Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 6543 2022-11-30 14:01:12 |
2 Reference format revised. -4221 word(s) 2322 2022-12-01 09:25:57 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Huang, C.;  Lee, C.;  Chang, C. Spontaneous Bacterial Peritonitis in Decompensated Liver Cirrhosis. Encyclopedia. Available online: https://encyclopedia.pub/entry/37350 (accessed on 27 July 2024).
Huang C,  Lee C,  Chang C. Spontaneous Bacterial Peritonitis in Decompensated Liver Cirrhosis. Encyclopedia. Available at: https://encyclopedia.pub/entry/37350. Accessed July 27, 2024.
Huang, Chien-Hao, Chen-Hung Lee, Ching Chang. "Spontaneous Bacterial Peritonitis in Decompensated Liver Cirrhosis" Encyclopedia, https://encyclopedia.pub/entry/37350 (accessed July 27, 2024).
Huang, C.,  Lee, C., & Chang, C. (2022, November 30). Spontaneous Bacterial Peritonitis in Decompensated Liver Cirrhosis. In Encyclopedia. https://encyclopedia.pub/entry/37350
Huang, Chien-Hao, et al. "Spontaneous Bacterial Peritonitis in Decompensated Liver Cirrhosis." Encyclopedia. Web. 30 November, 2022.
Spontaneous Bacterial Peritonitis in Decompensated Liver Cirrhosis
Edit

Spontaneous bacterial peritonitis (SBP) is defined as a bacterial infection of the ascitic fluid without a surgically treatable intra-abdominal infection source. SBP is a common, severe complication in cirrhosis patients with ascites, and if left untreated, in-hospital mortality may exceed 90%. However, the incidence of SBP has been lowered to approx. 20% through early diagnosis and antibiotic therapy. There are three types of SBP. Bacterial translocation from the gastrointestinal tract is the most common source of SBP. Distinguishing SBP from secondary bacterial peritonitis is essential because the conditions require different therapeutic strategies. The standard treatment for SBP is prompt broad-spectrum antibiotic administration and should be tailored according to community-acquired SBP, healthcare-associated or nosocomial SBP infections, and local resistance profile. Albumin supplementation, especially in patients with renal impairment, is also beneficial. Selective intestinal decontamination is associated with a reduced risk of bacterial infection and mortality in the high-risk group.

spontaneous bacterial peritonitis liver cirrhosis culture-negative neutrophilic ascites monomicrobial non-neutrocytic bacterascites bacterial translocation ascites fluid Gram-negative bacilli Gram-positive cocci antibiotic prophylaxis

1. Introduction

Cirrhotic patients have an altered defense against bacteria associated with reduced bacterial clearance [1]. This immune defect facilitates bacterial translocation induced by increased intestinal permeability and gut bacterial overgrowth [2]. Therefore, bacterial infection is either present on admission or develops during hospitalization in about 30% of patients with cirrhosis [3], and the most common form of these infections is spontaneous bacterial peritonitis (SBP) [3].
SBP is a severe complication in cirrhosis patients with ascites [4]. Ascites is mainly transudative fluid with poor opsonic activity, which provides a favorable environment for growth of bacteria. The prevalence of SBP is 1.5–3.5% among outpatients and 10–30% among hospitalized patients [5]. When first reported, in-hospital mortality from an episode of SBP exceeded 90%; however, this rate has been lowered to approximately 20% through early diagnosis and prompt antibiotic therapy [6].
SBP is diagnosed upon positive ascites culture and/or absolute neutrophil count (polymorphonuclear cell or PMN) within ascites fluid (AF) of ≧250 cells/mm3 [7][8]. Diagnosis is distinct from secondary peritonitis and hence is made in the absence of an intra-abdominal source of infection or other causes of an elevated ascites neutrophil count, such as hemorrhage, pancreatitis, peritoneal tuberculosis, and carcinomatosis [7], or an evident intra-abdominal, surgically treatable source [9][10].
There are three types of SBP (Table 1): (1) classic SBP; polymorphonuclear cell (PMN, also referred to as neutrophils) count in ascitic fluid is ≥250 cells/mm3 and positive ascites culture; (2) culture-negative neutrophilic ascites (CNNA), ascites with a PMN count of ≥250 cells/mm3 but with negative ascitic fluid culture; (3) monomicrobial non-neutrocytic bacterascites (MNB), PMN not elevated in ascites, but culture is positive.
Table 1. Three types of SBP.
Ascites Fluid Classic SBP CNNA 1 MNB 2
PMN count (cells/mm3) ≥250 ≥250 <250
Ascites culture positive negative positive
1 CNNA: culture-negative neutrophilic ascites; 2 MNB: monomicrobial non-neutrocytic bacterascites.

