8,535
Views
260
CrossRef citations to date
0
Altmetric
Review

Animal models of sepsis

Pages 143-153 | Received 25 Jun 2013, Accepted 07 Aug 2013, Published online: 19 Aug 2013

Abstract

Sepsis remains a common, serious, and heterogeneous clinical entity that is difficult to define adequately. Despite its importance as a public health problem, efforts to develop and gain regulatory approval for a specific therapeutic agent for the adjuvant treatment of sepsis have been remarkably unsuccessful. One step in the critical pathway for the development of a new agent for adjuvant treatment of sepsis is evaluation in an appropriate animal model of the human condition. Unfortunately, the animal models that have been used for this purpose have often yielded misleading findings. It is likely that there are multiple reasons for the discrepancies between the results obtained in tests of pharmacological agents in animal models of sepsis and the outcomes of human clinical trials. One of important reason may be that the changes in gene expression, which are triggered by trauma or infection, are different in mice, a commonly used species for preclinical testing, and humans. Additionally, many species, including mice and baboons, are remarkably resistant to the toxic effects of bacterial lipopolysaccharide, whereas humans are exquisitely sensitive. New approaches toward the use of animals for sepsis research are being investigated. But, at present, results from preclinical studies of new therapeutic agents for sepsis must be viewed with a degree of skepticism.

In a review article about animal models of sepsis published more than two decades ago, Stephen Heard and I wrote this sentence: “Despite the considerable progress in this field, sepsis is still an important cause of mortality.”Citation1 We also wrote this sentence: “Sepsis, as a clinical entity, is very heterogeneous and clinical data are invariably confounded by the effects of age, coexisting diseases, and supportive therapy.” Obviously, these words are as true today as they were in 1990. Sepsis remains a common, serious, and heterogeneous clinical entity, which is difficult to define adequately. Despite its importance as a public health problem, efforts to develop and gain regulatory approval for a specific therapeutic agent for the adjuvant treatment sepsis have been remarkably unsuccessful.

In 1982, Ziegler et al. reported results from a clinical trial in patients with septic shock of a polyclonal antibody directed against lipopolysaccharide (LPS) from a mutant strain of Escherichia coli called J5.Citation2 Since then, approximately 60 phase 2 (P2) or phase 3 (P3) randomized controlled clinical trials of pharmacological agents for the adjuvant treatment of sepsis have been conducted (). From inspection of , it quickly becomes apparent that survival has been favorably affected, at least in a statistically significant way, in only a very few of these trials. The previously mentioned study of an anti-J5 antiserum by Ziegler and colleagues yielded positive results, but not all patients enrolled in the study were included in the evaluation set and not all deaths were counted in the calculation of mortality rates.Citation2 The first P3 study of HA-1A, a human monoclonal antibody designed to neutralize the deleterious effects of LPS, showed improved survival for patients treated with the experimental agent.Citation2 The design and conduct of this study, however, were criticized by experts in the field,Citation3 and when HA-1A was tested in a canine peritonitis model, excess mortality was observed in the animals treated with the monoclonal antibody.Citation4 Accordingly, a follow-up multicentric randomized controlled clinical trial was performed, and HA-1A was not shown to provide any therapeutic benefit.Citation5 A statistically significant, dose-dependent reduction in 28-d mortality was observed in a preliminary, open label P2 trial of anakinra (recombinant human interleukin-1 receptor antagonist).Citation6 Two subsequent P3 studies, however, failed to validate the encouraging findings from the initial P2 trial.Citation7,Citation8 When results were adjusted using a logistic regression model that included the effects of pre-determined potential confounding variables, treatment with afelimomab, a F(ab')2 fragment of a murine anti-tumor necrosis factor monoclonal antibody, was shown to significantly improve survival in patients with sepsis and a high circulating concentration of interleukin (IL)-6.Citation9 A statistically significant improvement in survival was observed in a P2 dose-finding study of pafase (recombinant human platelet activating factor acetylhydrolase,Citation10 but a subsequent larger P3 trial showed no evidence of therapeutic benefit when patients were treated with the recombinant protein.Citation11 In a P2 trial, enteral administration of talactoferrin (recombinant human lactoferrin) improved survival of patients with severe sepsis and the treatment effect just missed being statistically significant (P = 0.052).Citation12 Unfortunately, when the same drug was evaluated in a much larger P3 study, enrollment of patients was stopped early by the data safety monitoring board (DSMB) because of excess mortality in the talactoferrin-treated arm.Citation13 The first P3 study of recombinant human activated protein C yielded unequivocally positive resultsCitation14 and led to regulatory approval of the agent in North America and Europe. Subsequent clinical trials of this agent, however, were uniformly disappointing,Citation15-Citation17 and the commercial product (Xigris®) was withdrawn from the market in 2012. A multicentric clinical trial of therapy with hydrocortisone plus a synthetic mineralocorticoid, which was conducted in 19 intensive care units in France, yielded marginally positive results,Citation18 but, because benefit was not confirmed in another, larger multicentric study of hydrocortisone,Citation19 the therapeutic value of adjunctive therapy with corticosteroids remains quite controversial.

Table 1. Summary of clinical trials of pharmacological interventions for the adjuvant treatment of sepsis, which have been reported since 1982

With the exception of the positive results mentioned in the previous paragraph, all other P3 trials of adjunctive pharmacological therapies for sepsis or septic shock performed since 1982 have failed to provide evidence for improved survival. Indeed, treatment with etanercept, a recombinant fusion protein that combines two extracellular binding domains of the human p75 TNF receptor (TNFR2) with the Fc portion of human IgG1, was shown to worsen outcome for septic patients.Citation20 Similarly, treatment with 546C88 (l-monomethyl arginine, an isoform unselective nitric oxide synthase inhibitor) significantly and dramatically worsened survival for patients with septic shock.Citation21

This dismal record likely has many causes. For example, neither of the two anti-LPS monoclonal antibodies, which have been tested in P3 clinical trials, has been shown to effectively neutralize the pro-inflammatory effects of LPS.Citation22,Citation23 In many trials, the duration of therapy with the experimental agent was arbitrarily determined rather than being adjusted based on clinical or biochemical data. Thus, the period of treatment in some cases might have been too short (or, for that matter, too long).

In a few cases, pharmacological agents were advanced into P2 or even P3 clinical trials in the absence of convincing evidence of efficacy from preliminary studies using animal models of sepsis or septic shock. However, in the vast majority of cases, preliminary studies, using various animal models of sepsis or septic shock, provided evidence for efficacy and supported the decision to carry out clinical trials. Some examples of compounds that failed in one or more P3 clinical trials but were effective in one or more animal models are listed in .

Table 2. Examples of some pharmacological agents, which have been evaluated in an animal model of sepsis and yielded negative results in one or more human clinical trials

The lack of a truly clinically relevant and predictive animal model appears to be one of the key barriers hampering the development of an effective therapeutic for the adjuvant treatment of sepsis. Over the past couple of decades, literally hundreds of different animal models of sepsis have been used by scientists. In some cases, studies using animal models of sepsis have been used as part of the development pathway for novel therapeutic agents. However, more often animal models have been used as a proxy for the human condition in order to gain insights into pathophysiology. Numerous previous reviews have cataloged in considerable detail the myriad approaches, which have been employed in an effort to recapitulate key features of severe sepsis or septic shock in human beings.Citation1,Citation24-Citation31 Accordingly, no effort will be made here to repeat this exercise. Rather, the discussion will focus on a concise critique of the most widely employed animal models of sepsis, focusing on why use of these models have failed to lead to the development of one or more useful pharmacological therapies for the syndrome in humans.

The most popular preclinical sepsis models use mice. Being very small mammals, mice pose little or no danger to laboratory personnel. Inbred strains are widely available from suppliers, and, generally speaking, are relatively inexpensive to acquire and maintain. Moreover, using genetically modified strains of mice is an elegant way to explore the importance of particular gene products in the pathogenesis of sepsis (or the response of septic animals to a pharmacological intervention). While laboratory experiments using any species of animal, especially ones that use organ dysfunction or mortality as a read-out, provoke intense scrutiny from regulatory and oversight groups, mice are not regarded as “companion animals” and, therefore, studies using mice may be regarded as being more ethical than are studies that enroll cats, dogs, horses, or non-human primates as research subjects. Finally, numerous immunological reagents and/or assay kits designed for murine systems are commercially available. These reagents and kits facilitate the measurement of cytokines or other mediators in biological fluids.

