733
Views
1
CrossRef citations to date
0
Altmetric
Review

An update on endotoxin neutralization strategies in Gram-negative bacterial infections

, , ORCID Icon, , , , & show all
Pages 495-517 | Received 19 Jan 2020, Accepted 07 Oct 2020, Published online: 19 Nov 2020

References

  • Sahly H, Aucken H, Benedi VJ, et al. Impairment of respiratory burst in polymorphonuclear leukocytes by extended-spectrum beta-lactamase-producing strains of Klebsiella pneumoniae. Eur J Clin Microbiol Infect Dis. 2004;23(1):20–26.
  • Asokan GV, Ramadhan T, Ahmed E, et al. WHO global priority pathogens list: a bibliometric analysis of medline-PubMed for knowledge mobilization to infection prevention and control practices in bahrain. Oman Med J. 2019;34:184–193.
  • Correa W, Heinbockel L, Behrends J, et al. Antibacterial action of synthetic antilipopolysaccharide peptides (SALP) involves neutralization of both membrane-bound and free toxins. Febs J. 2019;286:1576–1593.
  • Trautmann M, Heinemann M, Moricke A, et al. Endotoxin release due to ciprofloxacin measured by three different methods. J Chemother. 1999;11:248–254.
  • Rietschel ET, Brade L, Schade U, et al. Bacterial lipopolysaccharides: relationship of structure and conformation to endotoxic activity, serological specificity and biological function. Adv Exp Med Biol. 1990;256:81–99.
  • Poltorak A, He X, Smirnova I, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282:2085–2088.
  • Schromm AB, Lien E, Henneke P, et al. Molecular genetic analysis of an endotoxin nonresponder mutant cell line: a point mutation in a conserved region of MD-2 abolishes endotoxin-induced signaling. J Exp Med. 2001;194:79–88.
  • Schumann RR, Leong SR, Flaggs GW, et al. Structure and function of lipopolysaccharide binding protein. Science. 1990;249:1429–1431.
  • Kayagaki N, Wong MT, Stowe IB, et al. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science. 2013;341:1246–1249.
  • Park BS, Song DH, Kim HM, et al. The structural basis of lipopolysaccharide recognition by the TLR4-MD-2 complex. Nature. 2009;458:1191–1195.
  • Gay NJ, Symmons MF, Gangloff M, et al. Assembly and localization of Toll-like receptor signalling complexes. Nat Rev Immunol. 2014;14:546–558.
  • Casson CN, Yu J, Reyes VM, et al. 2015; Human caspase-4 mediates noncanonical inflammasome activation against gram-negative bacterial pathogens; Proceedings of the National Academy of Sciences of the United States of America. 112; p. 6688–6693.
  • Hagar JA, Powell DA, Aachoui Y, et al. (Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science. 2013;341:1250–1253.
  • Brandenburg K, Heinbockel L, Correa W, et al. Peptides with dual mode of action: killing bacteria and preventing endotoxin-induced sepsis. Biochim Biophys Acta. 2016;1858:971–979.
  • Brandenburg K, Wiese A. Endotoxins: relationships between structure, function, and activity. CurrTopMedChem. 2004;4:1127–1146.
  • Correa W, Heinbockel L, Martinez deTejada G, et al. Synthetic anti-lipopolysaccharide peptides (SALPs) as effective inhibitors of pathogen-associated molecular patterns (PAMPs). Adv Exp Med Biol. 2019;1117:111–129.
  • Zasloff M. Antimicrobial peptides of multicellular organisms. Nature. 2002;415:389–395.
  • Scott MG, Hancock REW. Cationic antimicrobial peptides and their multifunctional role in the immune system. CritRevImmunol. 2000;20:407–431.
  • Sun Y, Shang D. Inhibitory effects of antimicrobial peptides on lipopolysaccharide-induced inflammation. Mediators Inflamm. 2015;167572 (8 pages).
  • Schadich E, Mason D, Cole AL. Neutralization of bacterial endotoxins by frog antimicrobial peptides. Microbiol Immunol. 2013;57:159–161.
