ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Investigational and ineffective pharmacologic therapies for sepsis

Investigational and ineffective pharmacologic therapies for sepsis
Literature review current through: Jan 2024.
This topic last updated: Nov 13, 2023.

INTRODUCTION — A number of potential therapies have been investigated or are being evaluated in an attempt to improve clinical outcomes in sepsis. Therapies that are being investigated are the focus of this review, although none are recommended for use at this time because the ratio of potential benefits and harms has not been adequately studied. Therapies that have proven ineffective are also mentioned briefly. The definition, pathophysiology, and management of sepsis are discussed elsewhere. (See "Sepsis syndromes in adults: Epidemiology, definitions, clinical presentation, diagnosis, and prognosis" and "Evaluation and management of suspected sepsis and septic shock in adults".)

THERAPIES BEING INVESTIGATED — A number of potential therapies for sepsis appear promising in animal models, but have not yet been adequately studied in humans. Other potential therapies have been studied in humans, but have given conflicting results and require additional investigations to clarify their effects. These therapies are the focus of this section. Often there is no better therapy to offer a patient than enrollment onto a well-designed, scientifically valid, peer-reviewed clinical trial. Additional information and instructions for referring a patient to an appropriate research center can be obtained from the United States National Institutes of Health (www.clinicaltrials.gov).

Inhibition of innate immunity — Infecting microbes display highly conserved macromolecules (eg, lipopolysaccharides, peptidoglycans) on their surface. When these macromolecules are recognized by pattern-recognition receptors (called Toll-like receptors [TLRs]) on the surface of immune cells, the host's immune response is initiated. This may contribute to the excess systemic inflammatory response that characterizes sepsis. Inhibition of several TLRs is being evaluated as a potential therapy for sepsis:

Inhibition of TLR-4 with the antagonist, E5564 (Eritoran) was tested in humans with severe sepsis in the ACCESS (A Controlled Comparison of Eritoran Tetrasodium and Placebo in Patients with Severe Sepsis) Trial, showing no effect on 28-day mortality [1]. An earlier clinical trial that evaluated the inhibition of TLR-4 with another antagonist, TAK 242 (Resatorvid), found a non-statistically significant reduction in 28-day mortality [2].

The triggering receptor expressed on myeloid cells 1 (TREM-1) is an activating receptor expressed on innate immune cells and activated endothelial cells. TREM-1 activation results in dysregulation of innate immune responses [3]. Cellular TREM-1 is upregulated in septic shock, where it also exists in a soluble form (sTREM-1) following cleavage from the membrane-bound form. Concentrations of sTREM-1 correlate with Sequential Organ Failure Assessment (SOFA) scores, suggesting a causal role for TREM-1 in septic organ failures. Nangibotide, a ligand-trapping molecule that inhibits TREM-1 activation, was well tolerated in a small trial of septic shock [4]. The ASTONISH Phase IIb trial of nangibotide in septic shock is currently enrolling with a target of 450 subjects [5].

Intravenous immune globulin — It has been hypothesized that polyclonal intravenous (IV) immune globulin (IVIG) may benefit patients with sepsis by binding endotoxin. However, we and others believe that IVIG should not be administered to patients with sepsis [6-13]:

Suggesting that polyclonal IVIG has no benefit, a randomized trial of 653 patients with sepsis found that IVIG did not reduce 28-day mortality compared to placebo [6].

Suggesting that polyclonal IVIG has a benefit, meta-analyses of randomized trials found that polyclonal IVIG decreased mortality compared to either placebo or no IVIG [7-10]. The mortality benefit was greatest when IgA- or IgM-enriched IVIG was used [9,10]. However, all of the meta-analyses were limited by heterogeneity and two of the meta-analyses found that the mortality benefit disappeared when only the well-designed trials were analyzed [8,10].

Well-designed randomized trials are warranted to evaluate the impact of IgA- and IgM-enriched IVIG [12]. (See "Overview of intravenous immune globulin (IVIG) therapy".)

Cytokine and endotoxin inactivation or removal — Strategies to improve sepsis by either inactivating or removing endotoxin and/or inflammatory cytokines have been investigated:

Hemoperfusion through adsorptive materials – Perfusion of blood from patents with sepsis through adsorptive membranes or sorbent-containing cartridges can remove harmful circulating cytokines and endotoxin. While initial trials were hopeful [14-18], several trials have reported that hemoperfusion through a membranous polymyxin B fiber column (PBFC; high affinity for endotoxin) is not beneficial [19,20]. A post-hoc analysis suggested some possible benefit in those with high levels of endotoxin activity [21].

Hemoperfusion through sorbent-containing cartridges (hemadsorption) provides a larger surface area than membranous columns to enhance the removal of cytokines and inflammatory molecules involved in the pathogenesis of sepsis. Among the existing sorbents, CytoSorb (porous polymer beads) appears to be the most promising in animal models with limited data in humans [22-25]. However, it does not remove the two more potent inflammatory molecules, endotoxin and interleukin-10.

Plasma or whole blood exchange – Case series and observational studies with historical controls reported favorable outcomes when endotoxin was removed by plasma or whole blood exchange, including lower plasma endotoxin concentrations and, possibly, lower mortality [26-28].

Coupled plasma filtration adsorption (CPFA) – CPFA combines the separation of plasma from the cellular components of blood with a highly permeable filter (plasma filter) followed by sorbent adsorption of the plasma component with a styrene resin. In one multicenter trial of 330 adult patients with septic shock, compared to conventional care, CPFA was not associated with a mortality benefit (45 versus 47 percent) nor did it prevent the development of new organ failures or decrease length of ICU stay [29].

Interleukin-1 is a pro-inflammatory cytokine implicated in sepsis-related multiorgan dysfunction syndrome. Anakinra (a recombinant, non-glycosylated human interleukin-1 receptor antagonist) failed to improve survival in two clinical trials enrolling subjects with severe sepsis and septic shock [30,31]. Recent reports that anakinra is effective for patients with macrophage activation syndrome have rekindled interest in identifying subsets of severe sepsis in which the drug may be useful. When data from the original confirmatory phase III trial [30] were reanalyzed, grouping subjects according to the presence of hepatobiliary dysfunction and disseminated intravascular coagulation (as markers for macrophage activation), treatment with anakinra was associated with improved survival in those with macrophage activation, but not in those without [32]. The patients who met criteria for macrophage activation was a small subset of the overall group, so this finding (if confirmed) may pertain to few patients with septic shock.

Xuebijing (XBJ) is an herbal preparation that has an antagonistic effect on endotoxin and is an inhibitor of inflammatory mediator release by endotoxin-stimulated monocytes/macrophages [33-35]. Preliminary data suggest possible benefit in patients with sepsis [36,37]. One randomized trial of 1817 patients with sepsis reported that intravenous XBJ reduced 28-day mortality compared with placebo (19 versus 26 percent) [38]. The most common adverse event associated with XBJ was elevated transaminases and white blood cell count, but no difference in adverse event rates was reported between the groups. However, several limitations in the trial design may have influenced the outcome.

