could biomarkers direct therapy for the septic...

62
JPET #230797 1 Could Biomarkers Direct Therapy for the Septic Patient? Clark R. Sims, Trung C. Nguyen, and Philip R. Mayeux Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.R.S., P.R.M.), and Department of Pediatrics, Section of Critical Care Medicine, Baylor College of Medicine/Texas Children’s Hospital, Houston, Texas (T.C.N) This article has not been copyedited and formatted. The final version may differ from this version. JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797 at ASPET Journals on July 12, 2018 jpet.aspetjournals.org Downloaded from

Upload: lamduong

Post on 24-Jun-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

JPET #230797

1

Could Biomarkers Direct Therapy for the Septic Patient?

Clark R. Sims, Trung C. Nguyen, and Philip R. Mayeux

Department of Pharmacology and Toxicology, University of Arkansas for Medical

Sciences, Little Rock, Arkansas (C.R.S., P.R.M.), and Department of Pediatrics, Section

of Critical Care Medicine, Baylor College of Medicine/Texas Children’s Hospital,

Houston, Texas (T.C.N)

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

2

Running Title: (60 characters): Biomarker-Directed Therapy for the Septic Patient?

Corresponding Author:

Philip R. Mayeux, PhD

4301 West Markham Street, #611

Little Rock, AR 72205

501-686-8895

FAX: 501-686-5521

[email protected]

Number of text pages: 23

Number of tables: 0

Number of figures: 2

Number of references: 196

Number of words in Abstract: 190

Number of words in Introduction: 981

Number of words in Discussion: N/A

Section Assignment: Minireviews

Abbreviations: ADAMTS-13, a disintegrin and metalloproteinase with a thrombospondin

type 1 motif, member 13; Ang, angiopoietin; CARS, compensatory anti-inflammatory

response syndrome; cfu, colony forming units; CLP, cecal ligation and puncture; CRP,

C-reactive protein; DAMPs, damage-associated molecular patterns; DIC,

disseminated intravascular coagulation; EEG, electroencephalography; GM-CSF,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

3

granulocyte macrophage colony-stimulating factor; HLA-DR, human leukocyte antigen

- antigen D related; ICAM-1, intercellular adhesion molecule 1; ICU, intensive care

unit; IL, interleukin; KIM-1, kidney injury molecule 1; LPS, lipopolysaccharide; MMP-8,

matrix metalloproteinase-8; MODS, multiple organ dysfunction syndrome; MRI,

magnetic resonance imaging; NAG, N-acetyl-β-(D)-glucosaminidase; NGAL,

neutrophil gelatinase-associated lipocalin; NIRS, near-infrared spectroscopy; PAMPs,

pathogen-associated molecular patterns; PCT, procalcitonin; PD-1, programmed cell

death 1; S1P, sphingosine-1-phosphate; SIRS, systemic inflammatory response

syndrome; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor α; VCAM-1,

vascular cell adhesion molecule 1; VWF, von Willebrand factor

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

4

Abstract

Sepsis is a serious medical condition caused by a severe systemic inflammatory

response to a bacterial, fungal, or viral infection that most commonly affects neonates

and the elderly. Advances in understanding the pathophysiology of sepsis have resulted

in guidelines for care that have helped reduce the risk of dying from sepsis for both

children and older adults. Still, over the past three decades, a large number of clinical

trials have been undertaken to evaluate pharmacological agents for sepsis.

Unfortunately, all of these trials have failed, with the use of some agents even shown to

be harmful. One key issue in these trials was the heterogeneity of the patient population

that participated. What has emerged is the need to target therapeutic interventions to

the specific patient’s underlying pathophysiological processes, rather than looking for a

universal therapy that would be effective in a “typical” septic patient who does not exist.

This review supports the concept that identification of the right biomarkers that can

direct therapy and provide timely feedback on its effectiveness will enable critical care

physicians to decrease mortality of patients with sepsis and improve the quality of life of

survivors.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

5

Introduction

Sepsis is a serious medical condition caused by a severe systemic inflammatory

response to a bacterial, fungal, or viral infection (Martin, 2012; Dellinger et al., 2013). As

sepsis worsens, organ dysfunction (severe sepsis) or severe sepsis plus hypotension

(septic shock) can develop (Dellinger et al., 2013). Sepsis affects approximately

700,000 people annually in the U.S. alone (Chang et al., 2015). Over 25% of these

patients die before leaving the hospital, making sepsis the 9th leading cause of death

overall (Heron, 2015). In addition, relative to the total number of cases, sepsis is the

most costly medical condition, especially given that over 20% of sepsis survivors are

readmitted within 30 days and nearly 30% of these patients have recurrent sepsis (Torio

and Andrews, 2013; Chang et al., 2015). The estimated annual health care burden is

over $20 billion in the U.S. alone (Torio and Andrews, 2013).

While sepsis affects people of all ages, the most commonly affected populations

are neonates, due to an immature immune system and still developing organs, and the

elderly, due to chronic and comorbid conditions. In the U.S., sepsis is the 6th and 7th

leading cause of death in neonates and infants, respectively, and the 10th leading cause

of death in patients age 65 and older (Heron, 2015). Mortality rates for children with

septic shock range from 9% to 25% (Hartman et al., 2013; Weiss et al., 2015), and for

older adults mortality rates are as high as 45% (Daviaud et al., 2015). Although the

absolute number of deaths from sepsis is increasing due to increasing incidence (Martin,

2012; Gaieski et al., 2013), advances in the understanding of the pathophysiology of

sepsis have reduced the risk of dying from sepsis for both children (Kissoon et al., 2011;

Ruth et al., 2014) and older adults (Stevenson et al., 2014).

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

6

The pathophysiology of sepsis is very complex and still poorly understood.

Initially, local activation of the innate immune response by pathogen-associated

molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs)

results in a “cytokine storm” (Wang and Ma, 2008; Gustot, 2011; Laszlo et al., 2015). If

the inflammatory response becomes systemic it manifests as the systemic inflammatory

response syndrome (SIRS, (Conference, 1992)). Patients who survive the initial

hyperinflammatory state may progress to a hypoinflammatory state and develop the

compensatory anti-inflammatory response syndrome (CARS, (Bone, 1996; Ward et al.,

2008)), which increases the susceptibility to recurrence of the primary infection or a

secondary acquired infection. Disruption of the balance between these inflammatory

and anti-inflammatory responses in sepsis leads to multiple organ dysfunction

syndrome (MODS), which significantly increases mortality (Wang and Ma, 2008; Gustot,

2011; Boomer et al., 2014).

Mortality from sepsis has significantly decreased over the years because of early

recognition and resuscitation, as documented in the “Surviving Sepsis Campaign”

(Dellinger et al., 2013). Following recommendations from this document, current

therapies are mostly supportive care. Initial treatment includes broad-spectrum

antibiotics and then fluid resuscitation if necessary. As sepsis severity increases,

therapy may require administration of vasopressors, use of mechanical respiratory

support, and/or renal replacement therapy (Dellinger et al., 2013). Over the past three

decades, there have been over 60 large Phase II and III randomized, controlled clinical

trials of single pharmacological agents for sepsis, all of which have failed, with some

agents even found to be harmful (Fink and Warren, 2014). Reflecting on these failed

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

7

trials, many leaders in the field believe that the heterogeneity of the patient population

that participated in these trials is a key issue (Cohen et al., 2015). To address this issue,

various critical care societies are currently revising the definition of the sepsis syndrome

to refine the inclusion criteria for participants in future sepsis trials. One common theme

is surfacing: infection plus organ dysfunction should be considered as inclusion criteria

rather than just the current two or more SIRS criteria (Vincent et al., 2013; Cohen et al.,

2015; Drewry and Hotchkiss, 2015; Kaukonen et al., 2015). In addition it has been

suggested that there is a need to target therapeutic interventions according to the

specific patient’s underlying pathophysiological processes, rather than looking for a

universal therapy that would be effective in the "typical” patient population with sepsis

(Boomer et al., 2014; Christaki and Giamarellos-Bourboulis, 2014).