2. Pathogenesis

Mechanisms that may be involved in the pathogenesis of SBP are shown in Figure 1.
Figure 1. Mechanisms involved in the pathogenesis of SBP.

2.1. Gut Dysbiosis

One of the early stages in the development of SBP is the disturbance of gut flora leading to bacterial overgrowth and extra-intestinal dissemination of gut microorganisms [11]. Edema of the small intestine and ascending colon alters tight junction integrity and increases intestinal permeability, thus predisposing the patient to bacterial overgrowth in the presence of cirrhosis [11]. Altered small intestinal motility, presence of hypochlorhydria due to the use of proton pump inhibitors, and immunosuppression therapies commonly used during cirrhosis may also contribute to bacterial overgrowth.

2.2. Bacterial Translocation

Another important step following bacterial overgrowth is the translocation of enteric bacteria to extraintestinal sites, such as the mesenteric lymph nodes (MLN), which drain lymph from the gut [11]. Bacterial translocation (BT) has been defined as culture-positive MLN [12] and is culture-negative in healthy experimental animals without cirrhosis [12] but culture pathogenic bacteria in 78.1% of animals with cirrhosis and ascites [11]. The fact that SBP is monomicrobial implies that there are “filters” between polymicrobial intestinal sources and the ascitic fluid [11]. The first filter is the gut mucosa itself, and the second filter is the MLN [11]. If these MLN fail to sequester and destroy the bacteria, the pathogens can move from the mesenteric lymphatic system to systemic circulation and then percolate through the liver and extravasate across Glisson’s capsule to enter the ascitic fluid [13].

2.3. Impaired Host Defense System

Conversely, the host defense system also plays an important role in SBP pathogenesis. Once a microorganism enters the ascitic fluid, a battle ensues between the invading bacteria and the host’s immune system. Peritoneal macrophages are the first line of defense in the peritoneal cavity [14][15]. If these phagocytes fail to eradicate the invading microorganism, the complement system is activated, and cytokines are released [16]. Polymorphonuclear leukocytes (PMNs) then enter the peritoneum to eliminate the foreign bacteria.
However, cirrhotic patients are known to have impairment in neutrophil and reticuloendothelial function [17][18]. In addition, many cirrhotic patients with ascites have a complement or opsonin deficiency [19]. The opsonic activity of ascitic fluid parallels the ascitic fluid total protein concentration [20]. Since opsonins are required by phagocytic cells to eliminate the offending microorganisms, cirrhotic patients with an ascitic fluid protein concentration of less than or equal to (≦) 1 g/dL are 10-times more likely to develop SBP during hospitalization than those with a protein concentration greater than (>) 1 g/dL [19].