Injecting mice with purified LPS (“endotoxin”) via either the intraperitoneal (i.p.) route or the intravenous (i.v.) route leads to systemic activation of the innate immune system. If the dose of LPS is large enough, then the animals manifest physiological and biochemical changes that are reminiscent of certain very fulminant forms of gram-negative bacterial infection in humans, most notably overwhelming meningococcemia. Some of the manifestations of acute endotoxemia in mice include systemic arterial hypotension (i.e., shock), lactic acidosis, impaired myocardial contractility, a short-lived monophasic spike in the circulating level of TNF, a more prolonged elevation in the circulating level of IL-6, and a delayed increase in the circulating level of high mobility group box (HMGB)-1. Shock, lactic acidosis, impaired myocardial contractility, and increased circulating levels of TNF, IL-6, and HMGB1 all are features of sepsis or septic shock in humans, although the temporal kinetics and the magnitude of these changes from normal physiology are often different from what is observed in acute murine endotoxemia.

Activation of systemic inflammation by LPS in mice is largely mediated via interaction of the bacterial product with Toll-like receptor (TLR) 4, which is expressed on the surface of both “professional” immune cells, such as monocytes and macrophages, as well as many other cell types, including alveolar epithelial cells and myocardial cells. The intracellular signaling pathways, which are triggered by the interaction of LPS with TLR4, have been extensively investigated and there are many detailed reviews in the literature.Citation32,Citation33 Certain inbred strains, such as C3H/HeJ miceCitation34 and TLR4-deficient (“knockout”) mice,Citation34 are hyporesponsive to LPS. It is important to note, however, that C3H/HeJ mice are hyporesponsive but are not entirely unresponsive, to the toxic effects of LPS. For example, the dose of LPS required to induce weight loss in C3H/HeJ mice is about 40-fold less than the dose that is required to produce a similar degree of weight loss in congenic LPS-sensitive C3H/HeN mice.Citation35 Similarly, there is about a 40-fold difference between the lethal dose of LPS in LPS-responsive (A/HeJ) as compared with LPS-resistant (C3H/HeN) strains of mice.Citation36

Compared with humans, mice, even “normally responsive” strains, are remarkably less sensitive to the toxic or lethal effects of LPS. Thus, the dose of LPS, which leads to death in approximately half of mice (i.e., the LD50 dose) is about 1–25 mg/kg.Citation37-Citation39 This dose is about 1 000 000 times greater than the typical dose of LPS (2–4 ng/kg), which has been used in studies with human volunteer to induce symptoms (e.g., fever or myalgia) and the release of proinflammtory cytokines, such as TNF, into the circulation.Citation40,Citation41 The LD50 dose of LPS in mice is about 1000-fold to 10 000-fold greater than the dose of LPS that is required to induce severe illness and hypotension in humans.Citation42 In other words, the roughly 40-fold difference in the dose of LPS that is required to produce toxicity or death in “normally responsive” vs. “hyporesponsive” strains of mice is orders of magnitude smaller than is the difference in LPS dose that is required to produce death or serious illness in “normally responsive” mice as compared with human beings. The biological mechanism(s) that are responsible for the enormous difference between mice and humans with respect to responsiveness to LPS remain to be fully elucidated, but recent evidence obtained by Warren and colleagues suggests that one or more factors, which are present in murine sera, but are absent (or present in much lower concentrations) in human sera, are capable of suppressing the production of pro-inflammatory cytokines by murine (or human) mononuclear cells following stimulation with LPS or other TLR agonists, such as peptidoglycan (major component of the cell wall of gram-positive bacteria), zymosan (a glucan present in the cell wall of yeast) or bacterial DNA.Citation43 One of these factors may be the iron-binding acute phase protein, hemopexin.Citation44 Other factors, which have yet to be identified, may be important as well. In any event, the marked discrepancy in LPS sensitivity between mice and people suggests that data obtained using murine models of sepsis may be inapplicable to the human illness.

This view is strengthened by recently published findings from a systematic analysis of the genomic responses in three human conditions, which are associated with activation of the innate immune system, namely burn injury, trauma, and acute endotoxemia, and the corresponding insults in murine models.Citation45 In this important study, total cellular RNA was extracted from leukocytes isolated from blood, and gene expression profiles were obtained using appropriate Affymetrix GeneChip arrays. The samples from trauma and burn patients were obtained at 7 Level I trauma centers or 4 burn centers in the United States in the course of The Inflammation and Host Response to Injury, Large Scale Collaborative Research Program (“Glue Grant”), which was funded by the National Institutes of Health. The human endotoxemia samples were obtained from eight healthy volunteers, who were challenged with a small i.v. dose of LPS (2 ng/kg). Of note, there was high degree of similarity in the gene expression profiles in leukocytes from human subjects with burn injuries, major trauma, or acute endotoxemia. In marked contrast, there was minimal correlation of expression changes between the human conditions and their murine orthologs in the mouse models. For example, when the gene expression profile for human endotoxemia was compared with the gene expression profile for murine endotoxema, the Pearson correlation analysis yielded an R2 value of 0.01 (i.e., essential no correlation at all). Interestingly, the time to recovery (i.e., the time required to restore normal gene expression levels) was far longer in humans (months) as compared with mice (hours to days).

Of course, some concerns have been raised, regarding the findings from the Glue Grant program, which were summarized in the previous paragraph.Citation46 For example, the gene expression responses of humans were compared with those of only one inbred strain of mice, and it is well known that strain differences in the murine system can markedly affect immunological responses. Furthermore, some of the differences between “mice and men” might reflect the impact of therapeutic interventions rather than just differences between the species with regard to gene expression responses to acute insults, such as trauma or infection. Still, the results obtained by Seok et al.Citation45 should prompt all researchers to be appropriately skeptical about extrapolating findings from murine sepsis models to the problem of sepsis in patients.

All acute endotoxemia models, irrespective of whether the animals under study are mice or some other species, suffer from the same problem. In these models, activation of the innate immune system can only have deleterious effects; therefore, any intervention that blunts the inflammatory response is likely to be beneficial. In contrast, sepsis in patients is triggered by an infectious process and the immunological responses to microbial challenge can have both salutary and deleterious effects.

Because sepsis in patients often starts with a focus of infection in the lungs or peritoneal cavity,Citation31 investigators and funding agencies have grown enamored in recent years with animal, especially murine, models of pneumonia and peritonitis. Because of its simplicity and, when performed correctly, its reproducibility, cecal ligation and puncture (CLP), which is a murine model of bacterial peritonitis, has been regarded, at least until the publication of the paper by Seok et al. cited above, as the “gold standard” animal model of sepsis. CLP in mice reproduces a number of key features of secondary bacterial peritonitis in humans, including polymicrobial infection,Citation47 persistently elevated circulating HMGB1 levels,Citation48 hyperdynamic circulatory system,Citation49 and development of acute lung injury.Citation50 Moreover, some studies, using the murine CLP model, yielded results, which are concordant with the lack of efficacy in human clinical trials of some important pharmacological approaches for the adjuvant treatment of sepsis, such as the administration of antibodies against TNFCitation51,Citation52 or the administration of IL-1RA.Citation53 In other cases, however, studies performed using the murine CLP model suggested that therapeutic interventions, such as administration of a PAF receptor antagonistCitation54 or exogenous PAF acetylhydrolase,Citation55 would improve survival. In clinical trials, however, these approaches were unsuccessful.

Although the murine CLP paradigm has some features to recommend it, this model of severe sepsis also has some serious flaws. First, as already discussed, at a gene expression level, acute systemic inflammatory responses in mice appear to be quite different from those that occur in humans. Second, sepsis in humans is, by and large, a disease that occurs at the extremes of age.Citation56 In contrast, in the vast majority of studies using the murine CLP model, sepsis is induced in young adult animals without any co-morbid conditions. It is noteworthy in this regard that CLP-induced sepsis in young adult mice is not associated with the development of acute kidney injury (AKI), a common problem in human sepsis, whereas CLP-induced sepsis in aged animals does lead to development of AKI.Citation57 Third, septic patients typically receive multiple forms of supportive care, including: mechanical ventilation, if indicated; resuscitation of intravascular volume; infusion of vasopressors and/or inotropic agents; administration of anti-microbial agents; renal replacement therapy, if indicated; surgical source control, if indicated; and enteral (or, less commonly, parenteral) administration of nutritional supplements. Some of these forms of supportive of care, such as the administration of antibiotics, are sometimes included in murine studies. However, the more complex interventions, such as renal replacement therapy or prolonged mechanical ventilation, are rarely, if ever, employed. Fourth, the temporal course of the septic process—from the onset of symptoms to death—in the murine CLP model is compressed into an interval that lasts at most a few days. In contrast, patients with lethal sepsis often survive for weeks before succumbing to their illness.