The complex pathophysiology of sepsis and the ineffectiveness of current

targeted therapies suggest that treatments guided by biomarkers could provide a new

therapeutic strategy. A biomarker, as defined by an NIH working group, is “a

characteristic that is objectively measured and evaluated as an indicator of normal

biological processes, pathogenic processes, or pharmacologic responses to a

therapeutic intervention” (Group, 2001). There are general and overlapping categories

of biomarkers, which have been reviewed and discussed in detail elsewhere (Marshall

et al., 2009; Kaplan and Wong, 2011; Dupuy et al., 2013; Sandquist and Wong, 2014).

Biomarker categories include 1) screening biomarkers to identify patients at increased

risk and inform on prophylactic interventions, 2) diagnostic biomarkers to establish the

presence or absence of disease and inform treatment decisions, 3) stratification

biomarkers to stage severity and outcome of disease and help identify subgroups of

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

8

patients who may benefit or be harmed by therapy, 4) monitoring biomarkers to inform

on the effectiveness of a therapy, and 5) surrogate biomarkers to inform on disease

progression and potential outcomes of therapy. A number of excellent recent reviews

have been published on the potential diagnostic and prognostic value of biomarkers in

sepsis (Pierrakos and Vincent, 2010; Sandquist and Wong, 2014; Biron et al., 2015;

Cohen et al., 2015). Therefore, this review will not focus on the current status of sepsis

biomarkers. Rather, the goal of this review is to support the concept that the appropriate

biomarkers could be used to direct targeted therapies (Fig. 1). For example, diagnostic

biomarkers could direct therapies such as those targeting immune status, endothelial

injury, coagulation, and microcirculatory failure; and stratification biomarkers could

inform the decision to rapidly employ renal replacement therapy or mechanical

ventilation.

Patient Heterogeneity

Both the incidence and the outcome of sepsis are greatly influenced by patient

heterogeneity including patient-specific factors such as age, comorbid medical

conditions, and genetic predispositions. The very young (<1 year) and the elderly (>65

years) account for 76% of all deaths due to sepsis in the U.S. (Heron, 2015). Premature

and very-low-birth-weight infants suffer the highest mortality (Heron, 2015), which may

be due to increased hemodynamic fragility, low cardiac output, and organ hypoperfusion,

as well as a decreased ability to mount an appropriate immune response (Adkins et al.,

2004; Aneja and Carcillo, 2011; Shah and Padbury, 2014). Likewise, in elderly patients,

debilitated physiological processes, reflected in higher incidences of congestive heart

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

9

failure, coronary artery disease, diabetes, hypertension, and chronic obstructive

pulmonary disease, underlie a 13-fold higher relative risk for the development of sepsis

(Martin et al., 2006) and an increased mortality from sepsis (Boss and Seegmiller, 1981).

These physiological differences in sepsis with respect to age and comorbid medical

conditions suggest that treatments will need to be based on the specific conditions of

the individual patient.

The presence of MODS is an important factor in the treatment of sepsis. While

there is substantial variation in the percentages of patients with any specific organ

involvement, pulmonary, renal, cardiovascular, hepatic, neurological, and hematological

dysfunction may result from primary and secondary causes (Gaieski et al., 2013;

Iskander et al., 2013; Stubbs et al., 2013; Ziaja, 2013; Fiusa et al., 2015). In all

populations, the mortality rate of sepsis greatly increases as multiple organ systems fail

(Gaieski et al., 2013; Iskander et al., 2013; Romanovsky et al., 2013).

Because of the inherent heterogeneity of patients enrolled in clinical trials, a level

of heterogeneity in the therapeutic response should be expected. Most important,

however, while some patients will benefit from the intervention, others could be harmed

(Marshall, 2014). To demonstrate overall efficacy of a treatment, studying larger

numbers of patients to increase statistical power is required in trial design, but this will

increase costs and may still not uncover efficacy in the general population for the

reasons just described. Early-phase sepsis trials should focus on identifying an

appropriate study population and use more specific inclusion criteria to limit trials to the

target populations suggested by preclinical studies. This approach could reduce

heterogeneity and increase the likelihood of demonstrating efficacy.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

10

Biomarkers

Validation of biomarkers requires an extensive amount of research and

resources. A limitation of biomarker research in human sepsis is that there are rarely

data on preillness levels of a biomarker. Because many septic patients have comorbid

chronic inflammatory conditions such as coronary artery disease and diabetes,

biomarkers alone are unlikely to accurately inform on the severity of sepsis (Ginde et al.,

2014) but may inform on possible outcomes (Kojic et al., 2015). For example, the ideal

biomarker could identify those patients at risk for specific organ dysfunction and thereby

help direct more targeted therapy. In addition, gene expression patterns could lead to

candidate serum protein markers to predict organ-specific injury (Basu et al., 2011).

Given the complex nature of sepsis, achieving this goal will likely require monitoring

multiple biomarkers through the course of the disease and the course of therapy (Wong

et al., 2015a). Biomarkers also depend on the specific pathogen, or often pathogens,

causing sepsis. Consequently, there is a need to accurately and quickly distinguish

sepsis from noninfectious SIRS. These are all areas where proteomics and genomics

(Cao and Robinson, 2014; Cao et al., 2014; Christaki and Giamarellos-Bourboulis,

2014; DeCoux et al., 2015) and rapid identification of positive blood cultures (Bhatti et

al., 2014b; Bhatti et al., 2014a) could become very helpful.

Inflammatory Biomarkers

Should the goal of therapy for septic patients be to decrease inflammation or

boost host immunity? This is an extremely important and difficult question with no

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

11

simple answer. Hotchkiss and Sherwood (Hotchkiss and Sherwood, 2015) have

published a recent commentary on this issue. However, biomarkers have the potential

to help critical care physicians make important clinical decisions regarding

immunotherapy (Hotchkiss et al., 2013b).

In general, blood levels of soluble adhesion molecules such as E-, L- and P-

selectin, ICAM-1, and VCAM-1 correlate with the presence of sepsis in neonates,

children, and adults; however, there are age-related differences in the absolute levels,

and it is still unclear whether changes in levels might correlate positively or negatively

with outcomes (Zonneveld et al., 2014). C-reactive protein (CRP) is an acute-phase

protein released by the liver. It is used as an inflammatory biomarker but cannot

distinguish between infection and noninfectious diseases (Kojic et al., 2015).

Procalcitonin (PCT) is a propeptide of calcitonin released in the blood in response

bacteria-associated proinflammatory mediators and inflammatory cytokines. Its use as a

specific biomarker for sepsis has come under question (Talan, 2015). CRP and PCT

may be more general biomarkers of moderate to severe inflammation but could be

useful for informing on the response to treatment (Wacker et al., 2013; Rule et al., 2015).