While some of these therapies are promising, we believe that additional evidence from large, randomized trials are necessary before any of these strategies can be routinely used in patients with sepsis.

Interferon-gamma — A decrease in monocyte function has been observed late in the course of sepsis, which may predispose patients to life-threatening secondary infections. This observation prompted a study in which patients with sepsis were administered interferon-gamma [39]. The study found that interferon-gamma restored monocytic cell function. Controlled clinical trials measuring patient-important outcomes in patients with sepsis have not been reported, although a trial evaluating the impact of interferon-gamma in candidemia is underway [40].

Granulocyte-macrophage colony stimulating factor — Granulocyte-macrophage colony stimulating factor (GM-CSF, sargramostim, molgramostim) is a cytokine that promotes maturation of the progenitor cells of granulocytes, erythrocytes, megakaryocytes, and macrophages, as well as mature neutrophils, monocytes, macrophages, dendritic cells, T-lymphocytes, and plasma cells. Its use in sepsis has been studied in several controlled clinical trials.

One trial reported that GM-CSF increased the peripheral neutrophil count, but did not affect the mortality, duration of ICU stay, or rate of hospital discharge [41]. Another trial found that GM-CSF reduced the rate of infectious complications, the length of hospital stay, and the duration of antibiotic therapy, without affecting mortality [42]. And, a third trial found that GM-CSF induced a trend toward a shorter duration of mechanical ventilation (148 versus 207 hours), a shorter hospital stay (59 versus 69 days), and a shorter ICU stay (41 versus 52 days) [43]. All of the trials were small with few events, which is the reason why large absolute differences were often not statistically significant. We believe that larger controlled clinical trials are necessary to confirm the clinical effects reported in these trials and that GM-CSF should not be routinely used to treat sepsis until then.

The effects of granulocyte colony stimulating factor (G-CSF) are mentioned below. (See 'Ineffective therapies' below.)

Augmentation of immunomodulation — Antibodies directed against macrophage migration inhibition factor (MIF) prevented death in a study that used a cecal puncture animal model of sepsis, even when administered up to eight hours after the onset of peritonitis [44]. The mechanism of action is uncertain, but MIF inhibition might restore or augment the immunomodulatory actions of endogenous glucocorticoids [45]. While it is known that elevated MIF levels correlate with poor outcomes in humans with sepsis [45,46], the impact of MIF inhibition has not been studied in humans.

Stem cell therapy — Multipotent mesenchymal stem cells modulate several aspects of the immune response, reprogram the immune system to ameliorate host damage, enhance tissue repair, display antimicrobial properties, and reduce the propensity to multiorgan failure in animal models of sepsis. A growing safety record in human trials provides grounds for optimism that stem cell-based therapies may find a place in clinical medicine [47].

Immunostimulation — Anti-inflammatory pathways activated in septic patients may raise the risk of secondary infections. Treatments that stimulate immunity, such as IL-7, IL-15, or anti-PDL1, offer a promising option [48].

Inhibition of proinflammatory gene expression — A synthetic peptide has been developed that inhibits bacterial superantigen-induced expression of certain proinflammatory genes (interleukin-2, gamma interferon, and tumor necrosis factor-beta) by limiting T-cell activation. In one study, the peptide protected mice against lethal challenges with staphylococcal and streptococcal superantigen, when administered up to three hours after the superantigen [49]. This peptide has not been tested in humans.

Hemofiltration — It has been proposed that hemofiltration, as a mode of dialysis, may remove proinflammatory molecules that induce hemodynamic collapse in septic shock, thereby improving outcomes. However, data from small randomized trials of hemofiltration (high volume or continuous) in patients with septic shock suggest that there is insufficient evidence for the routine use of this mode of renal replacement therapy over conventional hemodialysis.

Initial studies suggested that the high volume hemofiltration offered benefit over conventional hemodialysis for the treatment of acute renal failure in sepsis [50-53]. However, meta-analyses of small randomized studies and one multicenter prospective study (IVOIRE) suggested no benefit [54,55].

Similarly, a randomized trial that evaluated continuous venovenous hemofiltration (CVVH) in patients with sepsis did not demonstrate any improvement in the clearance of inflammatory mediators or clinical outcomes [56]. Another trial was discontinued after an interim analysis showed more frequent and more severe organ failure in the hemofiltration group [57].

Anticoagulants — Heparin has anti-thrombotic and immunomodulating effects, both of which have the potential to be beneficial in sepsis. In a retrospective cohort study of patients with septic shock, intravenous therapeutic heparin was associated with decreased mortality, discontinuation of vasoactive drug infusions, and liberation from mechanical ventilation [58]. The risk of major hemorrhage or the need for transfusion was not increased. This study was followed by the HETRASE trial, which randomly assigned 319 patients with sepsis to receive low dose intravenous heparin (500 units per hour) or placebo for seven days [59]. The trial found no improvement in any clinical outcome. It has been suggested that the conflicting results were due to the randomized trial using a low dose of heparin; as a result, a randomized trial comparing various doses of heparin in patients with septic shock is being conducted [60].

Several early studies and a subsequent meta-analysis suggested a potential benefit of recombinant thrombomodulin in sepsis-induced disseminated intravascular coagulation [61,62]. However, a large randomized trial found no benefit to thrombomodulin therapy in sepsis-associated coagulopathy [63].

Therapeutic studies of anticoagulation in coronavirus disease 2019 (COVID-19) are discussed separately. (See "COVID-19: Hypercoagulability".)

Prostacyclin analogs — Iloprost is a prostacyclin analog that targets the microcirculatory perturbations of septic shock through vasodilatory and anti-thrombotic mechanisms. An initial case series of three patients receiving iloprost described improvements of hypoperfusion and arterial lactate concentrations without effects upon blood pressure [64]. A phase 2 trial enrolled 24 subjects randomized to placebo versus iloprost combined with the glycoprotein IIb/IIIa inhibitor eptifibatide, administered within 24 hours of onset of septic shock [65]. Compared to placebo, iloprost/eptifibatide was associated with a reduction in biomarkers of endothelial injury and fibrinolysis. Iloprost/eptifibatide was also associated with a reduction in SOFA scores. Phase 3 trials of iloprost are currently recruiting subjects [66,67].