3. Bacteriology

Bacterial translocation (BT) from the GI tract is the most common source of SBP. However, especially in nosocomial SBP, other sources, such as transient bacteremia due to invasive procedures, can also lead to SBP [21]. The bacteriology of SBP can be classified into Gram-negative bacilli, Gram-positive cocci, multidrug-resistant microorganisms, and anaerobes by bacterial spectrum, and classified into community acquired (CA), healthcare-associated (HCA), and nosocomial (hospital-acquired) infections by facilities.
Two thirds (66.7%) of SBP cases are caused by Gram-negative bacilli (GNB) from BT, almost exclusively Enterobacteriaceae, and occur independently from the site of acquisition [3][22]Escherichia coli (E. coli) is the most frequently isolated pathogen (46–70%) [3][23], followed by Streptococcus pneumoniae (18–19%) and Klebsiella (9–13%) [22][23].  On the other hand, Gram-positive organisms accounted for less than one third (33.3%) of SBP and were predominated by Streptococcus (60%) and Staphylococcus aureus (40%) [22][24]. The emergence of multidrug-resistant (MDR) microorganisms, such as extended-spectrum β-lactamase (ESBL)-producing GNB and enterococci [25], fluroquinolone-resistant (QR) GNB [26], cefoxitin/methicillin-resistant Staphylococcus aureus (MRSA) [27], vancomycin-resistant enterococcus (VRE), and other resistant microorganisms [25] have altered prior conceptions toward SBP bacteriology and treatment [28].  Although gut floras are responsible for the majority of SBP cases, anaerobes appear to be rare, presumably due to the high oxygen content of the intestinal wall and AF, as well as because of the relative inability of anaerobes to translocate across the intestinal mucosa [29][30].

4. Diagnosis

Spontaneous bacterial peritonitis (SBP) should be suspected in patients with cirrhosis who develop signs or symptoms, such as fever (69%), abdominal pain (59%), altered mental status (54%), abdominal tenderness (49%), diarrhea (32%), ileus (30%), hypotension/shock (21%), or hypothermia (17%) [29]. However, 10% of cases show no signs or symptoms, partly because a large volume of ascites prevents contact of the visceral and parietal peritoneal surfaces to elicit the spinal reflux that cause abdominal rigidity [29].

A diagnostic paracentesis should be performed in all patients with cirrhosis and ascites who require emergency room care or hospitalization, who demonstrate or report signs/symptoms mentioned above in the clinical presentations, or who present gastrointestinal bleeding, in order to confirm evidence of SBP [31]. However, low clinical suspicion for SBP does not preclude the necessity for paracentesis, since 10% of cases have no signs or symptoms [29]

Ascitic fluid tests should include cell count with a differential, Gram stain, culture, total protein, and albumin to calculate the serum-ascites albumin gradient (SAAG), if not already known [27]. When the diagnosis of cirrhosis is not definite, an ascites SAAG greater than or equal to (≧) 1.1 g/dl is ascribed to portal hypertension with approximately 97% accuracy [31]. Total ascitic fluid protein concentration should be measured to assess the risk of SBP since patients suffering from ascites with a total protein concentration lower than (<) 1.5 g/dL are at increased risk of SBP [7].

A diagnosis of (1) classic SBP is made if PMN count in the ascitic fluid is ≥250 cells/mm3, culture results are positive, and secondary causes of peritonitis are excluded [7][31]. A potential source of error in PMN count is hemorrhage into the ascitic fluid, such as with traumatic paracentesis, which can cause both red and white blood cells to enter the ascites. A corrected PMN count should be calculated if there are bloody ascites by subtracting one PMN from the absolute PMN count for every 250 red cells/mm3 [32]. Distinguishing SBP from secondary bacterial peritonitis is essential because the conditions require different therapeutic strategies. Mortality from SBP can be as high as 85% if a patient undergoes an unnecessary exploratory laparotomy [33], while mortality of secondary bacterial peritonitis can exceed 80% if treatment consists of antibiotics without surgical intervention [9].

5. Treatment

The standard treatment for SBP is prompt broad-spectrum antibiotic administration and albumin supplementation, especially in patients with renal impairment (RI) [34].