However, the levels of PCT and CRP in combination may be able discriminate bacterial

sepsis from other causes of SIRS (Han et al., 2015).

Cytokines including tumor necrosis factor α (TNF-α) and interleukin (IL)-1, IL-6,

IL-8, IL-27, and others have been evaluated individually and in combination as potential

biomarkers for screening and diagnosis (Xiao et al., 2015). Although cytokine levels are

relatively easily and rapidly measured with multiplex assays (Mera et al., 2011), the

cost–benefit of monitoring them has yet to be established. Serial cytokine

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

12

measurements would be required to inform on immune status; however, this would

require a thorough understanding of the complex kinetics of synthesis, release, and

clearance. Moreover, elderly septic patients present with generally higher levels of

inflammatory biomarkers due to comorbid conditions (Ginde et al., 2014), and because

pre-sepsis levels would be unknown, interpreting changes (or not) in cytokine levels

would be extremely difficulty. These issues may explain, at least in part, why clinical

trials targeting cytokines failed to demonstrate efficacy; use of some agents was even

harmful (Fink and Warren, 2014).

Septic patients in the immunosuppressive phase could potentially benefit from

immune-enhancing therapies, but these patients must first be identified. Potential

biomarkers, which have been suggested for this purpose, include decreased monocyte

human leukocyte antigen – antigen D related (HLA-DR) expression for monocyte

unresponsiveness, increased programmed cell death 1 (PD-1) expression for T cell

exhaustion, lymphopenia, and increased percentage of regulatory T cells, among others

(reviewed in (Hotchkiss et al., 2013b) and (Boomer et al., 2014)). The use of specific,

biomarker-directed immunotherapies such as granulocyte macrophage colony

stimulating factor (GM-CSF) to stimulate mature myeloid cell expansion in patients with

granulocytopenia (Mathias et al., 2015), and IL-7 to promote lymphocyte expansion in

patients with lymphopenia (Unsinger et al., 2010; Hutchins et al., 2014) are examples of

new therapeutic strategies with the potential to advance the treatment of the

immunosuppressed septic patient (Hotchkiss et al., 2013a; Boomer et al., 2014).

Endothelial Biomarkers

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

13

Sepsis is associated with severe injury to vascular endothelial cells. Injury to the

endothelium leads to dysregulation of vascular tone, hemostasis, and the permeability

barrier. Microvascular leakage is a hallmark of sepsis and results from damage to the

endothelial cell barrier lining the microvasculature. Identifying possible targeted therapy

to stabilize and even promote repair of the endothelial barrier is a very active area of

research (Darwish and Liles, 2013). Syndecan-1 and heparan sulfate are potential

biomarkers of damage to the endothelial glycocalyx, the antiadhesive and anticoagulant

surface of the endothelial cell (Schmidt et al., 2012) and, if confirmed as such, could be

used as diagnostic and monitoring biomarkers to direct and evaluate anticoagulant

therapy (Ostrowski et al., 2015; Yini et al., 2015).

A number of signaling pathways regulating vascular permeability appear to be

disrupted during sepsis. One such system is the endothelial-specific angiopoietin growth

factor (Ang) and its Tie-2 tyrosine kinase receptor (Maisonpierre et al., 1997; Ziegler et

al., 2013). Plasma levels of Ang-1, Ang-2, and Tie-2 have been show to predict the

onset of acute lung injury (Agrawal et al., 2013), 28-day mortality in septic adults (Fang

et al., 2015), and sepsis severity in children (Wang et al., 2014). If verified, levels of

Ang-1, Ang-2, and Tie-2 in septic patients may inform on the effectiveness and outcome

of therapy affecting the endothelium, such as the use of statins (Ghosh et al., 2015).

Another system is the sphingosine-1-phosphate (S1P) signaling pathway, a key

regulator of endothelial stability and vascular permeability (Lee et al., 1999; Wang and

Dudek, 2009). Serum levels of S1P in septic patients are decreased and inversely

associated with disease severity (Winkler et al., 2015). If S1P levels can be verified as a

biomarker of microvascular injury, S1P could be used to screen, diagnose, and/or

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

14

stratify patients and direct the use of S1P receptor 1 agonists, shown to protect the

endothelial glycocalyx in vitro (Zeng et al., 2014) and to heal the renal microcirculation

in a murine model of sepsis (Wang et al., 2015b).

During sepsis, injured endothelial cells release microparticles containing proteins

and lipids that can augment thrombosis, inflammation, and microvascular injury.

Understanding the role of microparticles in the progression of sepsis and organ injury is

also a very active area of research. Souza and colleagues (Souza et al., 2015) have

published an excellent recent review. As a potential biomarker, microparticles may help

stratify patients and inform on the potential outcomes of therapy targeting the

endothelium.

Coagulation Biomarkers

Sepsis with its associated inflammation induces a state of dysregulated

hemostasis. Up to 30–50% of septic patients have clinical overt disseminated

intravascular coagulation (DIC), which leads to dissemination of fibrin-rich microvascular

thromboses and contributes to MODS (Levi and van der Poll, 2013). Many specific

monotherapeutic agents that interfere with dysregulated hemostasis in septic patients

have been tried without success in large randomized, controlled trials. These agents

include heparin, antithrombin III, recombinant tissue pathway inhibitors, recombinant

activated protein C, and recombinant soluble thrombomodulin (Warren et al., 2001;

Abraham et al., 2003; Nadel et al., 2007; Saito et al., 2007; Jaimes et al., 2009; Ranieri

et al., 2012). These trials, however, enrolled heterogeneous septic patients and did not

use coagulation biomarkers as titratable therapeutic endpoints. On reflection, the failure

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

15

of these trials suggests that, as described earlier, study design and patient selection

rather than the specific agents may be at fault (Cohen et al., 2015). Currently, clinicians

rely on point-of-care global measurement of the hemostatic pathway including

prothrombin time, activated partial thromboplastin time, the fibrin degradation product D-

dimer, and platelet count to provide blood product support for hemostasis (Wada et al.,

2013). For future clinical trials, in addition to the point-of-care assays, measuring,

replacing, and normalizing the activities of heparin, antithrombin III, tissue factor,

activated protein C, and/or thrombomodulin in septic patients with a defined severe

coagulopathy perhaps might lead to better outcomes.

Sepsis-induced thrombotic microangiopathies driven by either von Willebrand

factor (VWF)/platelets and/or complement pathways will lead to dissemination of

VWF/platelet-rich microvascular thromboses and MODS (Nguyen et al., 2014;

Charchaflieh et al., 2015). Measuring VWF, the presence of ultralarge VWF multimers,

and ADAMTS-13 (VWF-cleaving protease) and directing therapies to normalize their

activities have been shown to improve outcome in children with sepsis-induced

thrombocytopenia-associated multiple organ failure (Nguyen et al., 2008; Fortenberry et

al., 2012). Low levels of ADAMTS-13, high VWF activity, and the presence of ultralarge

VWF multimers have been associated with worse outcomes in septic adults (Martin et

al., 2007; Bockmeyer et al., 2008). Sepsis-induced complement pathway dysregulation

leading to DIC and thrombotic microangiopathies has been demonstrated in human

clinical studies and animal models (Charchaflieh et al., 2015; Zhao et al., 2015).