  • Levin J. The horseshoe crab: a model for gram-negative sepsis in marine organisms and humans. Prog Clin Biol Res. 1988;272:3–15.
  • Brandenburg K, Howe J, Gutsman T, et al. The expression of endotoxic activity in the Limulus test as compared to cytokine production in immune cells. Curr Med Chem. 2009;16:2653–2660.
  • Saravanan R, Mohanram H, Mangesch J, et al. Structure, activity and interactions of the cysteine deleted analog of tachyplesin-1 with lipopolysaccharide micelle: mechanistic insights into outer-membrane permeabilization and endotoxin neutralization. Biochim Biophys Acta. 2012;1818(7):1613–1624.
  • Brandenburg K, Andrä J, Müller M, et al. Physicochemical properties of bacterial glycopolymers in relation to bioactivity. Carbohydr Res. 2003;338:2477–2489.
  • Brandenburg K, David A, Howe J, et al. Temperature dependence of the binding of endotoxins to the polycationic peptides polymyxin B and its nonapeptide. Biophys J. 2005;88:1845–1858.
  • Israelachvili JN. Thermodynamic principles of self-assembly in intermolecular & surface forces. London, San Diego, New York, Boston, Sydney, Tokyo, Toronto: Academic Press Ltd.; 1991. p. 341–365.
  • Hu L, Sun C, Wang S, et al. Lipopolysaccharide neutralization by a novel peptide derived from phosvitin. Int J Biochem Cell Biol. 2013;45:2622–2631.
  • Pulido D, Garcia-Mayoral MF, et al. Structural basis for endotoxin neutralization by the eosinophil cationic protein. Febs J. 2016;283:4176–4191.
  • Correa W, Brandenburg J, Behrends J, et al. Inactivation of bacteria by gamma-Irradiation to investigate the interaction with antimicrobial peptides. Biophys J. 2019;117:1805–1819.
  • Hao Y, Yang N, Wang X, et al. Killing of Staphylococcus aureus and Salmonella enteritidis and neutralization of lipopolysaccharide by 17-residue bovine lactoferricins: improved activity of Trp/Ala-containing molecules. Sci Rep. 2017;7:44278.
  • Li LH, Ju TC, Hsieh CY, et al. A synthetic cationic antimicrobial peptide inhibits inflammatory response and the NLRP3 inflammasome by neutralizing LPS and ATP. PloS One. 2017;12:e0182057.
  • Saravanan R, Holdbrook DA, Petrlova J, et al. Structural basis for endotoxin neutralisation and anti-inflammatory activity of thrombin-derived C-terminal peptides. Nat Commun. 2018;9:2762.
  • Shao C, Li W, Tan P, et al. Symmetrical modification of minimized dermaseptins to extend the spectrum of antimicrobials with endotoxin neutralization potency. Int J Mol Sci. 2019;20:20061417.
  • Ahmad B, Hanif Q, Xubiao W, et al. Expression and purification of hybrid LL-37Talpha1 peptide in Pichia pastoris and evaluation of Its Immunomodulatory and anti-inflammatory activities by LPS neutralization. Front Immunol. 2019;10:1365.
  • Brandenburg K, Jürgens G, Müller M, et al. Biophysical characterization of lipopolysaccharide and lipid A inactivation by lactoferrin. Biol Chem. 2001;382:1215–1225.
  • Vincent JL, Marshall JC, Dellinger RP, et al. I. S. S. I. talactoferrin in severe sepsis: results from the phase II/III oral tAlactoferrin in severe sepsis trial. Crit Care Med. 2015;43:1832–1838.
  • Brandenburg K, Howe J, Sanchez-Gomez, et al. Effective antimicrobial and anti-endotoxin activity of cationic peptides based on lactoferricin: a biophysical and microbiological study. Anti-infective Agents Med Chem. 2010;9:9–22. .
  • Brandenburg K, Koch MHJ, Seydel U. Phase diagram of deep rough mutant lipopolysaccharide from Salmonella minnesota R595. J Struct Biol. 1992;108:93–106.