5.1. Antibiotic Therapy

If SBP is suspected, antibiotic therapy must be initiated immediately after AF and culture to reduce complications and mortality [7]. Potentially nephrotoxic antibiotics (i.e., aminoglycosides) should be avoided [35] since patients with SBP are highly sensitive to aminoglycosides-related nephrotoxicity and fatal renal failure is common even at sub-toxic doses [29].
Two decades ago, most cases of SBP were attributed to third generation cephalosporin-sensitive Enterobacteriaceae. Now, risk factors, such as repeated hospitalizations, invasive procedures, and frequent exposure to antibiotics either as prophylaxis or as treatment [3], have led to the development of infections caused by MDR microorganisms. Bacterial resistance carries a 3.87-fold relative increased risk of mortality in patients with SBP [21]. Particularly, nosocomial SBP has been associated with multi-drug resistance (HR = 4.43) and poor outcome (50% in-hospital mortality). One prospective study demonstrated that failure of recommended empirical antibiotic regimens can have a negative impact on mortality [36]. Therefore, it is important to distinguish community-acquired SBP from healthcare-associated and nosocomial SBP, as well as consider the severity of infection and local resistance profile before implementing antibiotic therapy [34] (Figure 2). Subsequently, de-escalation according to bacterial susceptibility based on positive culture is recommended to minimize resistance selection pressure.
Figure 2. Recommended antibiotic treatment strategy for SBP since 2018.

5.2. Albumin Supplement in Patients with Renal Impairment

The administration of albumin (given 1.5 g/kg at diagnosis and 1 g/kg on day 3, maximum 100 g) is recommended in patients with SBP [37], especially in those with serum creatinine of >1 mg/dL, blood urea nitrogen of >30 mg/dL, or total bilirubin of >4 mg/dL [38]. In one meta-analysis of four RCTs (288 patients), albumin infusion prevented renal impairment and reduced mortality among patients with SBP [39].

5.3. Discontinue NSBB in Patients with SBP

Given that NSBB may interfere with systemic hemodynamics, effect on outcome was examined in a retrospective study of 607 patients with cirrhosis and ascites [40]. Once SBP developed, patients receiving NSBB suffered lower transplant-free survival (HR = 1.58; 95% CI: 1.098–2.274). In addition, a higher proportion of patients on NSBBs suffered hepatorenal syndrome (24% vs. 11%, p = 0.027) [40]. Therefore, NSBB should be discontinued after a diagnosis of SBP.

5.4. Other Novel Therapeutic Strategies

The emergence of MDR bacteria may require novel therapeutic strategies that do not involve the use of antibiotics. One potential approach is human amniotic mesenchymal stromal cell (hA-MSC) treatment. One in vitro study found hA-MSCs added to ascites fluid could significantly reduce the proliferation of both bacterial strains at 24 h as well as affect M1/M2 polarization, C3a complement protein, and ficolin 3 concentrations during the course of infection in a strain-dependent manner [41]. Validation of an in vivo model is warranted for future hA-MSC application in treating ascites infected with carbapenem-resistant bacteria [41].