Eculizumab, a monoclonal antibody against complement C5, has been shown to

improve outcome in severe Escherichia coli–induced hemolytic uremic syndrome, which

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

16

leads to thrombotic microangiopathy and MODS (Lapeyraque et al., 2011; Noris et al.,

2012). Research is needed to prove whether other types of infection-induced thrombotic

microangiopathy and MODS can be ameliorated by measuring and normalizing

complement activities.

Physiological Biomarkers

Biomarkers are generally assumed to mean small molecules. However,

physiological biomarkers are the most rapidly determined indices of organ dysfunction

and can often be measured at the bedside. The complex pathophysiology of sepsis can

have a major impact on the translation of experimental therapies to clinical practice.

This is one of the key reasons that animal models of sepsis have had, thus far, a limited

impact on therapy in humans (Dyson and Singer, 2009; Marshall, 2014; Efron et al.,

2015). Changes in cardiac output, organ perfusion, drug delivery to therapeutic target,

and clearance of the drug are dynamic and often unpredictable in septic patients.

Physiological biomarkers could be of great help in interpreting successes, failures, and

undesirable outcomes in sepsis clinical trials.

The application of transthoracic echocardiography with speckle-tracking

technology to detect subclinical myocardial dysfunction may have value in directing

cardioprotective strategies, even if the ejection fraction remains normal (Shahul et al.,

2015). This type of monitoring may also help in establishing prognosis in septic shock

patients because, despite a normal ejection fraction, severe right ventricular wall

longitudinal strain dysfunction is associated with a high rate of mortality in patients with

severe sepsis and septic shock (Orde et al., 2014). There is a pressing need for

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

17

biomarkers to screen for sepsis-induced cardiomyopathy. Extracellular histones may

emerge as such a biomarker (Alhamdi et al., 2015; Kalbitz et al., 2015).

There is also a need for minimally invasive techniques that permit direct

physiological measurements of microvascular flow and organ perfusion in critically ill

patients. One of the most routine measurements made in septic patients is blood lactate

concentration. Lactate production is a protective response to allow cellular energy

production to continue when tissue oxygen supply is inadequate for aerobic metabolism

(Suetrong and Walley, 2015). Serum lactate levels remain a strong, independent

predictor of lung injury (Mikkelsen et al., 2013). Elevated lactate levels are highly

associated with in-hospital mortality and can be used to stratify patient risk (Casserly et

al., 2015). Use of lactate as a biomarker to direct management of the microcirculation

during the later stages of sepsis can be complicated by changes in lactate clearance

(Chertoff et al., 2015). Better understanding of the relationships between lactate levels

and actual status of the microcirculation remains a critical need (De Backer et al., 2013).

Organ-specific microcirculation can be measured in animal models of

experimental sepsis and used to evaluate the therapeutic potential of new approaches

to improve organ perfusion (Dear et al., 2005; Wu et al., 2007; Holthoff et al., 2012; Fink

et al., 2013; Taccone et al., 2014). As therapeutic strategies emerge to target

microvascular dysfunction, physiological biomarkers measureable at the bedside could

assist in directing therapy and confirming efficacy (or revealing harmfulness) (Matejovic

et al., 2015). Cine phase-contrast MRI and contrast-enhanced ultrasound can be

performed on critically ill septic patients to monitor blood flow in the kidney (Schneider

et al., 2013). For example, cine phase-contrast MRI revealed that renal blood flow was

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

18

consistently reduced as a fraction of cardiac output in septic patients with established

acute kidney injury (Prowle et al., 2012). Although these minimally invasive techniques

for monitoring organ perfusion are still in the validation phase, their use could ultimately

guide the choice of therapy and evaluate effectiveness. For example, changes in renal

perfusion paired with renal injury biomarkers such as neutrophil gelatinase-associated

lipocalin (NGAL), kidney injury molecule 1 (KIM-1), N-acetyl-β-(D)-glucosaminidase

(NAG), and matrix metalloproteinase-8 (MMP-8) (Liangos et al., 2007; Basu et al., 2011;

Wang et al., 2015a) could direct the implementation of renal replacement therapies

(hemofiltration and/or dialysis) that could improve outcomes in some patients; however,

this approach is not without risk (Elseviers et al., 2010; Sun et al., 2014; Prowle and

Davenport, 2015; Wald et al., 2015).

Orthogonal polarization spectral imaging and side-stream dark-field imaging

techniques can be used at the bedside to obtain real-time measurements of the

sublingual microcirculation in humans with sepsis (De Backer et al., 2012). Even though

changes in the sublingual microcirculation cannot predict the status of other organ-

specific microvascular beds, there is an excellent correlation between decreases in the

proportion of perfused small sublingual vessels in early sepsis and decreased survival

rates (De Backer et al., 2013; De Backer et al., 2014). These techniques could be used

as diagnostic and monitoring physiological biomarkers to direct therapy targeting the

vasculature, as suggested in animal models of sepsis (Holthoff et al., 2013; Wang et al.,

2015b).

Sepsis-associated encephalopathy with cognitive impairment is a common

complication of sepsis that places a substantial burden on survivors of sepsis, their

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

19

families, and their caregivers (Iwashyna et al., 2010). While the pathophysiology is still

poorly understood, neuroinflammatory and ischemic process are likely contributors to

alterations in the blood–brain barrier, release of neurotoxic mediators, and microglial

activation (Adam et al., 2013). Changes in standard electroencephalogram (EEG) are

associated with increased mortality and delirium in septic ICU patients (Azabou et al.,

2015). Near-infrared spectroscopy (NIRS) is being evaluated in high-risk septic patients

as a method to monitor brain tissue oxygenation to predict the development of acute

neurological dysfunction (Wood et al., (in press)). Interest is growing in identifying

biomarkers for sepsis-induced neurological injury (Bersani et al., 2015), especially

because animal models are revealing long-term brain alterations that affect learning and

memory and persist into adulthood (Comim et al., 2015; Gao et al., 2015). Targeting

neurotoxic inflammatory pathways (Gao et al., 2015) may emerge as an approach that

could be guided by specific small-molecule and physiological biomarkers.

Accumulating data suggest that during sepsis intracellular redox processes result

in increased production of superoxide, hydrogen peroxide, and peroxynitrite, especially

in mitochondria (Mayeux and Macmillan-Crow, 2012; Duran-Bedolla et al., 2014). While

there are studies to suggest that mitochondrial injury occurs in septic patients (Jeger et

al., 2013), the notion that mitochondrial injury is a cause of organ dysfunction is

controversial (Fink, 2015). Nonetheless, if there were rapidly measurable mitochondrial

injury biomarkers, they could be used to direct mitochondria-targeted antioxidants, such

as ubiquinone (MitoQ), vitamin E (MitoVit E), and piperidine nitroxide (MitoTEMPO),

which have been shown in animal models to be protective against sepsis-induced

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

20

cardiomyopathy (Supavekin et al., 2003; Zang et al., 2012; Yao et al., 2015), renal injury

(Patil et al., 2014), and microcirculatory failure (Patil et al., 2014; Sims et al., 2014).