Naloxone — A meta-analysis of three randomized trials (61 patients) comparing naloxone to either placebo or no naloxone found that naloxone therapy led to hemodynamic improvement, but did not improve the case-fatality rate [68]. Adverse effects that have been associated with naloxone therapy include pulmonary edema, hypertension, and seizures [69]. Given the limitations of the meta-analysis that found a physiological benefit, the absence of patient-important benefits, and the potential for adverse effects, we believe that naloxone therapy is not warranted for patients with septic shock. However, large controlled trials are justified, since the studies included in the meta-analysis had too few events to definitively exclude a beneficial effect.

Pentoxifylline — Sepsis results in decreased red cell deformability and increased erythrocyte aggregation, effects that may be mitigated by pentoxifylline [70]. Pentoxifylline also inhibits neutrophil adhesion and activation, and modulates endotoxin-induced expression of proinflammatory cytokines [71]. In a trial of 51 surgical patients with severe sepsis who were randomly assigned to receive pentoxifylline or placebo, pentoxifylline improved the multiple organ dysfunction score and the arterial oxygen tension to fraction of inspired oxygen (PaO2/FiO2), but there was no improvement in the 28-day mortality [72]. Additional trials are needed to identify a true beneficial clinically meaningful effect.

Statins — HMG CoA reductase inhibitors (statins) are in wide clinical use and have an established role in the management of hyperlipidemia and cardiovascular disease. Statins also appear to have beneficial anti-inflammatory properties, such as suppression of endotoxin-induced up-regulation of TLR-4 and TLR-2. Although early studies of statins in patients with sepsis suggested benefit, most studies since then, including meta-analyses of randomized trials, show no benefit from statin use in this population [73-82].

Heart rate control — Beta-blocker therapy may potentially attenuate the deleterious effects of the sympathetic adrenergic response that occurs during septic shock. However, data are limited and some suggest harm such that this strategy is not routinely employed [83-87]:

Esmolol – One open-label, single-center trial randomized 154 patients with septic shock to an esmolol infusion or standard therapy [83]. All patients on esmolol achieved the preset target heart rate (HR) of 80 to 94 beats/min without a significant drop in mean arterial pressure (median dose 100 mg/hour). Compared with patients on standard therapy, patients receiving esmolol had improvements in the following variables at 96 hours:

Greater decline from baseline HR (by 28 beats/min versus 6 beats/min)

Higher left ventricular stroke work index (43 versus 31 mL/m2)

Reduced need for vasopressors (-0.11 mcg/kg/min versus -0.01 mcg/kg/min)

Reduced need for fluid replacement therapy (4L/day versus 5.4 L/day)

Improvements were also observed in secondary outcomes that included metabolic variables (eg, lactate and pH), markers of end organ function (eg, glomerular filtration rate), and 28-day mortality (50 versus 81 percent). A meta-analysis of seven randomized trials of esmolol (or landiolol) reported a possible increase in survival associated with short-acting beta blockade [87].

Landiolol – While one trial of landiolol in patients with sepsis-related tachycardia reported a reduction in the rate of new-onset tachyarrhythmia [84], another trial suggested harm [86]. In a randomized trial of 126 patients with septic shock-related tachycardia (heart rate ≥95/min) who were treated for at least 24 hours with continuous norepinephrine, landiolol did not reduce organ failure as measured by the SOFA score and was additionally associated with an increased 28-day mortality compared with standard care (37 versus 25 percent) [86].

Ivabrandine – Ivabradine also lowers heart rate through inhibition of ion channels in the sinoatrial node. In an animal model of sepsis, ivabradine mitigated septic tachycardia similar to atenolol [85]. Unlike atenolol, ivabradine did not decrease cardiac output or left ventricular ejection fraction. In a randomized, controlled study of clinical septic shock, enteral ivabradine led to decreased heart rates, lower SOFA scores, and increased stroke volume and ejection fraction compared with placebo. However, ivabradine had no effect on cardiac output or mortality. Accordingly, ivabradine cannot be recommended therapy for septic shock at this time.

Vitamin C — Vitamin C has gained popularity as a therapy for sepsis, either alone or in combination with thiamine and hydrocortisone. Hydrocortisone and vitamin C act synergistically on the inflammatory cascade. Vitamin C is also an antioxidant. Thiamine (vitamin B1) prevents renal oxalate crystallization when vitamin C is administered in high doses.

While one early retrospective study suggested a possible reduction in mortality, most studies since then have not been favorable for vitamin C-containing regimens [88-95], supporting the Society of Critical Care Medicine/European Society of ICU Medicine recommendation against its use [11].

Studies favoring vitamin C regimens:

One retrospective single institution study of 47 patients with sepsis or septic shock and a procalcitonin level >2 ng/mL, examined the impact of high-dose intravenous (IV) vitamin C (1.5 g every six hours for four days or until ICU discharge), IV thiamine (200 mg every 12 hours for four days or until ICU discharge), and IV hydrocortisone (50 mg every six hours for seven days or until ICU discharge) given early during the course of sepsis (ie, within the first 24 hours of intensive care [ICU] admission) [88]. This regimen was associated with a reduction in in-hospital mortality (8.5 versus 40.4 percent), more rapid weaning of vasopressors (18 versus 55 hours), and the prevention of progressive organ dysfunction compared with 47 historical controls who were treated in an era prior to the institution of the vitamin C protocol.

A preliminary randomized trial of 167 patients with sepsis and acute respiratory distress syndrome reported no effect of a 96-hour infusion of vitamin C on organ failure (as assessed by the SOFA score), levels of inflammatory markers (C-reactive protein), or vascular injury (thrombomodulin) [91]. While vitamin C reduced the crude 28-day mortality (30 versus 46.3 percent) and ICU- and hospital-free days, the analyses were not adjusted for confounding variables and are therefore considered exploratory.

Studies not favoring vitamin C:

Three randomized trials of patients with sepsis or septic shock compared a regimen of IV vitamin C (1.5 g every six hours), IV hydrocortisone (50 mg every six hours), and IV thiamine (200 mg every 12 hours) with either IV hydrocortisone alone [92] or placebo [94,95]. None of the trials showed improvement in 28-day mortality, ventilator- or vasopressor-free days, or rate of acute kidney injury. Adverse events included hyperglycemia (6 versus 3 percent), hypernatremia (5 versus 3 percent), and new hospital-acquired infection (6 percent each). However, the trials had several limitations, including open-label design and lack of blinded outcome assessment for one [92] and another was underpowered to detect a significant difference, which may affect the interpretation of these data.

Another study of 872 patients with septic shock who were receiving a vasopressor, IV vitamin C alone had no effect on 28-day mortality (53 versus 32 percent) or persistent organ dysfunction (defined by the use of vasopressors, invasive mechanical ventilation, or new renal-replacement therapy; 9 versus 7 percent) [96]. One patient had severe hypoglycemia and another had anaphylaxis in response to vitamin C.

Several meta-analyses suggest similar lack of benefit [97-99], but several studies remain in progress.

Targeting bioenergetic failure — Mitochondrial dysfunction is present in sepsis and could contribute to immune failure, cell death, and multiorgan impairment. For example, activating mitochondrial biogenesis using inhaled carbon monoxide reduced liver injury in a model of sepsis [100]. Treatments that protect mitochondria or enhance mitochondrial biogenesis might prove effective.

INEFFECTIVE THERAPIES — A number of potential therapies for sepsis have been investigated, but either caused harm or failed to improve clinical outcomes.

Despite promising initial data, recombinant human activated protein C (rhAPC) has not been confirmed to improve survival in patients with severe sepsis or septic shock, prompting withdrawal of this drug from the market [101]. Recombinant human activated protein C (also called drotrecogin alfa) promotes fibrinolysis and inhibits thrombosis. It was hypothesized that rhAPC may benefit patients with sepsis because it modulates the procoagulant response that is believed to contribute to multisystem organ dysfunction. This hypothesis was initially tested in the PROWESS trial, which reported that rhAPC improved 28-day mortality in patients with severe sepsis or septic shock, with its greatest benefit among patients with a high risk of death (ie, APACHE II score ≥25) [102]. However, conflicting data from subsequent studies eventually led to a new trial, the PROWESS-SHOCK trial. In this trial, 1696 patients with vasopressor-dependent septic shock were randomly assigned to receive rhAPC or placebo [103]. Preliminary analyses done by the maker of the drug indicated that rhAPC did not improve 28-day mortality (26.4 versus 24.2 percent for placebo; relative risk 1.09, 95% CI 0.92-1.28).

Additional therapies that have proven ineffective include the following:

The Toll-like receptor (TLR)-4 antagonist, TAK 242 (Resatorvid) [2]

The human anti-endotoxin monoclonal antibody, HA-1A [104,105]

The human anti-Enterobacteriaceae common antigen (ECA) monoclonal antibody [106]

Alkaline phosphatase [107]

Granulocyte colony-stimulating factor (filgrastim, G-CSF) [108]

Anti-tumor necrosis factor monoclonal antibody [109-111]

Tumor necrosis factor receptor antagonist [110,112]

Interleukin-1 receptor antagonist [30]

Antithrombin (formerly known as antithrombin III) [11,113-120]

Recombinant human tissue factor pathway inhibitor (tifacogin) [121,122]

Ibuprofen [123]

N-acetylcysteine [124,125]

Nitric oxide inhibitors [126-132]

The bradykinin antagonist, deltibant [133]

Growth hormone [134]

Intravenous selenium supplementation [11,135]

Talactoferrin – restores the barrier properties of the gastrointestinal mucosa [136-140]

Calcitriol [141]

Levosimendan [142]

Hypothermia [143]

Hyperoxia [144]

Hypertonic saline [144]

Hemoperfusion through a membranous polymyxin B fiber column (PBFC) [19,20]

Recombinant human soluble thrombomodulin (for sepsis-associated coagulopathy) [63]

High-dose vitamin D (in vitamin D deficient patients) [145]

SUMMARY AND RECOMMENDATIONS

A number of potential therapies for sepsis appear promising in animal models but have not yet been adequately studied in humans. These include toll-like receptor antagonists and neutralizing antibodies, iloprost, interferon gamma, macrophage migration inhibition factor neutralizing antibody, and a synthetic peptide that inhibits bacterial superantigen-induced expression of certain proinflammatory genes. (See 'Inhibition of innate immunity' above and 'Interferon-gamma' above and 'Augmentation of immunomodulation' above and 'Inhibition of proinflammatory gene expression' above.)

Other potential therapies for sepsis have been studied in humans, but have provided conflicting results and require additional investigations to clarify their effects. These include polyclonal intravenous immune globulin (IVIG), interleukin-1 receptor antagonists, hemoperfusion through adsorptive materials or membranes, plasma exchange, whole blood exchange, coupled plasma filtration adsorption, granulocyte-macrophage colony stimulating factor (GM-CSF), hemofiltration, anticoagulants, naloxone, pentoxifylline, and statins. (See 'Intravenous immune globulin' above and 'Cytokine and endotoxin inactivation or removal' above and 'Granulocyte-macrophage colony stimulating factor' above and 'Hemofiltration' above and 'Anticoagulants' above and 'Naloxone' above and 'Pentoxifylline' above and 'Statins' above.)

Potential therapies that require further validation of benefit in patients with septic shock include therapy with short-acting beta blockers esmolol or landiolol. (See 'Heart rate control' above.)

Potential therapies for sepsis have also been investigated that were found to either cause harm or not improve clinical outcomes. (See 'Ineffective therapies' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Peter F Clardy, MD, who contributed to earlier versions of this topic review.