6. Conclusions

Spontaneous bacterial peritonitis (SBP) is a severe complication in cirrhosis patients with ascites. Clinical awareness, prompt diagnosis by exclusion of secondary bacterial peritonitis, and immediate treatment are necessary to reduce mortality and morbidity in this patient group. However, the emergence of multidrug-resistant (MDR) microorganisms have changed the understanding of SBP bacteriology and treatment. Antibiotic therapy specific to either community-acquired or nosocomial/healthcare-acquired SBP is ideal, while liver transplantation remains the definitive treatment following SBP. Prevention of SBP recurrence by antibiotic prophylaxis while patients wait for a liver transplant is therefore an important clinical issue. The poorly absorbed antibiotic rifaximin may be effective for both primary and secondary SBP prophylaxis, but additional prospective studies are required. Further development of non-antibiotic strategies based on pathogenic mechanisms are also urgently needed. Blind studies that avoid post-randomization dropout and consider clinically relevant outcomes, such as mortality, health-related quality of life, and decompensation events, are desired for future research.
There are three types of SBP. Bacterial translocation from the GI tract is the most common source of SBP. Therefore, two thirds of SBP cases were caused by Gram-negative bacilli, almost exclusively Enterobacteriaceae. Escherichia coli (E. coli) is the most frequently isolated pathogen. However, a trend of Gram-positive cocci (GPC)-associated SBP has been demonstrated in recent years, representing a changing paradigm in the known bacteriology of SBP, especially in nosocomial SBP; other sources, such as transient bacteremia due to invasive procedures, can also lead to SBP Gram-positive cocci (GPC), such as Staphylococcus, Enterococcus, as well as multi-resistant bacteria have become common pathogens and have changed the conventional approach to treatment of SBP. Healthcare-associated and nosocomial SBP infections should prompt greater vigilance and consideration for alternative antibiotic coverage. Acid suppressive and beta-adrenergic antagonist therapies are strongly associated with SBP in at-risk individuals. A diagnostic paracentesis should be performed in all patients with cirrhosis and ascites who require emergency room care or hospitalization, who demonstrate or report signs/symptoms mentioned above in the clinical presentations, or who present gastrointestinal bleeding, in order to confirm evidence of SBP. Distinguishing SBP from secondary bacterial peritonitis is essential because the conditions require different therapeutic strategies. Since SBP may be regarded as the final clinical stage of liver cirrhosis [42], one-year overall mortality rates range from 53.9 [43] to 78% [43][44][45]. Liver transplantation should be seriously considered for SBP survivors who are good candidates for transplantation. The standard treatment for SBP is prompt broad-spectrum antibiotic administration and should be tailored according to either CAP or hospital-acquired, or to local resistance profiles. Albumin supplementation, especially in patients with renal impairment (RI) is also beneficial. Not all patients with cirrhosis and ascites require antibiotic prophylaxis, sometimes referred to as selective intestinal decontamination (SID). SID is associated with a reduced risk of bacterial infection [46][47][48][49] and mortality.