  • Brandenburg K, Mayer H, Koch MH, et al. Influence of the supramolecular structure of free lipid A on its biological activity. Eur J Biochem. 1993;218:555–563.
  • Shang D, Zhang Q, Dong W, et al. The effects of LPS on the activity of Trp-containing antimicrobial peptides against Gram-negative bacteria and endotoxin neutralization. Acta Biomater. 2016;33:153–165.
  • Ma Z, Wei D, Yan P, et al. Characterization of cell selectivity, physiological stability and endotoxin neutralization capabilities of alpha-helix-based peptide amphiphiles. Biomaterials. 2015;52:517–530.
  • Pristovsek P, Feher K, Szilagyi L, et al. Structure of a synthetic fragment of the LALF protein when bound to lipopolysaccharide. J Med Chem. 2005;48:1666–1670.
  • Brandenburg K, Andrä J, Garidel P, et al. Peptide-based treatment of sepsis. Appl Microbiol Biotechnol. 2011;90:799–808.
  • Dankesreiter S, Hoess A, Schneider-Mergener J, et al. Synthetic endotoxin-binding peptides block endotoxin-triggered TNF-à production by macrophages in vitro and in vivo and prevent endotoxin-mediated toxic shock. J Immunol. 2000;164:4804–4811.
  • Andrä J, Lamata M, Martinez de Tejada G, et al. Cyclic antimicrobial peptides based on Limulus anti-lipopolysaccharide factor for neutralization of lipopolysaccharide. Biochem Pharmacol. 2004;68:1297–1307.
  • Jiao B, Freudenberg M, Galanos C. Characterization of the lipid A component of genuine smooth-form lipopolysaccharide. Eur J Biochem. 1989;180:515–518.
  • Kaconis Y, Kowalski I, Howe J, et al. Biophysical mechanisms of endotoxin neutralization by cationic amphiphilic peptides. Biophys J. 2011;100:2652–2661.
  • Opal SM, Cohen J. Clinical Gram-positive sepsis: does it fundamentally differ from Gram-negative bacterial sepsis? Crit Care Med. 1999;27:1608–1616.
  • Brandenburg K, Heinbockel L, Correa W, et al. Supramolecular structure of enterobacterial wild-type lipopolysaccharides (LPS), fractions thereof, and their neutralization by Pep19-2.5. J Struct Biol. 2016;194:68–77.
  • Barcena-Varela S, Martinez de Tejada G, Martin L, et al. Coupling killing to neutralization: combined therapy with ceftriaxone/Pep19-2.5 counteracts sepsis in rabbits. Exp Mol Med. 2017;49:e345.
  • Galanos C, Freudenberg MA, Reutter W. Galactosamine-induced sensitization to the lethal effects of endotoxin. Proc Natl Acad Sci USA. 1979;76:5939–5943.
  • Heinbockel L, Sanchez-Gomez S, Martinez de Tejada G, et al. Preclinical investigations reveal the broad-spectrum neutralizing activity of peptide Pep19-2.5 on bacterial pathogenicity factors. Antimicrob Agents Chemother. 2013;57:1480–1487.
  • Tyers M, Wright GD. Drug combinations: a strategy to extend the life of antibiotics in the 21st century. Nat Rev Microbiol. 2019;17:141–155.
  • Miller KA, Suresh Kumar EV, Wood SJ, et al. Lipopolysaccharide sequestrants: structural correlates of activity and toxicity in novel acylhomospermines. J Med Chem. 2005;48:2589–2599.
  • David SA, Silverstein R, Amura CR, et al. Lipopolyamines: novel antiendotoxin compounds that reduce mortality in experimental sepsis caused by gram-negative bacteria. Antimicrob Agents Chemother. 1999;43:912–919.
  • Sil D, Heinbockel L, Kaconis Y, et al. Biophysical mechanisms of the neutralization of endotoxins by lipopolyamines. Open Biochem J. 2013;7:82–93.