Combinations of Biomarkers

The heterogeneity of septic patients suggests that multiple biomarkers may be

required to effectively guide therapy (Gibot et al., 2012; Liu et al., 2014; Sandquist and

Wong, 2014). Wong and collaborators have begun to use panels of biomarkers to

stratify pediatric septic shock patients to predict risk (Wong et al., 2014) and possibly

inform on therapeutic decisions (Wong et al., 2015a; Wong et al., 2015b). For example,

the PERSEVERE study used a panel consisting of C-C chemokine ligand 3, IL-8, heat

shock protein 70 kDa 1B, granzyme B, and MMP-8 to stratify pediatric sepsis patients

based on mortality probability (Wong et al., 2014). A panel of candidate biomarker

genes has also been used to guide the use of adjuvant corticosteroids (Wong et al.,

2015b). Due to the complexity of sepsis and the diversity of patient populations, it

should expected that combinations of biomarkers (small molecule and physiological) will

be required to best inform on interventional therapies (Fig. 1).

Animal Models

Animal models of sepsis are the subject of a number of excellent reviews that

discuss advantages and disadvantages of the various species studied and the various

methods used to induce sepsis (Zanotti-Cavazzoni and Goldfarb, 2009; Ward, 2012;

Fink, 2013; Efron et al., 2015). It is clear that no animal model of sepsis can fully

replicate the complexities and range of sepsis in humans. However, animal models are

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

21

essential for preclinical testing of new therapies to help suggest dose, potential toxicities,

and efficacy. Animal models are also necessary to identify potential biomarkers and test

new monitoring modalities. From a basic science point of view, in vivo models of sepsis

provide a means to investigate underlying mechanisms of organ injury that are not

possible in very sick septic patients.

Murine and rat models of sepsis are the most frequently used because of their

relatively low cost and their public acceptance as laboratory subjects. However, just

how relevant murine models of inflammation are to inflammatory diseases is an area of

intense debate (Seok et al., 2013; Osuchowski et al., 2014; Efron et al., 2015). Larger

species, such as pigs, sheep, dogs, and subhuman primates, are also used in sepsis

research, and some of these species better replicate more of the “typical”

cardiovascular defects observed in humans with sepsis than do small animals (Zanotti-

Cavazzoni and Goldfarb, 2009; Taylor et al., 2012). Not surprisingly, however, large-

animal models are rarely used in survival studies. This is an important consideration

because increased mortality is a key measure of efficacy in sepsis clinical trials.

Because sepsis is defined as a systemic inflammatory response due to an

infection (Dellinger et al., 2013), the most clinically relevant models of sepsis should

incorporate both a systemic inflammatory response and an active infection. Models of

sepsis that utilize endotoxemia (lipopolysaccharide [LPS] from the outer membrane of

gram-negative bacteria) in rodents and larger species are less clinically relevant

because these models rely solely on rapid activation of the innate immune system and

do not replicate the consequences of active microbial defense and subsequent immune

suppression observed in humans with sepsis (Stearns-Kurosawa et al., 2011).

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

22

In rodents, the cytokine profile and magnitude of cytokine release are very

different between endotoxemia models and active-infection models, such as

polymicrobial peritonitis induced by cecal ligation and puncture (CLP). LPS produces a

very rapid but transient increase in inflammatory cytokines to levels that can be orders

of magnitude higher than with CLP (Kawai et al., 1999; Remick et al., 2000; Miyaji et al.,

2003), where the magnitude and kinetics of cytokine release more closely mirror what

occurs in human sepsis (Rittirsch et al., 2007). Still, there are advantages to studying

endotoxemia. Endotoxemia models are attractive from an experimental point of view

because they are highly reproducible and do produce similar effects on the vasculature

(although perhaps through different mechanisms (Asano et al., 2015) and similar

mortality rates (Remick et al., 2000). A limitation of endotoxemia models is that the

systemic inflammatory effects are mediated through interactions with toll-like receptor-4

(TLR4) (Kawai et al., 1999; Palsson-McDermott and O'Neill, 2004), whereas this is not

the case in polymicrobial sepsis (Dear et al., 2006). These are mechanistically important

differences between endotoxin and polymicrobial models of sepsis that can impact the

translation of experimental therapies to clinical practice (Rittirsch et al., 2007).

Consequently, the CLP model and other models of active infection are generally

considered to be clinically relevant models of sepsis, especially when they incorporate

antibiotic and fluid therapy to more closely mimic the standard care of septic patients

(Miyaji et al., 2003; Rittirsch et al., 2007; Dyson and Singer, 2009; Rittirsch et al., 2009).

However, the CLP model exhibits high inherent variability because it depends on the

resident bacterial flora of the cecal contents, which can vary from animal to animal, and

on the consistency of a technician’s surgical skills.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

23

Depending on the dose, intravenous infusion of live E. coli in sheep can produce

mild hyperdynamic (increased cardiac output) sepsis or severe hypotensive sepsis

(Ramchandra et al., 2009; Ishikawa et al., 2011) to mimic the range of sepsis severity

and degree of cardiovascular dysfunction observed in humans. Similarly, in adult pigs,

intravenous administration of live bacteria can produce hypotensive sepsis associated

with pulmonary hypertension and a transient increase in systemic vascular resistance

(Garcia-Septien et al., 2010); in addition, when acute kidney injury develops, a

dissociation is produced between systemic hemodynamic changes and changes in the

renal circulation (Benes et al., 2011). In baboons, intravenous challenge with live E. coli

can be titrated to produce mild sepsis or severe sepsis with refractory hypotension and

coagulopathies (Taylor et al., 2012). An advantage of live-bacteria infusion models is

that gram-positive bacteria can also be studied (Soerensen et al., 2012), as well as

controlled combinations of bacteria. Another advantage of these intravenous live culture

models over the CLP model is that sepsis can be induced without subjecting the animal

to a surgical procedure or general anesthesia.

Autologous fecal peritonitis is a variation of the CLP model in which the cecal

contents are collected from an animal and administered at a later time. This model has

been used in sheep (He et al., 2012; Taccone et al., 2014) and pigs (Benes et al., 2011;

Correa et al., 2012; Wepler et al., 2013) to study sepsis-induced cardiovascular injury

and evaluate resuscitation protocols. A variation in which the fecal matter is derived

from multiple animals can also be used in mice too young or too small for the CLP

procedure (Wynn et al., 2007; Wynn et al., 2008). This model offers an additional

advantage in that cecal contents can be harvested from multiple animals, pooled,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

24

partially purified, stored at –80°C, and then administered to groups of animals (Starr et

al., 2014). The “dose” of fecal contents can be normalized to bacterial colony–forming

units (cfu)/body weight prior to administration, which should reduce interexperimental

variability.

As discussed earlier, sepsis is not identical in neonates, infants, and adults, and

it should not be assumed that results from adult animal models of sepsis are relevant for

translation to neonates or even infants with sepsis. Most preclinical studies are

performed in adult “middle-aged” animals, yet there are clear differences in the

immunological responses (Wynn et al., 2007; Wynn et al., 2008; Turnbull et al., 2009),

degree of organ injury (Miyaji et al., 2003; Starr et al., 2015), and mortality (Turnbull et

al., 2009; Starr et al., 2014) between very young, adult, and old mice made septic by

fecal peritonitis. CLP can be performed in young (weaning age) rat pups as a model of

infant sepsis. These animals develop rapid hemodynamic changes and microvascular

dysfunction that are not the same as in adult rats (Seely et al., 2011; Sims et al., 2014).