The CLP model likely can be improved, but the fixes are expensive and certainly not guaranteed to improve the model enough to make it a reliable step in the pathway toward the development of effective pharmacological agents for the adjuvant treatment of human sepsis. One improvement, already noted above, is to use aged mice, as described by the Star laboratory at the National Institutes of Health.Citation27,Citation57 Another improvement is to use “humanized mice” as described by Unsinger and colleagues.Citation58 These mice are generated by transplanting human CD34+ cord blood hematopoetic stem cells into gamma-irradiated neonatal NOD-scidIL2rγnull mice. These mice develop a complete lineage of human cells of the innate and adaptive immune systems, including monocytes, macrophages, plasmacytoid and myeloid dendritic cells, NK cells, T cells, and B cells. Sepsis is induced by performing CLP. Although the model might be useful, it is important to note that many key cell types, such as endothelial cells and intestinal and respiratory tract epithelial cells, are still murine. And, these cells, although not components of the “professional” immune system, are nonetheless important in the pathogenesis of the septic phenotype. Furthermore, generating “humanized” mice is a complex, time-consuming, and very expensive process, which is unlikely to be widely adopted. Finally, as yet, it is unknown whether results obtained with this model can predict the outcome of a clinical trial of a novel pharmacological intervention in patients with severe sepsis or septic shock.

Another way to solve some of the problems of the murine CLP model is to use a larger animal species, especially one that is more like humans with regard to sensitivity to LPS and possibly other pathogen-associated molecular pattern (PAMP) molecules. Following this sort of reasoning, our laboratory developed a rabbit peritonitis model, wherein sepsis was induced by i.p. injection of a known quantity of viable bacteria mixed with substances, namely hemoglobin and mucin, which are known to enhance the lethality of infections.Citation59 We employed a highly pathogenic strain of bacteria (E. coli O18:K1). The animals were treated with an appropriate antibiotic and were adequately resuscitated via an indwelling i.v. catheter. Studies performed with the rabbit paradigm indicated that tifacogin is an effective therapeutic agent, but contrary results were obtained in clinical trials.Citation60-Citation62 Thus, even this rabbit paradigm, which was developed to address many of the problems with earlier sepsis models, failed to predict the results of human clinical trials.

Sheep are docile large animals. Like humans, they are extremely sensitive to LPS. For example, continuous infusion of LPS at a rate as low as 9 ng/kg per h leads to marked changes in pulmonary arterial pressure, cardiac output, and lung microvascular permeability.Citation63 True sepsis, rather than endotoxemia, can be induced in sheep in a variety of ways, such as by infusing viable Pseudomonas aeruginosa i.v.Citation64 For ethical reasons, sheep are rarely used for survival studies, and thus it is not known whether ovine models of sepsis should be incorporated into the critical pathway for the development of new pharmacological agents for the treatment of sepsis.

In view of their close phylogenetic proximity to humans, nonhuman primate species, such as Papio anubis, would seem be good choices for preclinical models of sepsis. But, baboons and other monkey species tend to be remarkably resistant to the lethal effects of intravenously administered LPS or viable bacteria. Thus, in the classic Hinshaw model of lethal sepsis in baboons, the animals are infused with more than 1010 colony forming units (cfu)/kg of viable E. coli bacteria.Citation65 Blood cultures in this model contain 103 to 107 cfu/ml, i.e., levels of bacteria that are much greater than are commonly observed in lethal human septic shock. In numerous studies (), findings obtained using this model to test pharmacological interventions have failed to predict the outcome of human clinical trials.

In summary, most animal models of human sepsis are flawed. Studies using very complex models, such as ones that employ “humanized” mice, may represent a major advance, but, at present, data obtained in this way are quite limited. Experiments using animal models will remain in the critical pathway for the development of new agents for the pharmacological treatment of severe sepsis or septic shock. But, results from these preclinical studies never should be extrapolated directly to the problem of human sepsis.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