  • Brandenburg K, Schromm AB, Gutsmann T. Endotoxins: relationship between structure, function, and activity. Subcell Biochem. 2010;53:53–67.
  • Uppu DS, Haldar J. Lipopolysaccharide neutralization by cationic-amphiphilic polymers through pseudoaggregate formation. Biomacromolecules. 2016;17:862–873.
  • Richter W, Vogel V, Howe J, et al. Morphology, size distribution, and aggregate structure of lipopolysaccharide and lipid A dispersions from enterobacterial origin. Innate Immun. 2011;17:427–438.
  • Fink MP. Animal models of sepsis. Virulence. 2014;5:143–153.
  • Rittirsch D, Hoesel LM, Ward PA. The disconnect between animal models of sepsis and human sepsis. J Leukoc Biol. 2007;81:137–143.
  • Seok J, Warren HS, Cuenca AG, et al. Inflammation & host response to injury, L. S. C. R. P. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci USA. 2013;110:3507–3512.
  • Nemzek JA, Hugunin KM, Opp MR. Modeling sepsis in the laboratory: merging sound science with animal well-being. Comp Med. 2008;58:120–128.
  • Lilley E, Armstrong R, Clark N, et al. Refinement of animal models of sepsis and septic shock. Shock. 2015;43:304–316.
  • Lewis AJ, Seymour CW, Rosengart MR. Current murine models of sepsis. Surg Infect (Larchmt). 2016;17:385–393.
  • Mishra SK, Choudhury S. Experimental protocol for cecal ligation and puncture model of polymicrobial sepsis and assessment of vascular functions in mice. Methods Mol Biol. 2018;1717:161–187.
  • Parker SJ, Watkins PE. Experimental models of gram-negative sepsis. Brit J Surg. 2001;88:22–30.
  • Heinbockel L, Marwitz S, Barcena Varela S, et al. Therapeutical administration of peptide Pep19-2.5 and ibuprofen reduces inflammation and prevents lethal sepsis. PloS One. 2015;10:e0133291.
  • Silverstein R. D-galactosamine lethality model: scope and limitations. J Endotoxin Res. 2004;10:147–162.
  • Martinez de Tejada G, Heinbockel L, Ferrer-Espada R, et al. Lipoproteins/peptides are sepsis-inducing toxins from bacteria that can be neutralized by synthetic anti-endotoxin peptides. Sci Rep. 2015;5:14292.
  • van Lier D, Geven C, Leijte GP, et al. Experimental human endotoxemia as a model of systemic inflammation. Biochimie. 2019;159:99–106.
  • Takada H, Kotani S, Tsujimoto M, et al. Immunopharmacological activities of a synthetic counterpart of a biosynthetic lipid A precursor molecule and of its analogs. Infect Immun. 1985;48:219–227.
  • Fossum C, Hjertner B, Olofsson KM, et al. Expression of TLR4, MD2 and CD4 in equine blood leukocytes during endotoxin infusion and in intestinal tissues from healthy horses. Vet Immunol Immunopathol. 2012;150:141–148.
  • Ohto U, Fukase K, Miyake K, et al. Structural basis of species-specific endotoxin sensing by innate immune receptor TLR4/MD-2. Proc Natl Acad Sci USA. 2012;109:7421–7426.
  • Aida Y, Kusumoto K, Nakatomi K, et al. An analogue of lipid A and LPS from Rhodobacter sphaeroides inhibits neutrophil responses to LPS by blocking receptor recognition of LPS and by depleting LPS-binding protein in plasma. J Leukoc Biol. 1995;58:675–682.
  • Mullarkey M, Rose JR, Bristol J, et al. Inhibition of endotoxin response by e5564, a novel Toll-like receptor 4-directed endotoxin antagonist. J Pharmacol Exp Ther. 2003;304:1093–1102.
  • Rossignol DP, Wasan KM, Choo E, et al. Safety, pharmacokinetics, pharmacodynamics, and plasma lipoprotein distribution of eritoran (E5564) during continuous intravenous infusion into healthy volunteers. Antimicrob Agents Chemother. 2004;48:3233–3240.