Neonatal, 3-day-old piglets subjected to CLP show deterioration of cardiac output,

pulmonary hypertension, and shock (Goto et al., 2010), consistent with cold shock (low

cardiac output with high systemic vascular resistance) frequently observed in newborns

and infants with severe sepsis (Wheeler et al., 2011).

Confounding comorbid or predisposing conditions of most septic patients limit the

translation of findings from most animal models. However, researchers are now

beginning to study the pathophysiology of sepsis in the presence of comorbid conditions,

such as chronic kidney disease (Doi et al., 2008; Leelahavanichkul et al., 2011), type 1

(Osuchowski et al., 2010; Filgueiras et al., 2012) and type 2 (Jacob et al., 2008)

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

25

diabetes, and atherosclerosis (Wepler et al., 2013). Such studies are especially

important because they could uncover more targeted therapy for comorbid conditions.

They could also help explain why a therapy fails in septic patients with preexisting

conditions (Doi et al., 2008; Leelahavanichkul et al., 2011; Filgueiras et al., 2012) or

why survivors of sepsis may have increased susceptibility to subsequent organ injury

(Portella et al., 2013).

Murine models of sepsis are also being used to investigate the development and

potential causes of cognitive impairments following sepsis like those observed in human

survivors of sepsis (Iwashyna et al., 2010). For example, mice surviving severe sepsis

induced by CLP show time-dependent learning and memory impairments (Chavan et al.,

2012; Ji et al., 2015) associated with dendritic degeneration (Chavan et al., 2012) and

oxidative stress in the prefrontal cortex and hippocampus (Ji et al., 2015).

Preclinical research has several important roles to play in the development of

novel therapeutics. However, animal models have limitations that weaken the prediction

of clinical efficacy. Still, animal models are necessary for the discovery of plausible

biological targets and the demonstration that these targets can be manipulated to

improve outcomes, at least in the experimental animal. Preclinical testing seeks to

identify targeted therapy that may be effective. Equally important, however, is the use of

preclinical testing to suggest ineffective or harmful therapies. For example,

administration of TNF-α-neutralizing antibody, while protective in most gram-negative

preclinical models, is actually detrimental in gram-positive bacteria models and some

other types of pathogens, perhaps due to inhibition of innate antimicrobial defenses

(Lorente and Marshall, 2005). Preclinical studies testing efficacy against sepsis-induced

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

26

multiorgan failure are more challenging (Andonegui et al., 2009; Kapoor et al., 2010;

Coldewey et al., 2013) but are necessary to support translational research and clinical

trials.

Personalized Medicine

Septic patients still experience unacceptably high morbidity and mortality, even

with optimal goal-directed supportive therapy that meets approved guidelines. It is

important to consider that the likelihood of survivors of severe sepsis to be readmitted to

an inpatient health care facility is higher than matched nonsepsis survivors (Prescott et

al., 2014). This is most likely related to the significant long-term physical and mental

disabilities that can affect survivors. For example, estimates are that sepsis-associated

alterations in mental status and cognitive deficits occur in up to 87% of septic patients

(Iwashyna et al., 2012). While decreasing mortality is and should be the goal of therapy,

it is becoming increasingly clear that patients who initially survive sepsis experience

functional deficits that can diminish quality of life and even increase long-term mortality.

Consequently, biomarkers that also direct therapies that decrease the morbidity of

survivors would have a major impact on the overall health care burden of sepsis (Fig. 2).

Biomarkers and rapid noninvasive monitoring could offer better and more rapid

stratification of patients to identify those who might benefit (or be harmed) by more

targeted therapies. The advent of high-throughput technologies and access to advanced

bioinformatics have made possible the concept of personalized medicine to direct

therapy in septic patients. A personalized medicine approach may have the best chance

of identifying specific strategies for such a diverse patient population. A systems biology

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

27

approach could complement the use of biomarkers to stratify patients, aid in prognosis,

and help identify subsets of patients who may benefit or be harmed by therapy (Tsalik et

al., 2015). However, the costs are high, and it is too early to know whether knowledge of

the blood metabolome, proteome, and transcriptome could really direct therapy.

Perspectives

In the near future, biomarkers could direct the therapy of septic patients in order

to achieve an optimal outcome. Cohen and colleagues (Cohen et al., 2015) suggested

that the ideal profile of a sepsis biomarker should have these characteristics: fast

kinetics, high sensitivity and specificity, fully automated technology, short turnaround

time, availability as point-of-care tests, and low cost. Many clinicians already use

biomarkers to suggest an active infection (CRP, PCT), estimate the severity of under-

resuscitated septic shock (mixed venous blood oxygen saturation, lactate), and estimate

the severity of organ injury due to sepsis (liver enzymes, troponin, creatinine, ratio of

partial pressure of oxygen to fractional inspired oxygen, platelet count, activated partial

thromboplastin time, fibrinogen and D-dimer). Physiological biomarker monitoring

including echocardiography, pulmonary artery catheter, Doppler ultrasound, and/or

continuous pulse-contour cardiac analysis is already being used to direct sepsis

resuscitation and is recommended in the “Surviving Sepsis Campaign” document

(Dellinger et al., 2013).

After more than three decades of numerous failed large sepsis clinical trials,

sepsis is currently being redefined, which will most likely include “MODS due to non-

resolving inflammatory response to infection.” Future clinical trials of sepsis therapies

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

28

will most likely target an individual patient’s underlying pathophysiological mechanism

rather than a general patient population with sepsis. Some of the current proposed

mechanisms for sepsis-induced MODS include pathological immune activation,

immunoparalysis, mitochondrial dysfunction, disseminated microvascular thromboses,

and others (Carcillo et al., 2011). The genetic makeup of the host and pathological

organism, sex and age of the host, environmental factors, and specific interventions

from different hospitals will all influence the underlying pathophysiological mechanism of

the individual septic patient (Rautanen et al., 2015; Srinivasan et al., 2015). In this

context, future real-time measurements of biomarkers could identify the pathological

mechanism and direct therapeutic strategies in septic patients, including immune

modulation for pathological immune activation, boosting the immune response for

immunoparalysis, optimizing/rescuing mitochondrial function, reducing oxidative stress,

and/or normalizing dysregulated hemostasis. Eventually, more advanced physiological

monitoring, such as assessment of the microcirculation of specific organs, will give

instantaneous feedback of intervention where titration of therapeutic agents could be

optimized.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

29

Authorship Contributions

Participated in research design: NA.

Conducted experiments: NA.

Contributed new reagents or analytic tools: NA.

Performed data analysis: NA.

Wrote or contributed to the writing of the manuscript: Sims, Nguyen, Mayeux.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

30

References

Abraham E, Reinhart K, Opal S, Demeyer I, Doig C, Rodriguez AL, Beale R, Svoboda P,

Laterre PF, Simon S, Light B, Spapen H, Stone J, Seibert A, Peckelsen C, De

Deyne C, Postier R, Pettila V, Sprung CL, Artigas A, Percell SR, Shu V,

Zwingelstein C, Tobias J, Poole L, Stolzenbach JC, Creasey AA and Group OTS

(2003) Efficacy and safety of tifacogin (recombinant tissue factor pathway

inhibitor) in severe sepsis: a randomized controlled trial. J Am Med Assoc

290:238-247.

Adam N, Kandelman S, Mantz J, Chretien F and Sharshar T (2013) Sepsis-induced

brain dysfunction. Expert Rev Anti Infect Ther 11:211-221.

Adkins B, Leclerc C and Marshall-Clarke S (2004) Neonatal adaptive immunity comes

of age. Nat Rev Immunol 4:553-564.

Agrawal A, Matthay MA, Kangelaris KN, Stein J, Chu JC, Imp BM, Cortez A, Abbott J,

Liu KD and Calfee CS (2013) Plasma angiopoietin-2 predicts the onset of acute

lung injury in critically ill patients. Am J Respir Crit Care Med 187:736-742.

Alhamdi Y, Abrams ST, Cheng Z, Jing S, Su D, Liu Z, Lane S, Welters I, Wang G and

Toh CH (2015) Circulating Histones Are Major Mediators of Cardiac Injury in

Patients With Sepsis. Crit Care Med 43:2094-2103.

Andonegui G, Goring K, Liu D, McCafferty DM and Winston BW (2009) Characterization

of S. pneumoniae pneumonia-induced multiple organ dysfunction syndrome: an

experimental mouse model of gram-positive sepsis. Shock 31:423-428.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

31

Aneja RK and Carcillo JA (2011) Differences between adult and pediatric septic shock.

Minerva Anestesiol 77:986-992.

Asano S, Manne ND, Nandyala G, Ma B, Selvaraj V, Ravikumar A, Rice KM and Blough

E (2015) Cecal inoculum peritonitis: An alternative model for sepsis vascular

dysfunction study. Life Sci 141:108-118.

Azabou E, Magalhaes E, Braconnier A, Yahiaoui L, Moneger G, Heming N, Annane D,

Mantz J, Chretien F, Durand MC, Lofaso F, Porcher R, Sharshar T and Groupe

d'Explorations Neurologiques en R (2015) Early Standard Electroencephalogram

Abnormalities Predict Mortality in Septic Intensive Care Unit Patients. PLoS One

10:e0139969.

Basu RK, Standage SW, Cvijanovich NZ, Allen GL, Thomas NJ, Freishtat RJ, Anas N,

Meyer K, Checchia PA, Lin R, Shanley TP, Bigham MT, Wheeler DS, Devarajan

P, Goldstein SL and Wong HR (2011) Identification of candidate serum

biomarkers for severe septic shock-associated kidney injury via microarray. Crit

Care 15:R273.

Benes J, Chvojka J, Sykora R, Radej J, Krouzecky A, Novak I and Matejovic M (2011)

Searching for mechanisms that matter in early septic acute kidney injury: an

experimental study. Crit Care 15:R256.

Bersani I, Auriti C, Ronchetti MP, Prencipe G, Gazzolo D and Dotta A (2015) Use of

early biomarkers in neonatal brain damage and sepsis: state of the art and future

perspectives. Biomed Res Int 2015:253520.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

32

Bhatti MM, Boonlayangoor S, Beavis KG and Tesic V (2014a) Evaluation of FilmArray

and Verigene systems for rapid identification of positive blood cultures. J Clin

Microbiol 52:3433-3436.

Bhatti MM, Boonlayangoor S, Beavis KG and Tesic V (2014b) Rapid identification of

positive blood cultures by matrix-assisted laser desorption ionization-time of flight

mass spectrometry using prewarmed agar plates. J Clin Microbiol 52:4334-4338.

Biron BM, Ayala A and Lomas-Neira JL (2015) Biomarkers for Sepsis: What Is and

What Might Be? Biomark Insights 10:7-17.

Bockmeyer CL, Claus RA, Budde U, Kentouche K, Schneppenheim R, Losche W,

Reinhart K and Brunkhorst FM (2008) Inflammation-associated ADAMTS13

deficiency promotes formation of ultra-large von Willebrand factor.

Haematologica 93:137-140.

Bone RC (1996) Sir Isaac Newton, sepsis, SIRS, and CARS. Crit Care Med 24:1125-

1128.

Boomer JS, Green JM and Hotchkiss RS (2014) The changing immune system in

sepsis: is individualized immuno-modulatory therapy the answer? Virulence 5:45-

56.

Boss GR and Seegmiller JE (1981) Age-related physiological changes and their clinical

significance. West J Med 135:434-440.

Cao Z and Robinson RA (2014) The role of proteomics in understanding biological

mechanisms of sepsis. Proteomics Clin Appl 8:35-52.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

33

Cao Z, Yende S, Kellum JA, Angus DC and Robinson RA (2014) Proteomics reveals

age-related differences in the host immune response to sepsis. J Proteome Res

13:422-432.

Carcillo JA, Han YY, Doughty L, Hall M, Nguyen TC and Aneja R (2011) Inflammation

and Immunity: Systemic Inflammatory Response Syndrome, Sepsis, Acute Lung

Injury, and Multiple Organ Failure, in Fuhrman & Zimmerman’s Pediatric Critical

Care 4th edition (Fuhrman B ZJ, Carcillo J, Clark R, Rotta A, Tobias J, Relvas M,

Thompson A ed), Elsevier.

Casserly B, Phillips GS, Schorr C, Dellinger RP, Townsend SR, Osborn TM, Reinhart K,

Selvakumar N and Levy MM (2015) Lactate measurements in sepsis-induced

tissue hypoperfusion: results from the Surviving Sepsis Campaign database. Crit

Care Med 43:567-573.

Chang DW, Tseng CH and Shapiro MF (2015) Rehospitalizations Following Sepsis:

Common and Costly. Crit Care Med 43:2085-2093.

Charchaflieh J, Rushbrook J, Worah S and Zhang M (2015) Activated Complement

Factors as Disease Markers for Sepsis. Dis Markers 2015:382463.

Chavan SS, Huerta PT, Robbiati S, Valdes-Ferrer SI, Ochani M, Dancho M, Frankfurt M,

Volpe BT, Tracey KJ and Diamond B (2012) HMGB1 mediates cognitive

impairment in sepsis survivors. Mol Med 18:930-937.

Chertoff J, Chisum M, Garcia B and Lascano J (2015) Lactate kinetics in sepsis and

septic shock: a review of the literature and rationale for further research. J

Intensive Care 3:39.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

34

Christaki E and Giamarellos-Bourboulis EJ (2014) The beginning of personalized

medicine in sepsis: small steps to a bright future. Clin Genet 86:56-61.

Cohen J, Vincent JL, Adhikari NK, Machado FR, Angus DC, Calandra T, Jaton K,

Giulieri S, Delaloye J, Opal S, Tracey K, van der Poll T and Pelfrene E (2015)

Sepsis: a roadmap for future research. Lancet Infect Dis 15:581-614.

Coldewey SM, Rogazzo M, Collino M, Patel NS and Thiemermann C (2013) Inhibition of

IkappaB kinase reduces the multiple organ dysfunction caused by sepsis in the

mouse. Dis Model Mech 6:1031-1042.

Comim CM, Bussmann RM, Simao SR, Ventura L, Freiberger V, Patricio JJ, Palmas D,

Mendonca BP, Cassol-Jr OJ and Quevedo J (2015) Experimental Neonatal

Sepsis Causes Long-Term Cognitive Impairment. Mol Neurobiol:in press.

Conference NAC (1992) American College of Chest Physicians/Society of Critical Care

Medicine Consensus Conference: definitions for sepsis and organ failure and

guidelines for the use of innovative therapies in sepsis. Crit Care Med 20:864-

874.

Correa TD, Vuda M, Blaser AR, Takala J, Djafarzadeh S, Dunser MW, Silva E, Lensch

M, Wilkens L and Jakob SM (2012) Effect of treatment delay on disease severity

and need for resuscitation in porcine fecal peritonitis. Crit Care Med 40:2841-

2849.

Darwish I and Liles WC (2013) Emerging therapeutic strategies to prevent infection-

related microvascular endothelial activation and dysfunction. Virulence 4:572-582.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

35

Daviaud F, Grimaldi D, Dechartres A, Charpentier J, Geri G, Marin N, Chiche JD,

Cariou A, Mira JP and Pene F (2015) Timing and causes of death in septic shock.

Ann Intensive Care 5:58.

De Backer D, Donadello K and Cortes DO (2012) Monitoring the microcirculation. J Clin

Monit Comput 26:361-366.

De Backer D, Donadello K, Sakr Y, Ospina-Tascon G, Salgado D, Scolletta S and

Vincent JL (2013) Microcirculatory alterations in patients with severe sepsis:

impact of time of assessment and relationship with outcome. Crit Care Med

41:791-799.

De Backer D, Orbegozo Cortes D, Donadello K and Vincent JL (2014) Pathophysiology

of microcirculatory dysfunction and the pathogenesis of septic shock. Virulence

5:73-79.

Dear JW, Kobayashi H, Jo SK, Holly MK, Hu X, Yuen PS, Brechbiel MW and Star RA

(2005) Dendrimer-enhanced MRI as a diagnostic and prognostic biomarker of

sepsis-induced acute renal failure in aged mice. Kidney Int 67:2159-2167.

Dear JW, Yasuda H, Hu X, Hieny S, Yuen PS, Hewitt SM, Sher A and Star RA (2006)

Sepsis-induced organ failure is mediated by different pathways in the kidney and

liver: Acute renal failure is dependent on MyD88 but not renal cell apoptosis.

Kidney Int 69:832-836.

DeCoux A, Tian Y, DeLeon-Pennell KY, Nguyen NT, de Castro Bras LE, Flynn ER,

Cannon PL, Griswold ME, Jin YF, Puskarich MA, Jones AE and Lindsey ML

(2015) Plasma Glycoproteomics Reveals Sepsis Outcomes Linked to Distinct

Proteins in Common Pathways. Crit Care Med 43:2049-2058.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

36

Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, Sevransky JE,

Sprung CL, Douglas IS, Jaeschke R, Osborn TM, Nunnally ME, Townsend SR,

Reinhart K, Kleinpell RM, Angus DC, Deutschman CS, Machado FR, Rubenfeld

GD, Webb SA, Beale RJ, Vincent JL, Moreno R and Surviving Sepsis Campaign

Guidelines Committee including the Pediatric S (2013) Surviving sepsis

campaign: international guidelines for management of severe sepsis and septic

shock: 2012. Crit Care Med 41:580-637.

Doi K, Leelahavanichkul A, Hu X, Sidransky KL, Zhou H, Qin Y, Eisner C, Schnermann

J, Yuen PS and Star RA (2008) Pre-existing renal disease promotes sepsis-

induced acute kidney injury and worsens outcome. Kidney Int 74:1017-1025.

Drewry AM and Hotchkiss RS (2015) Sepsis: Revising definitions of sepsis. Nat Rev

Nephrol 11:326-328.

Dupuy AM, Philippart F, Pean Y, Lasocki S, Charles PE, Chalumeau M, Claessens YE,

Quenot JP, Guen CG, Ruiz S, Luyt CE, Roche N, Stahl JP, Bedos JP, Pugin J,

Gauzit R, Misset B, Brun-Buisson C and Maurice Rapin Institute Biomarkers G

(2013) Role of biomarkers in the management of antibiotic therapy: an expert

panel review: I - currently available biomarkers for clinical use in acute infections.

Ann Intensive Care 3:22.

Duran-Bedolla J, Montes de Oca-Sandoval MA, Saldana-Navor V, Villalobos-Silva JA,

Rodriguez MC and Rivas-Arancibia S (2014) Sepsis, mitochondrial failure and

multiple organ dysfunction. Clin Invest Med 37:E58-E69.

Dyson A and Singer M (2009) Animal models of sepsis: why does preclinical efficacy fail

to translate to the clinical setting? Crit Care Med 37:S30-S37.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

37

Efron PA, Mohr AM, Moore FA and Moldawer LL (2015) The future of murine sepsis

and trauma research models. J Leukoc Biol 98:945-952.

Elseviers MM, Lins RL, Van der Niepen P, Hoste E, Malbrain ML, Damas P, Devriendt J

and investigators S (2010) Renal replacement therapy is an independent risk

factor for mortality in critically ill patients with acute kidney injury. Crit Care

14:R221.

Fang Y, Li C, Shao R, Yu H, Zhang Q and Zhao L (2015) Prognostic significance of the

angiopoietin-2/angiopoietin-1 and angiopoietin-1/Tie-2 ratios for early sepsis in

an emergency department. Crit Care 19:367.

Filgueiras LR, Jr., Martins JO, Serezani CH, Capelozzi VL, Montes MB and Jancar S

(2012) Sepsis-induced acute lung injury (ALI) is milder in diabetic rats and

correlates with impaired NFkB activation. PLoS One 7:e44987.

Fink MP (2013) Animal models of sepsis. Virulence 5:143-153.

Fink MP (2015) Cytopathic hypoxia and sepsis: is mitochondrial dysfunction

pathophysiologically important or just an epiphenomenon. Pediatr Crit Care Med

16:89-91.

Fink MP and Warren HS (2014) Strategies to improve drug development for sepsis. Nat

Rev Drug Discov 13:741-758.

Fink T, Heymann P, Taha-Melitz S, Taha A, Wolf B, Rensing H, Volk T and Mathes AM

(2013) Dobutamine pretreatment improves survival, liver function, and hepatic

microcirculation after polymicrobial sepsis in rat. Shock 40:129-135.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

38

Fiusa MM, Carvalho-Filho MA, Annichino-Bizzacchi JM and De Paula EV (2015)

Causes and consequences of coagulation activation in sepsis: an evolutionary

medicine perspective. BMC Med 13:105.

Fortenberry JD, Knezevic A, Nguyen TC and Group TNS (2012) Outcome in Childrens

on ECMO with TAMOF Receiving Plasma Exchange: Results from the

Prospective Pediatric TAMOF Network Study Crit Care Med 40:A520.

Gaieski DF, Edwards JM, Kallan MJ and Carr BG (2013) Benchmarking the incidence

and mortality of severe sepsis in the United States. Crit Care Med 41:1167-1174.

Gao R, Kan MQ, Wang SG, Yang RH and Zhang SG (2015) Disrupted Tryptophan

Metabolism Induced Cognitive Impairment in a Mouse Model of Sepsis-

associated Encephalopathy. Inflammation:in press.

Garcia-Septien J, Lorente JA, Delgado MA, de Paula M, Nin N, Moscoso A, Sanchez-

Ferrer A, