12(2):301-307.
Ginés P, Rimola A, Planas R, et al. Norfloxacin prevents spontaneous bacterial peritonitis recurrence in cirrhosis: results of a double-blind, placebo-controlled trial. Hepatology. 1990;12(4 Pt 1):716-724.
Elfert A, Abo Ali L, Soliman S, Ibrahim S, Abd-Elsalam S. Randomized-controlled trial of rifaximin versus norfloxacin for secondary prophylaxis of spontaneous bacterial peritonitis. Eur J Gastroenterol Hepatol. 2016;28(12):1450-1454.
Angeli P, Bernardi M, Villanueva C, et al. EASL Clinical Practice Guidelines for the management of patients with decompensated cirrhosis. J Hepatol. 2018;69(2):406-460.
Vlachogiannakos J, Viazis N, Vasianopoulou P, et al. Long-term administration of rifaximin improves the prognosis of patients with decompensated alcoholic cirrhosis. J Gastroenterol Hepatol. 2013;28(3):450-455.
Horvath A, Leber B, Schmerboeck B, et al. Randomised clinical trial: the effects of a multispecies probiotic vs. placebo on innate immune function, bacterial translocation and gut permeability in patients with cirrhosis. Aliment Pharmacol Ther. 2016;44(9):926-935.
Prasad AS. Zinc is an antioxidant and anti-inflammatory agent: its role in human health. Front Nutr. 2014;1:14.
Anty R, Canivet CM, Patouraux S, et al. Severe vitamin D deficiency may be an additional cofactor for the occurrence of alcoholic steatohepatitis. Alcohol Clin Exp Res. 2015;39(6):1027-1033.
Abraldes JG, Villanueva C, Aracil C, et al. Addition of simvastatin to standard therapy for the prevention of variceal rebleeding does not reduce rebleeding but increases survival in patients with cirrhosis. Gastroenterology. 2016;150(5):1160-1170.e3.
Piano S, Brocca A, Mareso S, Angeli P. Infections complicating cirrhosis. Liver Int. 2018;38 Suppl 1:126-133.
Lee JY, Han SH, Park SH, et al. Ascitic fluid infection in patients with hepatic cirrhosis: culture-negative neutrocytic ascites versus spontaneous bacterial peritonitis. J Korean Med Sci. 2014;29(4):487-494.
Piano S, Fasolato S, Salinas F, et al. The empirical antibiotic treatment of nosocomial spontaneous bacterial peritonitis: Results of a randomized, controlled clinical trial. Hepatology. 2016;63(4):1299-1309.
Fernández J, Prado V, Trebicka J, et al. Multidrug-resistant bacterial infections in patients with decompensated cirrhosis and with acute-on-chronic liver failure in Europe. J Hepatol. 2019;70(3):398-411.
Bassetti M, Peghin M, Vena A, Giacobbe DR. Treatment of infections due to MDR Gram-negative bacteria. Front Med (Lausanne). 2019;6:74.
Brilhante RSN, Oliveira JS, Evangelista AJJ, et al. Candida albicans from patients with chronic kidney disease exhibit increased echinocandin susceptibility. Front Microbiol. 2018;9:2688.
Runyon BA. Monomicrobial nonneutrocytic bacterascites: a variant of spontaneous bacterial peritonitis. Hepatology. 1990;12(4 Pt 1):710-715.
Enomoto H, Inoue S, Matsuhisa A, Nishiguchi S. Diagnosis of spontaneous bacterial peritonitis and an in situ hybridization approach to detect an "unidentified" pathogen. Int J Hepatol. 2014;2014:634617.
Lutz P, Goeser F, Kaczmarek DJ, et al. Relative ascites polymorphonuclear cell count indicates bacterascites and risk of spontaneous bacterial peritonitis. Dig Dis Sci. 2017;62(9):2558-2568.
Moreau R, Jalan R, Gines P, et al. Acute-on-chronic liver failure is a distinct syndrome that develops in patients with acute decompensation of cirrhosis. Gastroenterology. 2013;144(7):1426-1437.e9.
Caraceni P, Riggio O, Angeli P, et al. Long-term albumin administration in decompensated cirrhosis (ANSWER): an open-label randomised trial. Lancet. 2018;391(10138):2417-2429.
Larsen FS, Schmidt LE, Bernsmeier C, et al. High-volume plasma exchange in patients with acute liver failure: an open randomised controlled trial. J Hepatol. 2016;64(1):69-78.
Duan XZ, Liu FF, Tong JJ, et al. Granulocyte-colony stimulating factor therapy improves survival in patients with hepatitis B virus-associated acute-on-chronic liver failure. World J Gastroenterol. 2013;19(7):1104-1110.
Artru F, Louvet A, Ruiz I, et al. Liver transplantation in the most severely ill cirrhotic patients: a multicenter study in acute-on-chronic liver failure grade 3. J Hepatol. 2017;67(4):708-715.
Runyon BA, Hoefs JC. Ascitic fluid analysis in the differentiation of spontaneous bacterial peritonitis from gastrointestinal tract perforation into ascitic fluid. Hepatology. 1984;4(3):447-450.
Kim JH, Kim YS, Choi HY, Kwon K, Won IS. Clinical and microbiological characteristics of spontaneous bacterial peritonitis in patients with malignant ascites. Korean J Hepatol. 2010;16(1):49-56.
Attia YM, Abou El Azm AR, Ibrahim SL, Ibrahim HM. Diagnostic value of ascitic calprotectin in the diagnosis of spontaneous bacterial peritonitis. Arab J Gastroenterol. 2020;21(3):164-169.
Schwabl P, Bucsics T, Soucek K, et al. Risk factors for development of spontaneous bacterial peritonitis and subsequent mortality in cirrhotic patients with ascites. Liver Int. 2015;35(9):2121-2128.
Bajaj JS, Vargas HE, Reddy KR, et al. Association between intestinal microbiota collected at hospital admission and outcomes of patients with cirrhosis. Clin Gastroenterol Hepatol. 2019;17(4):756-765.e3.
Bruns T, Zimmermann HW, Stallmach A. Risk factors and outcome of bacterial infections in cirrhosis. World J Gastroenterol. 2014;20(10):2542-2554.
Wiest R, Krag A, Gerbes A. Spontaneous bacterial peritonitis: recent guidelines and beyond. Gut. 2012;61(2):297-310.
Jalan R, Fernandez J, Wiest R, et al. Bacterial infections in cirrhosis: a position statement based on the EASL Special Conference 2013. J Hepatol. 2014;60(6):1310-1324.
Leevy CB, Phillips JA. Hospitalizations during the use of rifaximin versus lactulose for the treatment of hepatic encephalopathy. Dig Dis Sci. 2007;52(3):737-741.
Sanduzzi Zamparelli M, Rocco A, Compare D, Nardone G. The gut microbiota: a new potential driving force in liver cirrhosis and hepatocellular carcinoma. United European Gastroenterol J. 2017;5(7):944-953.
Bernsmeier C, van der Merwe S, Périanin A. The innate immune cells in cirrhosis. J Hepatol. 2020;73(1):186-201.
Harrison SA, Wong VW, Okanoue T, et al. Selonsertib for patients with bridging fibrosis or compensated cirrhosis due to NASH: Results from randomized phase III STELLAR trials. J Hepatol. 2020;73(1):26-39.
Shi M, Zhang Z, Xu R, et al. Human mesenchymal stem cell transfusion is safe and improves liver function in acute-on-chronic liver failure patients. Stem Cells Transl Med. 2012;1(10):725-731.
No comments:
Post a Comment