  • Tidswell M, Tillis W, Larosa SP, et al. Eritoran sepsis study, G. Phase 2 trial of eritoran tetrasodium (E5564), a toll-like receptor 4 antagonist, in patients with severe sepsis. Crit Care Med. 2010;38:72–83.
  • Guyatt GH, Tugwell PX, Feeny DH, et al. A framework for clinical evaluation of diagnostic technologies. CMAJ. 1986;134:587–594.
  • Kim HM, Park BS, Kim JI, et al. Crystal structure of the TLR4-MD-2 complex with bound endotoxin antagonist Eritoran. Cell. 2007;130:906–917.
  • Shirey KA, Lai W, Scott AJ, et al. The TLR4 antagonist Eritoran protects mice from lethal influenza infection. Nature. 2013;497:498–502.
  • Shirey KA, Sunday ME, Lai W, et al. Novel role of gastrin releasing peptide-mediated signaling in the host response to influenza infection. Mucosal Immunol. 2019;12:223–231.
  • Takashima K, Matsunaga N, Yoshimatsu M, et al. Analysis of binding site for the novel small-molecule TLR4 signal transduction inhibitor TAK-242 and its therapeutic effect on mouse sepsis model. Brit J Pharmacol. 2009;157:1250–1262.
  • Matsunaga N, Tsuchimori N, Matsumoto T, et al. TAK-242 (resatorvid), a small-molecule inhibitor of Toll-like receptor (TLR) 4 signaling, binds selectively to TLR4 and interferes with interactions between TLR4 and its adaptor molecules. Mol Pharmacol. 2011;79:34–41.
  • Sha T, Iizawa Y, Ii M. Combination of imipenem and TAK-242, a Toll-like receptor 4 signal transduction inhibitor, improves survival in a murine model of polymicrobial sepsis. Shock. 2011;35:205–209.
  • Hua F, Tang H, Wang J, et al. TAK-242, an antagonist for Toll-like receptor 4, protects against acute cerebral ischemia/reperfusion injury in mice. J Cereb Blood Flow Metab. 2015;35:536–542.
  • Rice TW, Wheeler AP, Bernard GR, et al. A randomized, double-blind, placebo-controlled trial of TAK-242 for the treatment of severe sepsis. Crit Care Med. 2010;38:1685–1694.
  • Piazza M, Rossini C, Della Fiorentina S, et al. Glycolipids and benzylammonium lipids as novel antisepsis agents: synthesis and biological characterization. J Med Chem. 2009;52:1209–1213.
  • Perrin-Cocon L, Aublin-Gex A, Sestito SE, et al. TLR4 antagonist FP7 inhibits LPS-induced cytokine production and glycolytic reprogramming in dendritic cells, and protects mice from lethal influenza infection. Sci Rep. 2017;7:40791.
  • Facchini FA, Zaffaroni L, Minotti A, et al. Structure-activity relationship in monosaccharide-based toll-like receptor 4 (TLR4) antagonists. J Med Chem. 2018;61:2895–2909.
  • Borio A, Holgado A, Garate JA, et al. Disaccharide-based anionic amphiphiles as potent inhibitors of lipopolysaccharide-induced inflammation. ChemMedChem. 2018;13:2317–2331.
  • Kuronuma K, Mitsuzawa H, Takeda K, et al. Anionic pulmonary surfactant phospholipids inhibit inflammatory responses from alveolar macrophages and U937 cells by binding the lipopolysaccharide-interacting proteins CD14 and MD-2. J Biol Chem. 2009;284:25488–25500.
  • Numata M, Kandasamy P, Nagashima Y, et al. Phosphatidylglycerol suppresses influenza A virus infection. Am J Respir Cell Mol Biol. 2012;46:479–487.
  • Spengler D, Winoto-Morbach S, Kupsch S, et al. Novel therapeutic roles for surfactant-inositols and -phosphatidylglycerols in a neonatal piglet ARDS model: a translational study. Am J Physiol Lung Cell Mol Physiol. 2018;314:L32–L53.