10.4161/viru.26083

References

  • Fink MP, Heard SO. Laboratory models of sepsis and septic shock. J Surg Res 1990; 49:186 - 96; http://dx.doi.org/10.1016/0022-4804(90)90260-9; PMID: 2199735
  • Ziegler EJ, McCutchan JA, Fierer J, Glauser MP, Sadoff JC, Douglas H, Braude AI. Treatment of gram-negative bacteremia and shock with human antiserum to a mutant Escherichia coli. N Engl J Med 1982; 307:1225 - 30; http://dx.doi.org/10.1056/NEJM198211113072001; PMID: 6752708
  • Warren HS, Danner RL, Munford RS. Anti-endotoxin monoclonal antibodies. N Engl J Med 1992; 326:1153 - 7; http://dx.doi.org/10.1056/NEJM199204233261711; PMID: 1552919
  • Quezado ZM, Natanson C, Alling DW, Banks SM, Koev CA, Elin RJ, Hosseini JM, Bacher JD, Danner RL, Hoffman WD. A controlled trial of HA-1A in a canine model of gram-negative septic shock. JAMA 1993; 269:2221 - 7; http://dx.doi.org/10.1001/jama.1993.03500170051033; PMID: 8474201
  • McCloskey RV, Straube RC, Sanders C, Smith SM, Smith CR, CHESS Trial Study Group. Treatment of septic shock with human monoclonal antibody HA-1A. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 1994; 121:1 - 5; http://dx.doi.org/10.7326/0003-4819-121-1-199407010-00001; PMID: 8198341
  • Fisher CJ Jr., Slotman GJ, Opal SM, Pribble JP, Bone RC, Emmanuel G, Ng D, Bloedow DC, Catalano MA, IL-1RA Sepsis Syndrome Study Group. Initial evaluation of human recombinant interleukin-1 receptor antagonist in the treatment of sepsis syndrome: a randomized, open-label, placebo-controlled multicenter trial. Crit Care Med 1994; 22:12 - 21; PMID: 8124953
  • Fisher CJ Jr., Dhainaut JF, Opal SM, Pribble JP, Balk RA, Slotman GJ, Iberti TJ, Rackow EC, Shapiro MJ, Greenman RL, et al. Recombinant human interleukin 1 receptor antagonist in the treatment of patients with sepsis syndrome. Results from a randomized, double-blind, placebo-controlled trial. Phase III rhIL-1ra Sepsis Syndrome Study Group. JAMA 1994; 271:1836 - 43; http://dx.doi.org/10.1001/jama.1994.03510470040032; PMID: 8196140
  • Opal SM, Fisher CJ Jr., Dhainaut JF, Vincent JL, Brase R, Lowry SF, Sadoff JC, Slotman GJ, Levy H, Balk RA, et al. Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: a phase III, randomized, double-blind, placebo-controlled, multicenter trial. The Interleukin-1 Receptor Antagonist Sepsis Investigator Group. Crit Care Med 1997; 25:1115 - 24; http://dx.doi.org/10.1097/00003246-199707000-00010; PMID: 9233735
  • Panacek EA, Marshall JC, Albertson TE, Johnson DH, Johnson S, MacArthur RD, Miller M, Barchuk WT, Fischkoff S, Kaul M, et al, Monoclonal Anti-TNF: a Randomized Controlled Sepsis Study Investigators. Efficacy and safety of the monoclonal anti-tumor necrosis factor antibody F(ab’)2 fragment afelimomab in patients with severe sepsis and elevated interleukin-6 levels. Crit Care Med 2004; 32:2173 - 82; PMID: 15640628
  • Schuster DP, Metzler M, Opal S, Lowry S, Balk R, Abraham E, Levy H, Slotman G, Coyne E, Souza S, et al, Pafase ARDS Prevention Study Group. Recombinant platelet-activating factor acetylhydrolase to prevent acute respiratory distress syndrome and mortality in severe sepsis: Phase IIb, multicenter, randomized, placebo-controlled, clinical trial. Crit Care Med 2003; 31:1612 - 9; http://dx.doi.org/10.1097/01.CCM.0000063267.79824.DB; PMID: 12794395
  • Opal S, Laterre PF, Abraham E, Francois B, Wittebole X, Lowry S, Dhainaut JF, Warren B, Dugernier T, Lopez A, et al, Controlled Mortality Trial of Platelet-Activating Factor Acetylhydrolase in Severe Sepsis Investigators. Recombinant human platelet-activating factor acetylhydrolase for treatment of severe sepsis: results of a phase III, multicenter, randomized, double-blind, placebo-controlled, clinical trial. Crit Care Med 2004; 32:332 - 41; http://dx.doi.org/10.1097/01.CCM.0000108867.87890.6D; PMID: 14758145
  • Guntupalli K, Dean N, Morris PE, Bandi V, Margolis B, Rivers E, Levy M, Lodato RF, Ismail PM, Reese A, et al, TLF LF-0801 Investigator Group. A phase 2 randomized, double-blind, placebo-controlled study of the safety and efficacy of talactoferrin in patients with severe sepsis. Crit Care Med 2013; 41:706 - 16; http://dx.doi.org/10.1097/CCM.0b013e3182741551; PMID: 23425819
  • Agennix AG. Agennix AG halts phase II/III oasis trial in severe sepsis [Internet] 2012 [cited 2012 Feb 2] Available from:http://agennix.com/index.php?option=com_content&view=category&id=23&Itemid=102&lang=en
  • Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, Lopez-Rodriguez A, Steingrub JS, Garber GE, Helterbrand JD, Ely EW, et al, Recombinant human protein C Worldwide Evaluation in Severe Sepsis (PROWESS) study group. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med 2001; 344:699 - 709; http://dx.doi.org/10.1056/NEJM200103083441001; PMID: 11236773
  • Abraham E, Laterre PF, Garg R, Levy H, Talwar D, Trzaskoma BL, François B, Guy JS, Brückmann M, Rea-Neto A, et al, Administration of Drotrecogin Alfa (Activated) in Early Stage Severe Sepsis (ADDRESS) Study Group. Drotrecogin alfa (activated) for adults with severe sepsis and a low risk of death. N Engl J Med 2005; 353:1332 - 41; http://dx.doi.org/10.1056/NEJMoa050935; PMID: 16192478
  • Nadel S, Goldstein B, Williams MD, Dalton H, Peters M, Macias WL, Abd-Allah SA, Levy H, Angle R, Wang D, et al, REsearching severe Sepsis and Organ dysfunction in children: a gLobal perspective (RESOLVE) study group. Drotrecogin alfa (activated) in children with severe sepsis: a multicentre phase III randomised controlled trial. Lancet 2007; 369:836 - 43; http://dx.doi.org/10.1016/S0140-6736(07)60411-5; PMID: 17350452
  • Ranieri VM, Thompson BT, Barie PS, Dhainaut JF, Douglas IS, Finfer S, Gårdlund B, Marshall JC, Rhodes A, Artigas A, et al, PROWESS-SHOCK Study Group. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med 2012; 366:2055 - 64; http://dx.doi.org/10.1056/NEJMoa1202290; PMID: 22616830
  • Annane D, Sébille V, Charpentier C, Bollaert PE, François B, Korach JM, Capellier G, Cohen Y, Azoulay E, Troché G, et al. Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA 2002; 288:862 - 71; http://dx.doi.org/10.1001/jama.288.7.862; PMID: 12186604
  • Sprung CL, Annane D, Keh D, Moreno R, Singer M, Freivogel K, Weiss YG, Benbenishty J, Kalenka A, Forst H, et al, CORTICUS Study Group. Hydrocortisone therapy for patients with septic shock. N Engl J Med 2008; 358:111 - 24; http://dx.doi.org/10.1056/NEJMoa071366; PMID: 18184957
  • Fisher CJ Jr., Agosti JM, Opal SM, Lowry SF, Balk RA, Sadoff JC, Abraham E, Schein RM, Benjamin E, The Soluble TNF Receptor Sepsis Study Group. Treatment of septic shock with the tumor necrosis factor receptor:Fc fusion protein. N Engl J Med 1996; 334:1697 - 702; http://dx.doi.org/10.1056/NEJM199606273342603; PMID: 8637514
  • López A, Lorente JA, Steingrub J, Bakker J, McLuckie A, Willatts S, Brockway M, Anzueto A, Holzapfel L, Breen D, et al. Multiple-center, randomized, placebo-controlled, double-blind study of the nitric oxide synthase inhibitor 546C88: effect on survival in patients with septic shock. Crit Care Med 2004; 32:21 - 30; http://dx.doi.org/10.1097/01.CCM.0000105581.01815.C6; PMID: 14707556
  • Marra MN, Thornton MB, Snable JL, Wilde CG, Scott RW. Endotoxin-binding and -neutralizing properties of recombinant bactericidal/permeability-increasing protein and monoclonal antibodies HA-1A and E5. Crit Care Med 1994; 22:559 - 65; http://dx.doi.org/10.1097/00003246-199404000-00009; PMID: 8143464
  • Chen TY, Warren HS, Greene E, Black KM, Frostell CG, Robinson DR, Zapol WM. Protective effects of anti-O polysaccharide and anti-lipid A monoclonal antibodies on pulmonary hemodynamics. J Appl Physiol 1993; 74:423 - 7; PMID: 8444723