References

  1. Wasmuth, H.E.; Kunz, D.; Yagmur, E.; Timmer-Stranghöner, A.; Vidacek, D.; Siewert, E.; Bach, J.; Geier, A.; Purucker, E.A.; Gressner, A.M. Patients with acute on chronic liver failure display ‘sepsis-like’immune paralysis. J. Hepatol. 2005, 42, 195–201.
  2. Wiest, R.; Garcia-Tsao, G. Bacterial translocation (BT) in cirrhosis. Hepatology 2005, 41, 422–433.
  3. Fernández, J.; Navasa, M.; Gómez, J.; Colmenero, J.; Vila, J.; Arroyo, V.; Rodés, J. Bacterial infections in cirrhosis: Epidemiological changes with invasive procedures and norfloxacin prophylaxis. Hepatology 2002, 35, 140–148.
  4. Huang, C.H.; Lin, C.Y.; Sheen, I.S.; Chen, W.T.; Lin, T.N.; Ho, Y.P.; Chiu, C.T. Recurrence of spontaneous bacterial peritonitis in cirrhotic patients non-prophylactically treated with norfloxacin: Serum albumin as an easy but reliable predictive factor. Liver Int. 2011, 31, 184–191.
  5. Aithal, G.P.; Palaniyappan, N.; China, L.; Harmala, S.; Macken, L.; Ryan, J.M.; Wilkes, E.A.; Moore, K.; Leithead, J.A.; Hayes, P.C.; et al. Guidelines on the management of ascites in cirrhosis. Gut 2021, 70, 9–29.
  6. Garcia-Tsao, G. Current management of the complications of cirrhosis and portal hypertension: Variceal hemorrhage, ascites, and spontaneous bacterial peritonitis. Gastroenterology 2001, 120, 726–748.
  7. Rimola, A.; García-Tsao, G.; Navasa, M.; Piddock, L.J.; Planas, R.; Bernard, B.; Inadomi, J.M. Diagnosis, treatment and prophylaxis of spontaneous bacterial peritonitis: A consensus document. J. Hepatol. 2000, 32, 142–153.
  8. Runyon, B.A.; AASLD. Introduction to the revised American Association for the Study of Liver Diseases Practice Guideline management of adult patients with ascites due to cirrhosis 2012. Hepatology 2013, 57, 1651–1653.
  9. Akriviadis, E.A.; Runyon, B.A. Utility of an algorithm in differentiating spontaneous from secondary bacterial peritonitis. Gastroenterology 1990, 98, 127–133.
  10. Runyon, B.A.; Committee, A.P.G. Management of adult patients with ascites due to cirrhosis: An update. Hepatology 2009, 49, 2087–2107.
  11. Runyon, B.A.; Squier, S.; Borzio, M. Translocation of gut bacteria in rats with cirrhosis to mesenteric lymph nodes partially explains the pathogenesis of spontaneous bacterial peritonitis. J. Hepatol. 1994, 21, 792–796.
  12. Berg, R.D.; Garlington, A.W. Translocation of certain indigenous bacteria from the gastrointestinal tract to the mesenteric lymph nodes and other organs in a gnotobiotic mouse model. Infect. Immun. 1979, 23, 403–411.
  13. Vilela, E.G.; Thabut, D.; Rudler, M.; Bittencourt, P.L. Management of Complications of Portal Hypertension. Can. J. Gastroenterol. Hepatol. 2019, 2019, 6919284.
  14. Dunn, D.L.; Barke, R.A.; Knight, N.B.; Humphrey, E.W.; Simmons, R.L. Role of resident macrophages, peripheral neutrophils, and translymphatic absorption in bacterial clearance from the peritoneal cavity. Infect. Immun. 1985, 49, 257–264.
  15. Ruiz-Alcaraz, A.J.; Martinez-Banaclocha, H.; Marin-Sanchez, P.; Carmona-Martinez, V.; Iniesta-Albadalejo, M.A.; Tristan-Manzano, M.; Tapia-Abellan, A.; Garcia-Penarrubia, P.; Machado-Linde, F.; Pelegrin, P.; et al. Isolation of functional mature peritoneal macrophages from healthy humans. Immunol. Cell Biol. 2020, 98, 114–126.
  16. Charles, A.; Janeway, J.; Travers, P.; Walport, M.; Shlomchik, M.J. Immunobiology: The Immune System in Health and Disease, 5th ed.; Garland Publishing: New York, NY, USA, 2001.