  • Pecorella F, Balsamo V, Collica F. [Neonatal thrombocytopenic purpura and hemolytic disease caused by Rh incompatibility in the female first-born infant of a transfused mother. Haematologica. 1966;51:453–484.
  • Pizzuto M, Lonez C, Baroja-Mazo A, et al. Saturation of acyl chains converts cardiolipin from an antagonist to an activator of Toll-like receptor-4. Cell Mol Life Sci. 2019;76:3667–3678.
  • Scott AJ, Oyler BL, Goodlett DR, et al. Lipid A structural modifications in extreme conditions and identification of unique modifying enzymes to define the Toll-like receptor 4 structure-activity relationship. Biochim Biophys Acta Mol Cell Biol Lipids. 1862;1439-1450:2017.
  • Perez-Regidor L, Zarioh M, Ortega L, et al. Virtual screening approaches towards the discovery of toll-like receptor modulators. Int J Mol Sci. 2016;17:1508.
  • Murgueitio MS, Rakers C, Frank A, et al. Balancing inflammation: computational design of small-molecule toll-like receptor modulators. Trends Pharmacol Sci. 2017;38:155–168.
  • Okamura Y, Watari M, Jerud ES, et al. The extra domain A of fibronectin activates Toll-like receptor 4. J Biol Chem. 2001;276(13):10229–10233. .
  • Walton KA, Hsieh X, Gharavi N, et al. Receptors involved in the oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine-mediated synthesis of interleukin-8. A role for Toll-like receptor 4 and a glycosylphosphatidylinositol-anchored protein. J Biol Chem. 2003;278:29661–29666.
  • Wang Y, Qian Y, Fang Q, et al. Saturated palmitic acid induces myocardial inflammatory injuries through direct binding to TLR4 accessory protein MD2. Nat Commun. 2017;8:13997.
  • Tsung A, Sahai R, Tanaka H. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med. 2005;201(7):1135–1143.
  • Yang D, Postnikov YV, Li Y, et al. High-mobility group nucleosome-binding protein 1 acts as an alarmin and is critical for lipopolysaccharide-induced immune responses. J Exp Med. 2012;209(2005):157–171.
  • Cao L, Tanga FY, Deleo JA. The contributing role of CD14 in toll-like receptor 4 dependent neuropathic pain. Neuroscience. 2009;158:896–903.
  • Erridge C. The roles of Toll-like receptors in atherosclerosis. J Innate Immun. 2009;1:340–349.
  • Shi J, Zhao Y, Wang Y, et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature. 2014;514:187–192.
  • Chu LH, Indramohan M, Ratsimandresy RA. The oxidized phospholipid oxPAPC protects from septic shock by targeting the non-canonical inflammasome in macrophages. Nat Commun. 2018;9:996.
  • Pfalzgraff A, Weindl G. Intracellular lipopolysaccharide sensing as a potential therapeutic target for sepsis. Trends Pharmacol Sci. 2019;40:187–197.
  • Opal SM, Laterre PF, Francois B, et al. Effect of Eritoran, an antagonist of MD2-TLR4, on mortality in patients with severe sepsis: the ACCESS randomized trial. JAMA. 2013;309:1154–1162.
  • Pfalzgraff A, Heinbockel L, Su Q, et al. Synthetic anti-endotoxin peptides inhibit cytoplasmic LPS-mediated responses. Biochem Pharmacol. 2017;140:64–72.
  • Vanaja SK, Russo AJ, Behl B, et al. Bacterial outer membrane vesicles mediate cytosolic localization of LPS and caspase-11 activation. Cell. 2016;165:1106–1119.
  • Bateman RM, Sharpe MD, Jagger JE, et al. Erratum to: 36th international symposium on intensive care and emergency medicine: Brussels, Belgium. 15-18 March 2016. Crit Care. 2016;20:347.
  • Park KS, Choi KH, Kim YS. Outer membrane vesicles derived from Escherichia coli induce systemic inflammatory response syndrome. PLoS One. 2010;5:e11334.