  1. 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.
  2. 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.
  3. Klesney-Tait J, Turnbull IR, Colonna M. The TREM receptor family and signal integration. Nat Immunol 2006; 7:1266.
  4. François B, Wittebole X, Ferrer R, et al. Nangibotide in patients with septic shock: a Phase 2a randomized controlled clinical trial. Intensive Care Med 2020; 46:1425.
  5. Francois B, Lambden S, Gibot S, et al. Rationale and protocol for the efficacy, safety and tolerability of nangibotide in patients with septic shock (ASTONISH) phase IIb randomised controlled trial. BMJ Open 2021; 11:e042921.
  6. Werdan K, Pilz G, Bujdoso O, et al. Score-based immunoglobulin G therapy of patients with sepsis: the SBITS study. Crit Care Med 2007; 35:2693.
  7. Alejandria MM, Lansang MA, Dans LF, Mantaring JB 3rd. Intravenous immunoglobulin for treating sepsis, severe sepsis and septic shock. Cochrane Database Syst Rev 2013; :CD001090.
  8. Pildal J, Gøtzsche PC. Polyclonal immunoglobulin for treatment of bacterial sepsis: a systematic review. Clin Infect Dis 2004; 39:38.
  9. Kreymann KG, de Heer G, Nierhaus A, Kluge S. Use of polyclonal immunoglobulins as adjunctive therapy for sepsis or septic shock. Crit Care Med 2007; 35:2677.
  10. Laupland KB, Kirkpatrick AW, Delaney A. Polyclonal intravenous immunoglobulin for the treatment of severe sepsis and septic shock in critically ill adults: a systematic review and meta-analysis. Crit Care Med 2007; 35:2686.
  11. Evans L, Rhodes A, Alhazzani W, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock 2021. Crit Care Med 2021; 49:e1063.
  12. Neugebauer EA. To use or not to use? Polyclonal intravenous immunoglobulins for the treatment of sepsis and septic shock. Crit Care Med 2007; 35:2855.
  13. Tagami T, Matsui H, Fushimi K, Yasunaga H. Intravenous immunoglobulin and mortality in pneumonia patients with septic shock: an observational nationwide study. Clin Infect Dis 2015; 61:385.
  14. Vincent JL, Laterre PF, Cohen J, et al. A pilot-controlled study of a polymyxin B-immobilized hemoperfusion cartridge in patients with severe sepsis secondary to intra-abdominal infection. Shock 2005; 23:400.
  15. Aoki H, Kodama M, Tani T, Hanasawa K. Treatment of sepsis by extracorporeal elimination of endotoxin using polymyxin B-immobilized fiber. Am J Surg 1994; 167:412.
  16. Iwama H, Komatsu T. Effect of an endotoxin-removing column containing immobilized polymyxin B fiber in a patient with septic shock from gram-positive infection. Acta Anaesthesiol Scand 1998; 42:590.
  17. Cruz DN, Antonelli M, Fumagalli R, et al. Early use of polymyxin B hemoperfusion in abdominal septic shock: the EUPHAS randomized controlled trial. JAMA 2009; 301:2445.
  18. Iwagami M, Yasunaga H, Doi K, et al. Postoperative polymyxin B hemoperfusion and mortality in patients with abdominal septic shock: a propensity-matched analysis. Crit Care Med 2014; 42:1187.
  19. Fujii T, Ganeko R, Kataoka Y, et al. Polymyxin B-immobilized hemoperfusion and mortality in critically ill adult patients with sepsis/septic shock: a systematic review with meta-analysis and trial sequential analysis. Intensive Care Med 2018; 44:167.
  20. Dellinger RP, Bagshaw SM, Antonelli M, et al. Effect of Targeted Polymyxin B Hemoperfusion on 28-Day Mortality in Patients With Septic Shock and Elevated Endotoxin Level: The EUPHRATES Randomized Clinical Trial. JAMA 2018; 320:1455.
  21. Klein DJ, Foster D, Walker PM, et al. Polymyxin B hemoperfusion in endotoxemic septic shock patients without extreme endotoxemia: a post hoc analysis of the EUPHRATES trial. Intensive Care Med 2018; 44:2205.
  22. Honore PM, Jacobs R, Joannes-Boyau O, et al. Newly designed CRRT membranes for sepsis and SIRS--a pragmatic approach for bedside intensivists summarizing the more recent advances: a systematic structured review. ASAIO J 2013; 59:99.
  23. Quintel M. CytoSorb™ whole blood cytokine adsorption–results of a controlled randomized trial. Presented at the 32nd International Symposium on Intensive Care and Emergency Medicine. March 20–23, 2012
  24. Peng ZY, Carter MJ, Kellum JA. Effects of hemoadsorption on cytokine removal and short-term survival in septic rats. Crit Care Med 2008; 36:1573.
  25. Schadler, D., Brederlau J., Jorres, A., Marx, G., Meier-Hellmann, A., Putensen, C., Quintel, M., Spies, C., Porzelius, C., Engel, C., Weiler, N., Kuhlmann, M. Extracorporeal cytokine hemoadsorption in patients with severe sepsis and acute lung injury. Am J Respr Crit Care Med , 2013, pp.A5241
  26. van Deuren M, Santman FW, van Dalen R, et al. Plasma and whole blood exchange in meningococcal sepsis. Clin Infect Dis 1992; 15:424.
  27. Reeves JH. A review of plasma exchange in sepsis. Blood Purif 2002; 20:282.
  28. Stegmayr BG. Plasmapheresis in severe sepsis or septic shock. Blood Purif 1996; 14:94.
  29. Livigni S, Bertolini G, Rossi C, et al. Efficacy of coupled plasma filtration adsorption (CPFA) in patients with septic shock: a multicenter randomised controlled clinical trial. BMJ Open 2014; 4:e003536.
  30. Opal SM, Fisher CJ Jr, Dhainaut JF, 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.
  31. Fisher CJ Jr, Dhainaut JF, Opal SM, 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.
  32. Shakoory B, Carcillo JA, Chatham WW, et al. Interleukin-1 Receptor Blockade Is Associated With Reduced Mortality in Sepsis Patients With Features of Macrophage Activation Syndrome: Reanalysis of a Prior Phase III Trial. Crit Care Med 2016; 44:275.
  33. Liu MW, Su MX, Zhang W, et al. Protective effect of Xuebijing injection on paraquat-induced pulmonary injury via down-regulating the expression of p38 MAPK in rats. BMC Complement Altern Med 2014; 14:498.
  34. Jiang M, Zhou M, Han Y, et al. Identification of NF-κB Inhibitors in Xuebijing injection for sepsis treatment based on bioactivity-integrated UPLC-Q/TOF. J Ethnopharmacol 2013; 147:426.
  35. Li J, Olaleye OE, Yu X, et al. High degree of pharmacokinetic compatibility exists between the five-herb medicine XueBiJing and antibiotics comedicated in sepsis care. Acta Pharm Sin B 2019; 9:1035.
  36. Li C, Wang P, Zhang L, et al. Efficacy and safety of Xuebijing injection (a Chinese patent) for sepsis: A meta-analysis of randomized controlled trials. J Ethnopharmacol 2018; 224:512.
  37. Shi H, Hong Y, Qian J, et al. Xuebijing in the treatment of patients with sepsis. Am J Emerg Med 2017; 35:285.