  • Fink MP. Animal models of sepsis and its complications. Kidney Int 2008; 74:991 - 3; http://dx.doi.org/10.1038/ki.2008.442; PMID: 18827799
  • Deitch EA. Rodent models of intra-abdominal infection. Shock 2005; 24:Suppl 1 19 - 23; http://dx.doi.org/10.1097/01.shk.0000191386.18818.0a; PMID: 16374368
  • Deitch EA. Animal models of sepsis and shock: a review and lessons learned. Shock 1998; 9:1 - 11; http://dx.doi.org/10.1097/00024382-199801000-00001; PMID: 9466467
  • Doi K, Leelahavanichkul A, Yuen PS, Star RA. Animal models of sepsis and sepsis-induced kidney injury. J Clin Invest 2009; 119:2868 - 78; http://dx.doi.org/10.1172/JCI39421; PMID: 19805915
  • Dyson A, Singer M. Animal models of sepsis: why does preclinical efficacy fail to translate to the clinical setting?. Crit Care Med 2009; 37:Suppl S30 - 7; http://dx.doi.org/10.1097/CCM.0b013e3181922bd3; PMID: 19104223
  • Zanotti-Cavazzoni SL, Goldfarb RD. Animal models of sepsis. [vii-viii.] Crit Care Clin 2009; 25:703 - 19, vii-viii; http://dx.doi.org/10.1016/j.ccc.2009.08.005; PMID: 19892248
  • Freise H, Brückner UB, Spiegel HU. Animal models of sepsis. J Invest Surg 2001; 14:195 - 212; http://dx.doi.org/10.1080/089419301750420232; PMID: 11680530
  • van der Poll T. Preclinical sepsis models. Surg Infect (Larchmt) 2012; 13:287 - 92; http://dx.doi.org/10.1089/sur.2012.105; PMID: 23046077
  • Rossol M, Heine H, Meusch U, Quandt D, Klein C, Sweet MJ, Hauschildt S. LPS-induced cytokine production in human monocytes and macrophages. Crit Rev Immunol 2011; 31:379 - 446; http://dx.doi.org/10.1615/CritRevImmunol.v31.i5.20; PMID: 22142165
  • Yamamoto M, Akira S. Lipid A receptor TLR4-mediated signaling pathways. Adv Exp Med Biol 2010; 667:59 - 68; http://dx.doi.org/10.1007/978-1-4419-1603-7_6; PMID: 20665200
  • Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y, Takeda K, Akira S. Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J Immunol 1999; 162:3749 - 52; PMID: 10201887
  • Vogel SN, Moore RN, Sipe JD, Rosenstreich DL. BCG-induced enhancement of endotoxin sensitivity in C3H/HeJ mice. I. In vivo studies. J Immunol 1980; 124:2004 - 9; PMID: 6154089
  • Sultzer BM. Genetic factors in leucocyte responses to endotoxin: further studies in mice. J Immunol 1969; 103:32 - 8; PMID: 4894397
  • Glode LM, Mergenhagen SE, Rosenstreich DL. Significant contribution of spleen cells in mediating the lethal effects of endotoxin in vivo. Infect Immun 1976; 14:626 - 30; PMID: 965087
  • McCuskey RS, McCuskey PA, Urbaschek R, Urbaschek B. Species differences in Kupffer cells and endotoxin sensitivity. Infect Immun 1984; 45:278 - 80; PMID: 6376358
  • Reynolds K, Novosad B, Hoffhines A, Gipson J, Johnson J, Peters J, Gonzalez F, Gimble J, Hill M. Pretreatment with troglitazone decreases lethality during endotoxemia in mice. J Endotoxin Res 2002; 8:307 - 14; PMID: 12230920
  • Barber AE, Coyle SM, Fischer E, Smith C, van der Poll T, Shires GT, Lowry SF. Influence of hypercortisolemia on soluble tumor necrosis factor receptor II and interleukin-1 receptor antagonist responses to endotoxin in human beings. Surgery 1995; 118:406 - 10, discussion 410-1; http://dx.doi.org/10.1016/S0039-6060(05)80352-6; PMID: 7638758
  • Suffredini AF, Reda D, Banks SM, Tropea M, Agosti JM, Miller R. Effects of recombinant dimeric TNF receptor on human inflammatory responses following intravenous endotoxin administration. J Immunol 1995; 155:5038 - 45; PMID: 7594512
  • Taveira da Silva AM, Kaulbach HC, Chuidian FS, Lambert DR, Suffredini AF, Danner RL. Brief report: shock and multiple-organ dysfunction after self-administration of Salmonella endotoxin. N Engl J Med 1993; 328:1457 - 60; http://dx.doi.org/10.1056/NEJM199305203282005; PMID: 8479465
  • Warren HS, Fitting C, Hoff E, Adib-Conquy M, Beasley-Topliffe L, Tesini B, Liang X, Valentine C, Hellman J, Hayden D, et al. Resilience to bacterial infection: difference between species could be due to proteins in serum. J Infect Dis 2010; 201:223 - 32; http://dx.doi.org/10.1086/649557; PMID: 20001600
  • Liang X, Lin T, Sun G, Beasley-Topliffe L, Cavaillon JM, Warren HS. Hemopexin down-regulates LPS-induced proinflammatory cytokines from macrophages. J Leukoc Biol 2009; 86:229 - 35; http://dx.doi.org/10.1189/jlb.1208742; PMID: 19395472
  • Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DR, McDonald-Smith GP, Gao H, Hennessy L, et al, Inflammation and Host Response to Injury, Large Scale Collaborative Research Program. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A 2013; 110:3507 - 12; http://dx.doi.org/10.1073/pnas.1222878110; PMID: 23401516
  • Of men, not mice. Nat Med 2013; 19:379; http://dx.doi.org/10.1038/nm.3163; PMID: 23558605
  • Ozment TR, Ha T, Breuel KF, Ford TR, Ferguson DA, Kalbfleisch J, Schweitzer JB, Kelley JL, Li C, Williams DL. Scavenger receptor class a plays a central role in mediating mortality and the development of the pro-inflammatory phenotype in polymicrobial sepsis. PLoS Pathog 2012; 8:e1002967; http://dx.doi.org/10.1371/journal.ppat.1002967; PMID: 23071440
  • Ulloa L, Ochani M, Yang H, Tanovic M, Halperin D, Yang R, Czura CJ, Fink MP, Tracey KJ. Ethyl pyruvate prevents lethality in mice with established lethal sepsis and systemic inflammation. Proc Natl Acad Sci U S A 2002; 99:12351 - 6; http://dx.doi.org/10.1073/pnas.192222999; PMID: 12209006
  • Hollenberg SM, Dumasius A, Easington C, Colilla SA, Neumann A, Parrillo JE. Characterization of a hyperdynamic murine model of resuscitated sepsis using echocardiography. Am J Respir Crit Care Med 2001; 164:891 - 5; http://dx.doi.org/10.1164/ajrccm.164.5.2010073; PMID: 11549551
  • Doerschug KC, Powers LS, Monick MM, Thorne PS, Hunninghake GW. Antibiotics delay but do not prevent bacteremia and lung injury in murine sepsis. Crit Care Med 2004; 32:489 - 94; http://dx.doi.org/10.1097/01.CCM.0000109450.66450.23; PMID: 14758168
  • Eskandari MK, Bolgos G, Miller C, Nguyen DT, DeForge LE, Remick DG. Anti-tumor necrosis factor antibody therapy fails to prevent lethality after cecal ligation and puncture or endotoxemia. J Immunol 1992; 148:2724 - 30; PMID: 1315357
  • Kato T, Murata A, Ishida H, Toda H, Tanaka N, Hayashida H, Monden M, Matsuura N. Interleukin 10 reduces mortality from severe peritonitis in mice. Antimicrob Agents Chemother 1995; 39:1336 - 40; http://dx.doi.org/10.1128/AAC.39.6.1336; PMID: 7574526
  • Ashare A, Powers LS, Butler NS, Doerschug KC, Monick MM, Hunninghake GW. Anti-inflammatory response is associated with mortality and severity of infection in sepsis. Am J Physiol Lung Cell Mol Physiol 2005; 288:L633 - 40; http://dx.doi.org/10.1152/ajplung.00231.2004; PMID: 15579629
  • Rios-Santos F, Benjamim CF, Zavery D, Ferreira SH, Cunha FdeQ. A critical role of leukotriene B4 in neutrophil migration to infectious focus in cecal ligaton and puncture sepsis. Shock 2003; 19:61 - 5; http://dx.doi.org/10.1097/00024382-200301000-00012; PMID: 12558146
  • Gomes RN, Bozza FA, Amâncio RT, Japiassú AM, Vianna RC, Larangeira AP, Gouvêa JM, Bastos MS, Zimmerman GA, Stafforini DM, et al. Exogenous platelet-activating factor acetylhydrolase reduces mortality in mice with systemic inflammatory response syndrome and sepsis. Shock 2006; 26:41 - 9; http://dx.doi.org/10.1097/01.shk.0000209562.00070.1a; PMID: 16783197
  • Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 2001; 29:1303 - 10; http://dx.doi.org/10.1097/00003246-200107000-00002; PMID: 11445675