  17. Huang, C.H.; Jeng, W.J.; Ho, Y.P.; Teng, W.; Hsieh, Y.C.; Chen, W.T.; Chen, Y.C.; Lin, H.H.; Sheen, I.S.; Lin, C.Y. Increased EMR2 expression on neutrophils correlates with disease severity and predicts overall mortality in cirrhotic patients. Sci. Rep. 2016, 6, 38250.
  18. Rimola, A.; Soto, R.; Bory, F.; Arroyo, V.; Piera, C.; Rodes, J. Reticuloendothelial system phagocytic activity in cirrhosis and its relation to bacterial infections and prognosis. Hepatology 1984, 4, 53–58.
  19. Runyon, B.A. Low-protein-concentration ascitic fluid is predisposed to spontaneous bacterial peritonitis. Gastroenterology 1986, 91, 1343–1346.
  20. Hoefs, J.C.; Runyon, B.A. Spontaneous bacterial peritonitis. Dis. Mon. 1985, 31, 1–48.
  21. Wiest, R.; Krag, A.; Gerbes, A. Spontaneous bacterial peritonitis: Recent guidelines and beyond. Gut 2012, 61, 297–310.
  22. Oladimeji, A.A.; Temi, A.P.; Adekunle, A.E.; Taiwo, R.H.; Ayokunle, D.S. Prevalence of spontaneous bacterial peritonitis in liver cirrhosis with ascites. Pan Afr. Med. J. 2013, 15, 128.
  23. Bhuva, M.; Ganger, D.; Jensen, D. Spontaneous bacterial peritonitis: An update on evaluation, management, and prevention. Am. J. Med. 1994, 97, 169–175.
  24. Runyon, B.A. Monomicrobial nonneutrocytic bacterascites: A variant of spontaneous bacterial peritonitis. Hepatology 1990, 12, 710–715.
  25. Alexopoulou, A.; Papadopoulos, N.; Eliopoulos, D.G.; Alexaki, A.; Tsiriga, A.; Toutouza, M.; Pectasides, D. Increasing frequency of gram-positive cocci and gram-negative multidrug-resistant bacteria in spontaneous bacterial peritonitis. Liver Int. 2013, 33, 975–981.
  26. Fernandez, J.; Acevedo, J.; Castro, M.; Garcia, O.; de Lope, C.R.; Roca, D.; Pavesi, M.; Sola, E.; Moreira, L.; Silva, A.; et al. Prevalence and risk factors of infections by multiresistant bacteria in cirrhosis: A prospective study. Hepatology 2012, 55, 1551–1561.
  27. Dever, J.B.; Sheikh, M.Y. Review article: Spontaneous bacterial peritonitis--bacteriology, diagnosis, treatment, risk factors and prevention. Aliment. Pharmacol. Ther. 2015, 41, 1116–1131.
  28. Reuken, P.A.; Pletz, M.W.; Baier, M.; Pfister, W.; Stallmach, A.; Bruns, T. Emergence of spontaneous bacterial peritonitis due to enterococci—Risk factors and outcome in a 12-year retrospective study. Aliment Pharm. 2012, 35, 1199–1208.
  29. Such, J.; Runyon, B.A. Spontaneous bacterial peritonitis. Clin. Infect. Dis. 1998, 27, 669–674, quiz 675–666.
  30. Sheckman, P.; Onderdonk, A.B.; Bartlett, J.G. Anaerobes in spontaneous peritonitis. Lancet 1977, 2, 1223.
  31. European Association for the Study of the Liver. EASL clinical practice guidelines on the management of ascites, spontaneous bacterial peritonitis, and hepatorenal syndrome in cirrhosis. J. Hepatol. 2010, 53, 397–417.
  32. Hoefs, J.C. Increase in ascites white blood cell and protein concentrations during diuresis in patients with chronic liver disease. Hepatology 1981, 1, 249–254.
  33. Garrison, R.N.; Cryer, H.M.; Howard, D.A.; Polk, H.C., Jr. Clarification of risk factors for abdominal operations in patients with hepatic cirrhosis. Ann. Surg. 1984, 199, 648–655.
  34. 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.
  35. Cabrera, J.; Arroyo, V.; Ballesta, A.M.; Rimola, A.; Gual, J.; Elena, M.; Rodes, J. Aminoglycoside nephrotoxicity in cirrhosis. Value of urinary beta 2-microglobulin to discriminate functional renal failure from acute tubular damage. Gastroenterology 1982, 82, 97–105.