  38. Liu S, Yao C, Xie J, et al. Effect of an Herbal-Based Injection on 28-Day Mortality in Patients With Sepsis: The EXIT-SEP Randomized Clinical Trial. JAMA Intern Med 2023; 183:647.
  39. Döcke WD, Randow F, Syrbe U, et al. Monocyte deactivation in septic patients: restoration by IFN-gamma treatment. Nat Med 1997; 3:678.
  40. Efficacy of Interferon-gamma in Combination With Anidulafungin for the Treatment of Candidemia. http://www.clinicaltrials.gov/ct2/show/NCT01270490 (Accessed on June 12, 2011).
  41. Presneill JJ, Harris T, Stewart AG, et al. 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.
  42. Orozco H, Arch J, Medina-Franco H, et al. Molgramostim (GM-CSF) associated with antibiotic treatment in nontraumatic abdominal sepsis: a randomized, double-blind, placebo-controlled clinical trial. Arch Surg 2006; 141:150.
  43. Meisel C, Schefold JC, Pschowski R, 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.
  44. Calandra T, Echtenacher B, Roy DL, et al. Protection from septic shock by neutralization of macrophage migration inhibitory factor. Nat Med 2000; 6:164.
  45. Martin TR. MIF mediation of sepsis. Nat Med 2000; 6:140.
  46. Bozza FA, Gomes RN, Japiassú AM, et al. Macrophage migration inhibitory factor levels correlate with fatal outcome in sepsis. Shock 2004; 22:309.
  47. Keane C, Jerkic M, Laffey JG. Stem Cell-based Therapies for Sepsis. Anesthesiology 2017; 127:1017.
  48. Venet F, Rimmelé T, Monneret G. Management of Sepsis-Induced Immunosuppression. Crit Care Clin 2018; 34:97.
  49. Arad G, Levy R, Hillman D, Kaempfer R. Superantigen antagonist protects against lethal shock and defines a new domain for T-cell activation. Nat Med 2000; 6:414.
  50. Borthwick EMJ, Hill CJ, Rabindranath KS, et al. High-volume haemofiltration for sepsis. Cochrane Database Syst Rev 2013; :CD008075.
  51. Honore PM, Jamez J, Wauthier M, et al. Prospective evaluation of short-term, high-volume isovolemic hemofiltration on the hemodynamic course and outcome in patients with intractable circulatory failure resulting from septic shock. Crit Care Med 2000; 28:3581.
  52. Cornejo R, Downey P, Castro R, et al. High-volume hemofiltration as salvage therapy in severe hyperdynamic septic shock. Intensive Care Med 2006; 32:713.
  53. Piccinni P, Dan M, Barbacini S, et al. Early isovolaemic haemofiltration in oliguric patients with septic shock. Intensive Care Med 2006; 32:80.
  54. Joannes-Boyau O, Honoré PM, Perez P, et al. High-volume versus standard-volume haemofiltration for septic shock patients with acute kidney injury (IVOIRE study): a multicentre randomized controlled trial. Intensive Care Med 2013; 39:1535.
  55. Borthwick EM, Hill CJ, Rabindranath KS, et al. High-volume haemofiltration for sepsis. Cochrane Database Syst Rev 2013; :CD008075.
  56. Cole L, Bellomo R, Hart G, et al. A phase II randomized, controlled trial of continuous hemofiltration in sepsis. Crit Care Med 2002; 30:100.
  57. Payen D, Mateo J, Cavaillon JM, et al. Impact of continuous venovenous hemofiltration on organ failure during the early phase of severe sepsis: a randomized controlled trial. Crit Care Med 2009; 37:803.
  58. Zarychanski R, Doucette S, Fergusson D, et al. Early intravenous unfractionated heparin and mortality in septic shock. Crit Care Med 2008; 36:2973.
  59. Jaimes F, De La Rosa G, Morales C, et al. Unfractioned heparin for treatment of sepsis: A randomized clinical trial (The HETRASE Study). Crit Care Med 2009; 37:1185.
  60. Safety of Heparin in Patients with Septic Shock. http://www.clinicaltrials.gov/ct2/show/NCT01234285 (Accessed on June 13, 2011).
  61. Yamakawa K, Aihara M, Ogura H, et al. Recombinant human soluble thrombomodulin in severe sepsis: a systematic review and meta-analysis. J Thromb Haemost 2015; 13:508.
  62. Hayakawa M, Kudo D, Saito S, et al. Antithrombin Supplementation and Mortality in Sepsis-Induced Disseminated Intravascular Coagulation: A Multicenter Retrospective Observational Study. Shock 2016; 46:623.
  63. Vincent JL, Francois B, Zabolotskikh I, et al. Effect of a Recombinant Human Soluble Thrombomodulin on Mortality in Patients With Sepsis-Associated Coagulopathy: The SCARLET Randomized Clinical Trial. JAMA 2019; 321:1993.
  64. Dépret F, Sitbon A, Soussi S, et al. Intravenous iloprost to recruit the microcirculation in septic shock patients? Intensive Care Med 2018; 44:121.
  65. Berthelsen RE, Ostrowski SR, Bestle MH, Johansson PI. Co-administration of iloprost and eptifibatide in septic shock (CO-ILEPSS)-a randomised, controlled, double-blind investigator-initiated trial investigating safety and efficacy. Crit Care 2019; 23:301.
  66. Legrand M, Oufella HA, De Backer D, et al. The I-MICRO trial, Ilomedin for treatment of septic shock with persistent microperfusion defects: a double-blind, randomized controlled trial-study protocol for a randomized controlled trial. Trials 2020; 21:601.
  67. Bestle MH, Clausen NE, Søe-Jensen P, et al. Efficacy and safety of iloprost in patients with septic shock-induced endotheliopathy-Protocol for the multicenter randomized, placebo-controlled, blinded, investigator-initiated trial. Acta Anaesthesiol Scand 2020; 64:705.
  68. Boeuf B, Gauvin F, Guerguerian AM, et al. Therapy of shock with naloxone: a meta-analysis. Crit Care Med 1998; 26:1910.
  69. Rock P, Silverman H, Plump D, et al. Efficacy and safety of naloxone in septic shock. Crit Care Med 1985; 13:28.
  70. Kirschenbaum LA, Aziz M, Astiz ME, et al. Influence of rheologic changes and platelet-neutrophil interactions on cell filtration in sepsis. Am J Respir Crit Care Med 2000; 161:1602.
  71. Kleinschmidt S, Wanner GA, Bussmann D, et al. Proinflammatory cytokine gene expression in whole blood from patients undergoing coronary artery bypass surgery and its modulation by pentoxifylline. Shock 1998; 9:12.
  72. Staubach KH, Schröder J, Stüber F, et al. Effect of pentoxifylline in severe sepsis: results of a randomized, double-blind, placebo-controlled study. Arch Surg 1998; 133:94.
  73. Papazian L, Roch A, Charles PE, et al. Effect of statin therapy on mortality in patients with ventilator-associated pneumonia: a randomized clinical trial. JAMA 2013; 310:1692.
  74. Hackam DG, Mamdani M, Li P, Redelmeier DA. Statins and sepsis in patients with cardiovascular disease: a population-based cohort analysis. Lancet 2006; 367:413.