  • Miyaji T, Hu X, Yuen PS, Muramatsu Y, Iyer S, Hewitt SM, Star RA. Ethyl pyruvate decreases sepsis-induced acute renal failure and multiple organ damage in aged mice. Kidney Int 2003; 64:1620 - 31; http://dx.doi.org/10.1046/j.1523-1755.2003.00268.x; PMID: 14531793
  • Unsinger J, McDonough JS, Shultz LD, Ferguson TA, Hotchkiss RS. Sepsis-induced human lymphocyte apoptosis and cytokine production in “humanized” mice. J Leukoc Biol 2009; 86:219 - 27; http://dx.doi.org/10.1189/jlb.1008615; PMID: 19369639
  • Camerota AJ, Creasey AA, Patla V, Larkin VA, Fink MP. Delayed treatment with recombinant human tissue factor pathway inhibitor improves survival in rabbits with gram-negative peritonitis. J Infect Dis 1998; 177:668 - 76; http://dx.doi.org/10.1086/514246; PMID: 9498446
  • Abraham E, Reinhart K, Opal S, Demeyer I, Doig C, Rodriguez AL, Beale R, Svoboda P, Laterre PF, Simon S, et al, OPTIMIST Trial Study Group. Efficacy and safety of tifacogin (recombinant tissue factor pathway inhibitor) in severe sepsis: a randomized controlled trial. JAMA 2003; 290:238 - 47; http://dx.doi.org/10.1001/jama.290.2.238; PMID: 12851279
  • Abraham E, Reinhart K, Svoboda P, Seibert A, Olthoff D, Dal Nogare A, Postier R, Hempelmann G, Butler T, Martin E, et al. Assessment of the safety of recombinant tissue factor pathway inhibitor in patients with severe sepsis: a multicenter, randomized, placebo-controlled, single-blind, dose escalation study. Crit Care Med 2001; 29:2081 - 9; http://dx.doi.org/10.1097/00003246-200111000-00007; PMID: 11700399
  • Wunderink RG, Laterre PF, Francois B, Perrotin D, Artigas A, Vidal LO, Lobo SM, Juan JS, Hwang SC, Dugernier T, et al, CAPTIVATE Trial Group. Recombinant tissue factor pathway inhibitor in severe community-acquired pneumonia: a randomized trial. Am J Respir Crit Care Med 2011; 183:1561 - 8; http://dx.doi.org/10.1164/rccm.201007-1167OC; PMID: 21297074
  • Traber DL, Redl H, Schlag G, Herndon DN, Kimura R, Prien T, Traber LD. Cardiopulmonary responses to continuous administration of endotoxin. Am J Physiol 1988; 254:H833 - 9; PMID: 3364588
  • Booke M, Hinder F, McGuire R, Traber LD, Traber DL. Noradrenaline and nomega-monomethyl-L-arginine (L-NMMA): effects on haemodynamics and regional blood flow in healthy and septic sheep. Clin Sci (Lond) 2000; 98:193 - 200; http://dx.doi.org/10.1042/CS19990143; PMID: 10657275
  • Hinshaw LB, Beller-Todd BK, Archer LT, Benjamin B, Flournoy DJ, Passey R, Wilson MF. Effectiveness of steroid/antibiotic treatment in primates administered LD100 Escherichia coli. Ann Surg 1981; 194:51 - 6; http://dx.doi.org/10.1097/00000658-198107000-00009; PMID: 7018430
  • Opal SM, Palardy JE, Parejo NA, Creasey AA. The activity of tissue factor pathway inhibitor in experimental models of superantigen-induced shock and polymicrobial intra-abdominal sepsis. Crit Care Med 2001; 29:13 - 7; http://dx.doi.org/10.1097/00003246-200101000-00003; PMID: 11176151
  • Ziegler EJ, Fisher CJ Jr., Sprung CL, Straube RC, Sadoff JC, Foulke GE, Wortel CH, Fink MP, Dellinger RP, Teng NN, 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 - 36; http://dx.doi.org/10.1056/NEJM199102143240701; PMID: 1988827
  • Greenman RL, Schein RM, Martin MA, Wenzel RP, MacIntyre NR, Emmanuel G, Chmel H, Kohler RB, McCarthy M, Plouffe J, et al, The XOMA Sepsis Study Group. A controlled clinical trial of E5 murine monoclonal IgM antibody to endotoxin in the treatment of gram-negative sepsis. JAMA 1991; 266:1097 - 102; http://dx.doi.org/10.1001/jama.1991.03470080067031; PMID: 1865542
  • Bone RC, Balk RA, Fein AM, Perl TM, Wenzel RP, Reines HD, Quenzer RW, Iberti TJ, Macintyre N, Schein RM. A second large controlled clinical study of E5, a monoclonal antibody to endotoxin: results of a prospective, multicenter, randomized, controlled trial. The E5 Sepsis Study Group. Crit Care Med 1995; 23:994 - 1006; http://dx.doi.org/10.1097/00003246-199506000-00003; PMID: 7774238
  • Angus DC, Birmingham MC, Balk RA, Scannon PJ, Collins D, Kruse JA, Graham DR, Dedhia HV, Homann S, MacIntyre N. E5 murine monoclonal antiendotoxin antibody in gram-negative sepsis: a randomized controlled trial. E5 Study Investigators. JAMA 2000; 283:1723 - 30; http://dx.doi.org/10.1001/jama.283.13.1723; PMID: 10755499
  • Albertson TE, Panacek EA, MacArthur RD, Johnson SB, Benjamin E, Matuschak GM, Zaloga G, Maki D, Silverstein J, Tobias JK, et al, MAB-T88 Sepsis Study Group. Multicenter evaluation of a human monoclonal antibody to Enterobacteriaceae common antigen in patients with Gram-negative sepsis. Crit Care Med 2003; 31:419 - 27; http://dx.doi.org/10.1097/01.CCM.0000045564.51812.3F; PMID: 12576946
  • Levin M, Quint PA, Goldstein B, Barton P, Bradley JS, Shemie SD, Yeh T, Kim SS, Cafaro DP, Scannon PJ, et al. Recombinant bactericidal/permeability-increasing protein (rBPI21) as adjunctive treatment for children with severe meningococcal sepsis: a randomised trial. rBPI21 Meningococcal Sepsis Study Group. Lancet 2000; 356:961 - 7; http://dx.doi.org/10.1016/S0140-6736(00)02712-4; PMID: 11041396
  • Abraham E, Glauser MP, Butler T, Garbino J, Gelmont D, Laterre PF, Kudsk K, Bruining HA, Otto C, Tobin E, et al. p55 Tumor necrosis factor receptor fusion protein in the treatment of patients with severe sepsis and septic shock. A randomized controlled multicenter trial. Ro 45-2081 Study Group. JAMA 1997; 277:1531 - 8; http://dx.doi.org/10.1001/jama.1997.03540430043031; PMID: 9153367
  • Abraham E, Laterre PF, Garbino J, Pingleton S, Butler T, Dugernier T, Margolis B, Kudsk K, Zimmerli W, Anderson P, et al, Lenercept Study Group. Lenercept (p55 tumor necrosis factor receptor fusion protein) in severe sepsis and early septic shock: a randomized, double-blind, placebo-controlled, multicenter phase III trial with 1,342 patients. Crit Care Med 2001; 29:503 - 10; http://dx.doi.org/10.1097/00003246-200103000-00006; PMID: 11373411
  • Abraham E, Wunderink R, Silverman H, Perl TM, Nasraway S, Levy H, Bone R, Wenzel RP, Balk R, Allred R, et al. Efficacy and safety of monoclonal antibody to human tumor necrosis factor alpha in patients with sepsis syndrome. A randomized, controlled, double-blind, multicenter clinical trial. TNF-alpha MAb Sepsis Study Group. JAMA 1995; 273:934 - 41; http://dx.doi.org/10.1001/jama.1995.03520360048038; PMID: 7884952
  • Cohen J, Carlet J, International Sepsis Trial Study Group. INTERSEPT: an international, multicenter, placebo-controlled trial of monoclonal antibody to human tumor necrosis factor-alpha in patients with sepsis. Crit Care Med 1996; 24:1431 - 40; http://dx.doi.org/10.1097/00003246-199609000-00002; PMID: 8797612
  • Abraham E, Anzueto A, Gutierrez G, Tessler S, San Pedro G, Wunderink R, Dal Nogare A, Nasraway S, Berman S, Cooney R, et al, NORASEPT II Study Group. Double-blind randomised controlled trial of monoclonal antibody to human tumour necrosis factor in treatment of septic shock. Lancet 1998; 351:929 - 33; PMID: 9734938
  • Rice TW, Wheeler AP, Morris PE, Paz HL, Russell JA, Edens TR, Bernard GR. Safety and efficacy of affinity-purified, anti-tumor necrosis factor-alpha, ovine fab for injection (CytoFab) in severe sepsis. Crit Care Med 2006; 34:2271 - 81; http://dx.doi.org/10.1097/01.CCM.0000230385.82679.34; PMID: 16810105
  • Reinhart K, Wiegand-Löhnert C, Grimminger F, Kaul M, Withington S, Treacher D, Eckart J, Willatts S, Bouza C, Krausch D, et al. Assessment of the safety and efficacy of the monoclonal anti-tumor necrosis factor antibody-fragment, MAK 195F, in patients with sepsis and septic shock: a multicenter, randomized, placebo-controlled, dose-ranging study. Crit Care Med 1996; 24:733 - 42; http://dx.doi.org/10.1097/00003246-199605000-00003; PMID: 8706447
  • Reinhart K, Menges T, Gardlund B, Harm Zwaveling J, Smithes M, Vincent JL, Tellado JM, Salgado-Remigio A, Zimlichman R, Withington S, et al. Randomized, placebo-controlled trial of the anti-tumor necrosis factor antibody fragment afelimomab in hyperinflammatory response during severe sepsis: The RAMSES Study. Crit Care Med 2001; 29:765 - 9; http://dx.doi.org/10.1097/00003246-200104000-00015; PMID: 11373466
  • Dhainaut JF, Vincent JL, Richard C, Lejeune P, Martin C, Fierobe L, Stephens S, Ney UM, Sopwith M. CDP571, a humanized antibody to human tumor necrosis factor-alpha: safety, pharmacokinetics, immune response, and influence of the antibody on cytokine concentrations in patients with septic shock. CPD571 Sepsis Study Group. Crit Care Med 1995; 23:1461 - 9; http://dx.doi.org/10.1097/00003246-199509000-00004; PMID: 7664546
  • Fisher CJ Jr., Opal SM, Dhainaut JF, Stephens S, Zimmerman JL, Nightingale P, Harris SJ, Schein RM, Panacek EA, Vincent JL, et al. Influence of an anti-tumor necrosis factor monoclonal antibody on cytokine levels in patients with sepsis. The CB0006 Sepsis Syndrome Study Group. Crit Care Med 1993; 21:318 - 27; http://dx.doi.org/10.1097/00003246-199303000-00006; PMID: 8440099
  • Dhainaut JF, Tenaillon A, Le Tulzo Y, Schlemmer B, Solet JP, Wolff M, Holzapfel L, Zeni F, Dreyfuss D, Mira JP, et al. Platelet-activating factor receptor antagonist BN 52021 in the treatment of severe sepsis: a randomized, double-blind, placebo-controlled, multicenter clinical trial. BN 52021 Sepsis Study Group. Crit Care Med 1994; 22:1720 - 8; PMID: 7956274
  • Dhainaut JF, Tenaillon A, Hemmer M, Damas P, Le Tulzo Y, Radermacher P, Schaller MD, Sollet JP, Wolff M, Holzapfel L, et al. Confirmatory platelet-activating factor receptor antagonist trial in patients with severe gram-negative bacterial sepsis: a phase III, randomized, double-blind, placebo-controlled, multicenter trial. BN 52021 Sepsis Investigator Group. Crit Care Med 1998; 26:1963 - 71; http://dx.doi.org/10.1097/00003246-199812000-00021; PMID: 9875905
  • Vincent JL, Spapen H, Bakker J, Webster NR, Curtis L. Phase II multicenter clinical study of the platelet-activating factor receptor antagonist BB-882 in the treatment of sepsis. Crit Care Med 2000; 28:638 - 42; http://dx.doi.org/10.1097/00003246-200003000-00006; PMID: 10752807
  • Suputtamongkol Y, Intaranongpai S, Smith MD, Angus B, Chaowagul W, Permpikul C, Simpson JA, Leelarasamee A, Curtis L, White NJ. A double-blind placebo-controlled study of an infusion of lexipafant (Platelet-activating factor receptor antagonist) in patients with severe sepsis. Antimicrob Agents Chemother 2000; 44:693 - 6; http://dx.doi.org/10.1128/AAC.44.3.693-696.2000; PMID: 10681340
  • Froon AM, Greve JW, Buurman WA, van der Linden CJ, Langemeijer HJ, Ulrich C, Bourgeois M. Treatment with the platelet-activating factor antagonist TCV-309 in patients with severe systemic inflammatory response syndrome: a prospective, multi-center, double-blind, randomized phase II trial. Shock 1996; 5:313 - 9; http://dx.doi.org/10.1097/00024382-199605000-00001; PMID: 9156785
  • Poeze M, Froon AH, Ramsay G, Buurman WA, Greve JW. Decreased organ failure in patients with severe SIRS and septic shock treated with the platelet-activating factor antagonist TCV-309: a prospective, multicenter, double-blind, randomized phase II trial. TCV-309 Septic Shock Study Group. Shock 2000; 14:421 - 8; http://dx.doi.org/10.1097/00024382-200014040-00001; PMID: 11049104
  • Rice TW, Wheeler AP, Bernard GR, Vincent JL, Angus DC, Aikawa N, Demeyer I, Sainati S, Amlot N, Cao C, et al. A randomized, double-blind, placebo-controlled trial of TAK-242 for the treatment of severe sepsis. Crit Care Med 2010; 38:1685 - 94; http://dx.doi.org/10.1097/CCM.0b013e3181e7c5c9; PMID: 20562702
  • Tidswell M, Tillis W, Larosa SP, Lynn M, Wittek AE, Kao R, Wheeler J, Gogate J, Opal SM, Eritoran Sepsis Study Group. 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; http://dx.doi.org/10.1097/CCM.0b013e3181b07b78; PMID: 19661804
  • Opal SM, Laterre PF, Francois B, LaRosa SP, Angus DC, Mira JP, Wittebole X, Dugernier T, Perrotin D, Tidswell M, et al, ACCESS Study Group. Effect of eritoran, an antagonist of MD2-TLR4, on mortality in patients with severe sepsis: the ACCESS randomized trial. JAMA 2013; 309:1154 - 62; http://dx.doi.org/10.1001/jama.2013.2194; PMID: 23512062
  • Vincent JL, Ramesh MK, Ernest D, LaRosa SP, Pachl J, Aikawa N, Hoste E, Levy H, Hirman J, Levi M, et al. A randomized, double-blind, placebo-controlled, phase-2B study to evaluate the safety and efficacy of recombinant human soluble thrombomodulin, ART-123, in patients with sepsis and suspected disseminated intravascular coagulation. Crit Care Med 2013; Forthcoming
  • Dellinger RP, Tomayko JF, Angus DC, Opal S, Cupo MA, McDermott S, Ducher A, Calandra T, Cohen J, Lipid Infusion and Patient Outcomes in Sepsis (LIPOS) Investigators. Efficacy and safety of a phospholipid emulsion (GR270773) in Gram-negative severe sepsis: results of a phase II multicenter, randomized, placebo-controlled, dose-finding clinical trial. Crit Care Med 2009; 37:2929 - 38; http://dx.doi.org/10.1097/CCM.0b013e3181b0266c; PMID: 19770753
  • Fein AM, Bernard GR, Criner GJ, Fletcher EC, Good JT Jr., Knaus WA, Levy H, Matuschak GM, Shanies HM, Taylor RW, et al. Treatment of severe systemic inflammatory response syndrome and sepsis with a novel bradykinin antagonist, deltibant (CP-0127). Results of a randomized, double-blind, placebo-controlled trial. CP-0127 SIRS and Sepsis Study Group. JAMA 1997; 277:482 - 7; http://dx.doi.org/10.1001/jama.1997.03540300050033; PMID: 9020273
  • Bernard GR, Wheeler AP, Russell JA, Schein R, Summer WR, Steinberg KP, Fulkerson WJ, Wright PE, Christman BW, Dupont WD, et al, The Ibuprofen in Sepsis Study Group. The effects of ibuprofen on the physiology and survival of patients with sepsis. N Engl J Med 1997; 336:912 - 8; http://dx.doi.org/10.1056/NEJM199703273361303; PMID: 9070471
  • Díaz-Cremades JM, Lorenzo R, Sánchez M, Moreno MJ, Alsar MJ, Bosch JM, Fajardo L, González D, Guerrero D. Use of antithrombin III in critical patients. Intensive Care Med 1994; 20:577 - 80; http://dx.doi.org/10.1007/BF01705725; PMID: 7706571
  • Fourrier F, Chopin C, Huart JJ, Runge I, Caron C, Goudemand J. Double-blind, placebo-controlled trial of antithrombin III concentrates in septic shock with disseminated intravascular coagulation. Chest 1993; 104:882 - 8; http://dx.doi.org/10.1378/chest.104.3.882; PMID: 8365305
  • Eisele B, Lamy M, Thijs LG, Keinecke HO, Schuster HP, Matthias FR, Fourrier F, Heinrichs H, Delvos U. Antithrombin III in patients with severe sepsis. A randomized, placebo-controlled, double-blind multicenter trial plus a meta-analysis on all randomized, placebo-controlled, double-blind trials with antithrombin III in severe sepsis. Intensive Care Med 1998; 24:663 - 72; http://dx.doi.org/10.1007/s001340050642; PMID: 9722035
  • Baudo F, Caimi TM, de Cataldo F, Ravizza A, Arlati S, Casella G, Carugo D, Palareti G, Legnani C, Ridolfi L, et al. Antithrombin III (ATIII) replacement therapy in patients with sepsis and/or postsurgical complications: a controlled double-blind, randomized, multicenter study. Intensive Care Med 1998; 24:336 - 42; http://dx.doi.org/10.1007/s001340050576; PMID: 9609411
  • Warren BL, Eid A, Singer P, Pillay SS, Carl P, Novak I, Chalupa P, Atherstone A, Pénzes I, Kübler A, et al, KyberSept Trial Study Group. Caring for the critically ill patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial. JAMA 2001; 286:1869 - 78; http://dx.doi.org/10.1001/jama.286.15.1869; PMID: 11597289
  • Sprung CL, Caralis PV, Marcial EH, Pierce M, Gelbard MA, Long WM, Duncan RC, Tendler MD, Karpf M. The effects of high-dose corticosteroids in patients with septic shock. A prospective, controlled study. N Engl J Med 1984; 311:1137 - 43; http://dx.doi.org/10.1056/NEJM198411013111801; PMID: 6384785
  • Bone RC, Fisher CJ Jr., Clemmer TP, Slotman GJ, Metz CA, Balk RA. A controlled clinical trial of high-dose methylprednisolone in the treatment of severe sepsis and septic shock. N Engl J Med 1987; 317:653 - 8; http://dx.doi.org/10.1056/NEJM198709103171101; PMID: 3306374
  • Bollaert PE, Charpentier C, Levy B, Debouverie M, Audibert G, Larcan A. Reversal of late septic shock with supraphysiologic doses of hydrocortisone. Crit Care Med 1998; 26:645 - 50; http://dx.doi.org/10.1097/00003246-199804000-00010; PMID: 9559600
  • Oppert M, Schindler R, Husung C, Offermann K, Gräf KJ, Boenisch O, Barckow D, Frei U, Eckardt KU. Low-dose hydrocortisone improves shock reversal and reduces cytokine levels in early hyperdynamic septic shock. Crit Care Med 2005; 33:2457 - 64; http://dx.doi.org/10.1097/01.CCM.0000186370.78639.23; PMID: 16276166
  • Arabi YM, Aljumah A, Dabbagh O, Tamim HM, Rishu AH, Al-Abdulkareem A, Knawy BA, Hajeer AH, Tamimi W, Cherfan A. Low-dose hydrocortisone in patients with cirrhosis and septic shock: a randomized controlled trial. CMAJ 2010; 182:1971 - 7; http://dx.doi.org/10.1503/cmaj.090707; PMID: 21059778
  • Briegel J, Forst H, Haller M, Schelling G, Kilger E, Kuprat G, Hemmer B, Hummel T, Lenhart A, Heyduck M, et al. Stress doses of hydrocortisone reverse hyperdynamic septic shock: a prospective, randomized, double-blind, single-center study. Crit Care Med 1999; 27:723 - 32; http://dx.doi.org/10.1097/00003246-199904000-00025; PMID: 10321661
  • Root RK, Lodato RF, Patrick W, Cade JF, Fotheringham N, Milwee S, Vincent JL, Torres A, Rello J, Nelson S, Pneumonia Sepsis Study Group. Multicenter, double-blind, placebo-controlled study of the use of filgrastim in patients hospitalized with pneumonia and severe sepsis. Crit Care Med 2003; 31:367 - 73; http://dx.doi.org/10.1097/01.CCM.0000048629.32625.5D; PMID: 12576938
  • Presneill JJ, Harris T, Stewart AG, Cade JF, Wilson JW. A randomized phase II trial of granulocyte-macrophage colony-stimulating factor therapy in severe sepsis with respiratory dysfunction. Am J Respir Crit Care Med 2002; 166:138 - 43; http://dx.doi.org/10.1164/rccm.2009005; PMID: 12119223
  • Orozco H, Arch J, Medina-Franco H, Pantoja JP, González QH, Vilatoba M, Hinojosa C, Vargas-Vorackova F, Sifuentes-Osornio J. Molgramostim (GM-CSF) associated with antibiotic treatment in nontraumatic abdominal sepsis: a randomized, double-blind, placebo-controlled clinical trial. Arch Surg 2006; 141:150 - 3, discussion 154; http://dx.doi.org/10.1001/archsurg.141.2.150; PMID: 16490891
  • Meisel C, Schefold JC, Pschowski R, Baumann T, Hetzger K, Gregor J, Weber-Carstens S, Hasper D, Keh D, Zuckermann H, et al. Granulocyte-macrophage colony-stimulating factor to reverse sepsis-associated immunosuppression: a double-blind, randomized, placebo-controlled multicenter trial. Am J Respir Crit Care Med 2009; 180:640 - 8; http://dx.doi.org/10.1164/rccm.200903-0363OC; PMID: 19590022
  • Jaimes F, De La Rosa G, Morales C, Fortich F, Arango C, Aguirre D, Muñoz A. Unfractioned heparin for treatment of sepsis: A randomized clinical trial (The HETRASE Study). Crit Care Med 2009; 37:1185 - 96; http://dx.doi.org/10.1097/CCM.0b013e31819c06bc; PMID: 19242322
  • Staubach KH, Schröder J, Stüber F, Gehrke K, Traumann E, Zabel P. Effect of pentoxifylline in severe sepsis: results of a randomized, double-blind, placebo-controlled study. Arch Surg 1998; 133:94 - 100; http://dx.doi.org/10.1001/archsurg.133.1.94; PMID: 9438767
  • Bakker J, Grover R, McLuckie A, Holzapfel L, Andersson J, Lodato R, Watson D, Grossman S, Donaldson J, Takala J, Glaxo Wellcome International Septic Shock Study Group. Administration of the nitric oxide synthase inhibitor NG-methyl-L-arginine hydrochloride (546C88) by intravenous infusion for up to 72 hours can promote the resolution of shock in patients with severe sepsis: results of a randomized, double-blind, placebo-controlled multicenter study (study no. 144-002). Crit Care Med 2004; 32:1 - 12; http://dx.doi.org/10.1097/01.CCM.0000105118.66983.19; PMID: 14707554
  • Alexander HR, Doherty GM, Buresh CM, Venzon DJ, Norton JA. A recombinant human receptor antagonist to interleukin 1 improves survival after lethal endotoxemia in mice. J Exp Med 1991; 173:1029 - 32; http://dx.doi.org/10.1084/jem.173.4.1029; PMID: 1826127
  • Fischer E, Marano MA, Van Zee KJ, Rock CS, Hawes AS, Thompson WA, DeForge L, Kenney JS, Remick DG, Bloedow DC, et al. Interleukin-1 receptor blockade improves survival and hemodynamic performance in Escherichia coli septic shock, but fails to alter host responses to sublethal endotoxemia. J Clin Invest 1992; 89:1551 - 7; http://dx.doi.org/10.1172/JCI115748; PMID: 1533231
  • Van Zee KJ, Moldawer LL, Oldenburg HS, Thompson WA, Stackpole SA, Montegut WJ, Rogy MA, Meschter C, Gallati H, Schiller CD, et al. Protection against lethal Escherichia coli bacteremia in baboons (Papio anubis) by pretreatment with a 55-kDa TNF receptor (CD120a)-Ig fusion protein, Ro 45-2081. J Immunol 1996; 156:2221 - 30; PMID: 8690912
  • Redl H, Schlag G, Paul E, Bahrami S, Buurman WA, Strieter RM, Kunkel SL, Davies J, Foulkes R. Endogenous modulators of TNF and IL-1 response are under partial control of TNF in baboon bacteremia. Am J Physiol 1996; 271:R1193 - 8; PMID: 8945953
  • Myers AK, Robey JW, Price RM. Relationships between tumour necrosis factor, eicosanoids and platelet-activating factor as mediators of endotoxin-induced shock in mice. Br J Pharmacol 1990; 99:499 - 502; http://dx.doi.org/10.1111/j.1476-5381.1990.tb12957.x; PMID: 2110016
  • Giral M, Balsa D, Ferrando R, Merlos M, Garcia-Rafanell J, Forn J. Effects of UR-12633, a new antagonist of platelet-activating factor, in rodent models of endotoxic shock. Br J Pharmacol 1996; 118:1223 - 31; http://dx.doi.org/10.1111/j.1476-5381.1996.tb15527.x; PMID: 8818347
  • Ogata M, Matsumoto T, Koga K, Takenaka I, Kamochi M, Sata T, Yoshida S, Shigematsu A. An antagonist of platelet-activating factor suppresses endotoxin-induced tumor necrosis factor and mortality in mice pretreated with carrageenan. Infect Immun 1993; 61:699 - 704; PMID: 8423096
  • Takashima K, Matsunaga N, Yoshimatsu M, Hazeki K, Kaisho T, Uekata M, Hazeki O, Akira S, Iizawa Y, Ii M. Analysis of binding site for the novel small-molecule TLR4 signal transduction inhibitor TAK-242 and its therapeutic effect on mouse sepsis model. Br J Pharmacol 2009; 157:1250 - 62; http://dx.doi.org/10.1111/j.1476-5381.2009.00297.x; PMID: 19563534
  • Sha T, Sunamoto M, Kitazaki T, Sato J, Ii M, Iizawa Y. Therapeutic effects of TAK-242, a novel selective Toll-like receptor 4 signal transduction inhibitor, in mouse endotoxin shock model. Eur J Pharmacol 2007; 571:231 - 9; http://dx.doi.org/10.1016/j.ejphar.2007.06.027; PMID: 17632100
  • Creasey AA, Chang AC, Feigen L, Wün TC, Taylor FB Jr., Hinshaw LB. Tissue factor pathway inhibitor reduces mortality from Escherichia coli septic shock. J Clin Invest 1993; 91:2850 - 60; http://dx.doi.org/10.1172/JCI116529; PMID: 8514893