  36. Umgelter, A.; Reindl, W.; Miedaner, M.; Schmid, R.M.; Huber, W. Failure of current antibiotic first-line regimens and mortality in hospitalized patients with spontaneous bacterial peritonitis. Infection 2009, 37, 2–8.
  37. Sort, P.; Navasa, M.; Arroyo, V.; Aldeguer, X.; Planas, R.; Ruiz-del-Arbol, L.; Castells, L.; Vargas, V.; Soriano, G.; Guevara, M.; 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, 403–409.
  38. Sigal, S.H.; Stanca, C.M.; Fernandez, J.; Arroyo, V.; Navasa, M. Restricted use of albumin for spontaneous bacterial peritonitis. Gut 2007, 56, 597–599.
  39. Salerno, F.; Navickis, R.J.; Wilkes, M.M. Albumin infusion improves outcomes of patients with spontaneous bacterial peritonitis: A meta-analysis of randomized trials. Clin. Gastroenterol. Hepatol. 2013, 11, 123–130.e1.
  40. Mandorfer, M.; Bota, S.; Schwabl, P.; Bucsics, T.; Pfisterer, N.; Kruzik, M.; Hagmann, M.; Blacky, A.; Ferlitsch, A.; Sieghart, W.; et al. Nonselective beta blockers increase risk for hepatorenal syndrome and death in patients with cirrhosis and spontaneous bacterial peritonitis. Gastroenterology 2014, 146, 1680–1690.e1.
  41. Pampalone, M.; Vitale, G.; Gruttadauria, S.; Amico, G.; Iannolo, G.; Douradinha, B.; Mularoni, A.; Conaldi, P.G.; Pietrosi, G. Human Amnion-Derived Mesenchymal Stromal Cells: A New Potential Treatment for Carbapenem-Resistant Enterobacterales in Decompensated Cirrhosis. Int. J. Mol. Sci. 2022, 23, 857.
  42. Huang, C.H.; Tseng, H.J.; Amodio, P.; Chen, Y.L.; Wang, S.F.; Chang, S.H.; Hsieh, S.Y.; Lin, C.Y. Hepatic Encephalopathy and Spontaneous Bacterial Peritonitis Improve Cirrhosis Outcome Prediction: A Modified Seven-Stage Model as a Clinical Alternative to MELD. J. Pers. Med. 2020, 10, 186.
  43. Hung, T.H.; Tsai, C.C.; Hsieh, Y.H.; Tsai, C.C. The long-term mortality of spontaneous bacterial peritonitis in cirrhotic patients: A 3-year nationwide cohort study. Turk. J. Gastroenterol. 2015, 26, 159–162.
  44. Andreu, M.; Sola, R.; Sitges-Serra, A.; Alia, C.; Gallen, M.; Vila, M.C.; Coll, S.; Oliver, M.I. Risk factors for spontaneous bacterial peritonitis in cirrhotic patients with ascites. Gastroenterology 1993, 104, 1133–1138.
  45. Bac, D.J. Spontaneous bacterial peritonitis: An indication for liver transplantation? Scand. J. Gastroenterol. Suppl. 1996, 218, 38–42.
  46. Gines, P.; Rimola, A.; Planas, R.; Vargas, V.; Marco, F.; Almela, M.; Forne, M.; Miranda, M.L.; Llach, J.; Salmeron, J.M.; et al. Norfloxacin prevents spontaneous bacterial peritonitis recurrence in cirrhosis: Results of a double-blind, placebo-controlled trial. Hepatology 1990, 12, 716–724.
  47. Soriano, G.; Guarner, C.; Teixido, M.; Such, J.; Barrios, J.; Enriquez, J.; Vilardell, F. Selective intestinal decontamination prevents spontaneous bacterial peritonitis. Gastroenterology 1991, 100, 477–481.
  48. Soriano, G.; Guarner, C.; Tomas, A.; Villanueva, C.; Torras, X.; Gonzalez, D.; Sainz, S.; Anguera, A.; Cusso, X.; Balanzo, J.; et al. Norfloxacin prevents bacterial infection in cirrhotics with gastrointestinal hemorrhage. Gastroenterology 1992, 103, 1267–1272.
  49. Grange, J.D.; Roulot, D.; Pelletier, G.; Pariente, E.A.; Denis, J.; Ink, O.; Blanc, P.; Richardet, J.P.; Vinel, J.P.; Delisle, F.; et al. Norfloxacin primary prophylaxis of bacterial infections in cirrhotic patients with ascites: A double-blind randomized trial. J. Hepatol. 1998, 29, 430–436.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 487
Revisions: 2 times (View History)
Update Date: 01 Dec 2022
1000/1000
Video Production Service