  75. Thomsen RW, Hundborg HH, Johnsen SP, et al. Statin use and mortality within 180 days after bacteremia: a population-based cohort study. Crit Care Med 2006; 34:1080.
  76. Merx MW, Weber C. Statins: a preventive strike against sepsis in patients with cardiovascular disease? Lancet 2006; 367:372.
  77. Majumdar SR, McAlister FA, Eurich DT, et al. Statins and outcomes in patients admitted to hospital with community acquired pneumonia: population based prospective cohort study. BMJ 2006; 333:999.
  78. Kruger PS, Harward ML, Jones MA, et al. Continuation of statin therapy in patients with presumed infection: a randomized controlled trial. Am J Respir Crit Care Med 2011; 183:774.
  79. Yende S, Milbrandt EB, Kellum JA, et al. Understanding the potential role of statins in pneumonia and sepsis. Crit Care Med 2011; 39:1871.
  80. Thomas G, Hraiech S, Loundou A, et al. Statin therapy in critically-ill patients with severe sepsis: a review and meta-analysis of randomized clinical trials. Minerva Anestesiol 2015; 81:921.
  81. Deshpande A, Pasupuleti V, Rothberg MB. Statin therapy and mortality from sepsis: a meta-analysis of randomized trials. Am J Med 2015; 128:410.
  82. Dinglas VD, Hopkins RO, Wozniak AW, et al. One-year outcomes of rosuvastatin versus placebo in sepsis-associated acute respiratory distress syndrome: prospective follow-up of SAILS randomised trial. Thorax 2016; 71:401.
  83. Morelli A, Ertmer C, Westphal M, et al. Effect of heart rate control with esmolol on hemodynamic and clinical outcomes in patients with septic shock: a randomized clinical trial. JAMA 2013; 310:1683.
  84. Kakihana Y, Nishida O, Taniguchi T, et al. Efficacy and safety of landiolol, an ultra-short-acting β1-selective antagonist, for treatment of sepsis-related tachyarrhythmia (J-Land 3S): a multicentre, open-label, randomised controlled trial. Lancet Respir Med 2020; 8:863.
  85. Bedet A, Voiriot G, Ternacle J, et al. Heart Rate Control during Experimental Sepsis in Mice: Comparison of Ivabradine and β-Blockers. Anesthesiology 2020; 132:321.
  86. Whitehouse T, Hossain A, Perkins GD, et al. Landiolol and Organ Failure in Patients With Septic Shock: The STRESS-L Randomized Clinical Trial. JAMA 2023; 330:1641.
  87. Hasegawa D, Sato R, Prasitlumkum N, et al. Effect of Ultrashort-Acting β-Blockers on Mortality in Patients With Sepsis With Persistent Tachycardia Despite Initial Resuscitation: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Chest 2021; 159:2289.
  88. Marik PE, Khangoora V, Rivera R, et al. Hydrocortisone, Vitamin C, and Thiamine for the Treatment of Severe Sepsis and Septic Shock: A Retrospective Before-After Study. Chest 2017; 151:1229.
  89. Putzu A, Daems AM, Lopez-Delgado JC, et al. The Effect of Vitamin C on Clinical Outcome in Critically Ill Patients: A Systematic Review With Meta-Analysis of Randomized Controlled Trials. Crit Care Med 2019; 47:774.
  90. Fowler AA 3rd, Syed AA, Knowlson S, et al. Phase I safety trial of intravenous ascorbic acid in patients with severe sepsis. J Transl Med 2014; 12:32.
  91. Fowler AA 3rd, Truwit JD, Hite RD, et al. Effect of Vitamin C Infusion on Organ Failure and Biomarkers of Inflammation and Vascular Injury in Patients With Sepsis and Severe Acute Respiratory Failure: The CITRIS-ALI Randomized Clinical Trial. JAMA 2019; 322:1261.
  92. Fujii T, Luethi N, Young PJ, et al. Effect of Vitamin C, Hydrocortisone, and Thiamine vs Hydrocortisone Alone on Time Alive and Free of Vasopressor Support Among Patients With Septic Shock: The VITAMINS Randomized Clinical Trial. JAMA 2020; 323:423.
  93. Chang P, Liao Y, Guan J, et al. Combined Treatment With Hydrocortisone, Vitamin C, and Thiamine for Sepsis and Septic Shock: A Randomized Controlled Trial. Chest 2020; 158:174.
  94. Moskowitz A, Huang DT, Hou PC, et al. Effect of Ascorbic Acid, Corticosteroids, and Thiamine on Organ Injury in Septic Shock: The ACTS Randomized Clinical Trial. JAMA 2020; 324:642.
  95. Sevransky JE, Rothman RE, Hager DN, et al. Effect of Vitamin C, Thiamine, and Hydrocortisone on Ventilator- and Vasopressor-Free Days in Patients With Sepsis: The VICTAS Randomized Clinical Trial. JAMA 2021; 325:742.
  96. Lamontagne F, Masse MH, Menard J, et al. Intravenous Vitamin C in Adults with Sepsis in the Intensive Care Unit. N Engl J Med 2022; 386:2387.
  97. Sato R, Hasegawa D, Prasitlumkum N, et al. Effect of IV High-Dose Vitamin C on Mortality in Patients With Sepsis: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Crit Care Med 2021; 49:2121.
  98. Assouline B, Faivre A, Verissimo T, et al. Thiamine, Ascorbic Acid, and Hydrocortisone As a Metabolic Resuscitation Cocktail in Sepsis: A Meta-Analysis of Randomized Controlled Trials With Trial Sequential Analysis. Crit Care Med 2021; 49:2112.
  99. Agarwal A, Basmaji J, Fernando SM, et al. Administration of Parenteral Vitamin C in Patients With Severe Infection: Protocol for a Systematic Review and Meta-analysis. JMIR Res Protoc 2022; 11:e33989.
  100. MacGarvey NC, Suliman HB, Bartz RR, et al. Activation of mitochondrial biogenesis by heme oxygenase-1-mediated NF-E2-related factor-2 induction rescues mice from lethal Staphylococcus aureus sepsis. Am J Respir Crit Care Med 2012; 185:851.
  101. Martí-Carvajal AJ, Solà I, Lathyris D, Cardona AF. Human recombinant activated protein C for severe sepsis. Cochrane Database Syst Rev 2012; :CD004388.
  102. Bernard GR, Vincent JL, Laterre PF, et al. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med 2001; 344:699.
  103. FDA Drug Safety Communication: Voluntary Market Withdrawal of Xigris [(Drotrecogin alfa (activated)] due to failure to show a survival benefit. http://www.fda.gov/Drugs/DrugSafety/ucm277114.htm (Accessed on October 25, 2011).
  104. McCloskey RV, Straube RC, Sanders C, et al. Treatment of septic shock with human monoclonal antibody HA-1A. A randomized, double-blind, placebo-controlled trial. CHESS Trial Study Group. Ann Intern Med 1994; 121:1.
  105. 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.
  106. Albertson TE, Panacek EA, MacArthur RD, et al. Multicenter evaluation of a human monoclonal antibody to Enterobacteriaceae common antigen in patients with Gram-negative sepsis. Crit Care Med 2003; 31:419.
  107. Heemskerk S, Masereeuw R, Moesker O, et al. Alkaline phosphatase treatment improves renal function in severe sepsis or septic shock patients. Crit Care Med 2009; 37:417.
  108. Root RK, Lodato RF, Patrick W, et al. 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.