  • Pfalzgraff A, Correa W, Heinbockel L, et al. LPS-neutralizing peptides reduce outer membrane vesicle-induced inflammatory responses. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864:1503–1513.
  • Pfalzgraff A, Heinbockel L, Su Q, et al. Synthetic antimicrobial and LPS-neutralising peptides suppress inflammatory and immune responses in skin cells and promote keratinocyte migration. Sci Rep. 2016;6:31577.
  • Pfalzgraff A, Barcena-Varela S, Heinbockel L, et al. Antimicrobial endotoxin-neutralizing peptides promote keratinocyte migration via P2X7 receptor activation and accelerate wound healing in vivo. Br J Pharmacol. 2018;175:3581–3593.
  • Nunnally ME, Patel A. Sepsis - What’s new in 2019? Curr Opin Anaesthesiol. 2019;32:163–168.
  • Cavaillon JM, Chretien F. From septicemia to sepsis 3.0-from ignaz semmelweis to Louis Pasteur. Genes Immun. 2019;20:371–382.
  • Singer M, Deutschman CS, Seymour CW, et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 2016;315:801–810.
  • Luhr R, Cao Y, Soderquist B, et al. Trends in sepsis mortality over time in randomised sepsis trials: a systematic literature review and meta-analysis of mortality in the control arm, 2002-2016. Crit Care. 2019;23:241.
  • Vincent JL, Sakr Y. Clinical trial design for unmet clinical needs: a spotlight on sepsis. Expert Rev Clin Pharmacol. 2019;12:893–900.
  • Manocha S, Feinstein D, Kumar A, et al. Novel therapies for sepsis: antiendotoxin therapies. Expert Opin Investig Drugs. 2002;11:1795–1812.
  • Giamarellos-Bourboulis EJ, Opal SM. The role of genetics and antibodies in sepsis. Ann Transl Med. 2016;4:328.
  • Greisman SE, Johnston CA. Failure of antisera to J5 and R595 rough mutants to reduce endotoxemic lethality. J Infect Dis. 1988;157:54–64.
  • Shankar-Hari M, Spencer J, Sewell WA, et al. Bench-to-bedside review: immunoglobulin therapy for sepsis - biological plausibility from a critical care perspective. Crit Care. 2012;16:206.
  • Greisman HA, Pabo CO. A general strategy for selecting high-affinity zinc finger proteins for diverse DNA target sites. Science. 1997;275:657–661.
  • Welte T, Dellinger RP, Ebelt H, et al. Efficacy and safety of trimodulin, a novel polyclonal antibody preparation, in patients with severe community-acquired pneumonia: a randomized, placebo-controlled, double-blind, multicenter, phase II trial (CIGMA study). Intensive Care Med. 2018;44:438–448.
  • Trautmann M, Held TK, Susa M, et al. Bacterial lipopolysaccharide (LPS)-specific antibodies in commercial human immunoglobulin preparations: superior antibody content of an IgM-enriched product. Clin Exp Immunol. 1998;111:81–90.
  • Schwab I, Nimmerjahn F. Intravenous immunoglobulin therapy: how does IgG modulate the immune system? Nat Rev Immunol. 2013;13:176–189.
  • Almansa R, Tamayo E, Andaluz-Ojeda D, et al. The original sins of clinical trials with intravenous immunoglobulins in sepsis. Crit Care. 2015;19:90.
  • Welte T, Dellinger RP, Ebelt H, et al. Concept for a study design in patients with severe community-acquired pneumonia: A randomised controlled trial with a novel IGM-enriched immunoglobulin preparation - The CIGMA study. Respir Med. 2015;109:758–767.
  • Quintiliani R, Nightingale CH. Antimicrobials and therapeutic decision making: an historical perspective. Pharmacotherapy. 1991;11:6S–13S.
  • Olsen KM, Campbell GD Jr. E5 monoclonal immunoglobulin M antibody for the treatment of gram-negative sepsis. DICP. 1991;25:784–790.
  • Kobayashi H, Kawai S, Sakayori S, et al. [Phase I study of edobacomab (E5) in patients with gram-negative sepsis]. Kansenshogaku Zasshi. 1994;68:59–80.
  • Wortel CH, Ziegler EJ, van Deventer SJ. Therapy of gram-negative sepsis in man with anti-endotoxin antibodies: a review. Prog Clin Biol Res. 1991;367:161–178.
  • Gorelick KJ, Chmel H. The role of monoclonal antibodies in the management of gram-negative sepsis. Experience with the E5 antibody. Infect Dis Clin North Am. 1991;5:899–913.
  • Bouter AS, van Kessel KP, Cornelissen JJ, et al. Antibiotics enhance binding of lipid A-specific murine monoclonal antibody E5 to Gram-negative bacteria. Int J Antimicrob Agents. 1994;4:191–195.
  • Marra MN, Thornton MB, Snable JL, et al. Endotoxin-binding and -neutralizing properties of recombinant bactericidal/permeability-increasing protein and monoclonal antibodies HA-1A and E5. Crit Care Med. 1994;22:559–565.
  • Manzullo EF. Sepsis: the role of steroids and monoclonal antibodies in treatment. Oncol (Williston Park). 1994;8:115–20; discussion 120, 123.
  • Angus DC, Birmingham MC, Balk RA, et al. E5 murine monoclonal antiendotoxin antibody in gram-negative sepsis: a randomized controlled trial. E5 Study Investigators. JAMA. 2000;283:1723–1730.
  • Fomsgaard A. [Septomonab (Centoxin) in the treatment of gram-negative septicemia]. Ugeskr Laeger. 1993;155:1066–1071.
  • Fang KC. Monoclonal antibodies to endotoxin in the management of sepsis. West J Med. 1993;158:393–399.
  • Murphy ST, Bellamy MC. The quest for the magic bullet: Centoxin, drotrecogin alfa and lessons not learned. Trends Anaesth Crit Care. 2013;3:316–319.
  • Teng NNH, Kaplan HS, Hebert JM, et al. Protection against Gram-negative bacteremia and endotoxemia with human monoclonal IgM antibodies. Proc Natl Acad Sci USA. 1985;82(6):1790–1794.
  • Ziegler EJ, Fisher CJ Jr., Sprung CL, et al. Treatment of gram-negative bacteremia and septic shock with HA-1A human monoclonal antibody against endotoxin. A randomized, double-blind, placebo-controlled trial. The HA-1A sepsis study group. N Engl J Med. 1991;324:429–436.
  • Derkx B, Wittes J, McCloskey R. Randomized, placebo-controlled trial of HA-1A, a human monoclonal antibody to endotoxin, in children with meningococcal septic shock. European pediatric meningococcal septic shock trial study group. Clin Infect Dis. 1999;28:770–777.
  • Reinhart K, Gluck T, Ligtenberg J, et al. CD14 receptor occupancy in severe sepsis: results of a phase I clinical trial with a recombinant chimeric CD14 monoclonal antibody (IC14). Crit Care Med. 2004;32:1100–1108.
  • Marks L. The birth pangs of monoclonal antibody therapeutics: the failure and legacy of Centoxin. MAbs. 2012;4:403–412.
  • Axtelle T, Pribble J. An overview of clinical studies in healthy subjects and patients with severe sepsis with IC14, a CD14-specific chimeric monoclonal antibody. J Endotoxin Res. 2003;9:385–389.
  • Werdan K. Immunoglobulin treatment in sepsis–is the answer “no”? Crit Care Med. 2006;34:1542–1544.
  • Kreymann KG, de Heer G, Nierhaus A, et al. Use of polyclonal immunoglobulins as adjunctive therapy for sepsis or septic shock. Crit Care Med. 2007;35:2677–2685.
  • Alejandria MM, Lansang MA, Dans LF, et al. Intravenous immunoglobulin for treating sepsis, severe sepsis and septicshock. Cochrane Database Syst Rev. 2013;CD001090.(90 pages).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.