  109. Abraham E, Wunderink R, Silverman H, 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.
  110. Cohen J, Carlet J. INTERSEPT: an international, multicenter, placebo-controlled trial of monoclonal antibody to human tumor necrosis factor-alpha in patients with sepsis. International Sepsis Trial Study Group. Crit Care Med 1996; 24:1431.
  111. Abraham E, Anzueto A, Gutierrez G, et al. Double-blind randomised controlled trial of monoclonal antibody to human tumour necrosis factor in treatment of septic shock. NORASEPT II Study Group. Lancet 1998; 351:929.
  112. Abraham E, Laterre PF, Garbino J, et al. 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.
  113. Opal SM, Kessler CM, Roemisch J, Knaub S. Antithrombin, heparin, and heparan sulfate. Crit Care Med 2002; 30:S325.
  114. Warren BL, Eid A, Singer P, et al. Caring for the critically ill patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial. JAMA 2001; 286:1869.
  115. Afshari A, Wetterslev J, Brok J, Møller A. Antithrombin III in critically ill patients: systematic review with meta-analysis and trial sequential analysis. BMJ 2007; 335:1248.
  116. Baudo F, Caimi TM, de Cataldo F, 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.
  117. Wiedermann CJ, Hoffmann JN, Juers M, et al. High-dose antithrombin III in the treatment of severe sepsis in patients with a high risk of death: efficacy and safety. Crit Care Med 2006; 34:285.
  118. Allingstrup M, Afshari A. Selenium supplementation for critically ill adults. Cochrane Database Syst Rev 2015; :CD003703.
  119. Allingstrup M, Wetterslev J, Ravn FB, et al. Antithrombin III for critically ill patients: a systematic review with meta-analysis and trial sequential analysis. Intensive Care Med 2016; 42:505.
  120. Allingstrup M, Wetterslev J, Ravn FB, et al. Antithrombin III for critically ill patients. Cochrane Database Syst Rev 2016; 2:CD005370.
  121. Abraham E, Reinhart K, Svoboda P, 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.
  122. Abraham E, Reinhart K, Opal S, et al. Efficacy and safety of tifacogin (recombinant tissue factor pathway inhibitor) in severe sepsis: a randomized controlled trial. JAMA 2003; 290:238.
  123. Bernard GR, Wheeler AP, Russell JA, et al. The effects of ibuprofen on the physiology and survival of patients with sepsis. The Ibuprofen in Sepsis Study Group. N Engl J Med 1997; 336:912.
  124. Spapen HD, Diltoer MW, Nguyen DN, et al. Effects of N-acetylcysteine on microalbuminuria and organ failure in acute severe sepsis: results of a pilot study. Chest 2005; 127:1413.
  125. Szakmany T, Hauser B, Radermacher P. N-acetylcysteine for sepsis and systemic inflammatory response in adults. Cochrane Database Syst Rev 2012; :CD006616.
  126. Petros A, Lamb G, Leone A, et al. Effects of a nitric oxide synthase inhibitor in humans with septic shock. Cardiovasc Res 1994; 28:34.
  127. Lorente JA, Landín L, De Pablo R, et al. L-arginine pathway in the sepsis syndrome. Crit Care Med 1993; 21:1287.
  128. Avontuur JA, Boomsma F, van den Meiracker AH, et al. Endothelin-1 and blood pressure after inhibition of nitric oxide synthesis in human septic shock. Circulation 1999; 99:271.
  129. Grover R, Zaccardelli D, Colice G, et al. An open-label dose escalation study of the nitric oxide synthase inhibitor, N(G)-methyl-L-arginine hydrochloride (546C88), in patients with septic shock. Glaxo Wellcome International Septic Shock Study Group. Crit Care Med 1999; 27:913.
  130. Schoonover LL, Stewart AS, Clifton GD. Hemodynamic and cardiovascular effects of nitric oxide modulation in the therapy of septic shock. Pharmacotherapy 2000; 20:1184.
  131. López A, Lorente JA, Steingrub J, 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.
  132. Vincent JL, Privalle CT, Singer M, et al. Multicenter, randomized, placebo-controlled phase III study of pyridoxalated hemoglobin polyoxyethylene in distributive shock (PHOENIX). Crit Care Med 2015; 43:57.
  133. Fein AM, Bernard GR, Criner GJ, 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.
  134. Takala J, Ruokonen E, Webster NR, et al. Increased mortality associated with growth hormone treatment in critically ill adults. N Engl J Med 1999; 341:785.
  135. Angstwurm MW, Engelmann L, Zimmermann T, et al. Selenium in Intensive Care (SIC): results of a prospective randomized, placebo-controlled, multiple-center study in patients with severe systemic inflammatory response syndrome, sepsis, and septic shock. Crit Care Med 2007; 35:118.
  136. Safety and Efficacy of Recombinant Human Lactoferrin to Treat Severe Sepsis. http://www.clinicaltrials.gov/ct2/show/NCT00630656 (Accessed on June 12, 2011).
  137. Talactoferrin alfa reduces mortality in severe sepsis: Results of a Phase 2 randomized, placebo-controlled, double-blind study. http://www.thoracic.org/newsroom/press-releases/resources/late-breaking-abstract-pdf/talactoferrin.pdf (Accessed on June 12, 2011).
  138. Guntupalli KK, Dean NC, Morris PE, et al . Talactoferrin alfa reduces mortality in severe. Am J Respir Crit Care Med 2010; 181:A6768.
  139. Safety and Efficacy of Talactoferrin Alfa in Patients with Severe Sepsis (OASIS). http://www.clinicaltrials.gov/ct2/show/NCT01273779 (Accessed on June 12, 2011).
  140. Vincent JL, Marshall JC, Dellinger RP, et al. Talactoferrin in Severe Sepsis: Results From the Phase II/III Oral tAlactoferrin in Severe sepsIS Trial. Crit Care Med 2015; 43:1832.
  141. Leaf DE, Raed A, Donnino MW, et al. Randomized controlled trial of calcitriol in severe sepsis. Am J Respir Crit Care Med 2014; 190:533.
  142. Gordon AC, Perkins GD, Singer M, et al. Levosimendan for the Prevention of Acute Organ Dysfunction in Sepsis. N Engl J Med 2016; 375:1638.
  143. Itenov TS, Johansen ME, Bestle M, et al. Induced hypothermia in patients with septic shock and respiratory failure (CASS): a randomised, controlled, open-label trial. Lancet Respir Med 2018; 6:183.
  144. Asfar P, Schortgen F, Boisramé-Helms J, et al. Hyperoxia and hypertonic saline in patients with septic shock (HYPERS2S): a two-by-two factorial, multicentre, randomised, clinical trial. Lancet Respir Med 2017; 5:180.
  145. National Heart, Lung, and Blood Institute PETAL Clinical Trials Network, Ginde AA, Brower RG, et al. Early High-Dose Vitamin D3 for Critically Ill, Vitamin D-Deficient Patients. N Engl J Med 2019; 381:2529.
Topic 1631 Version 63.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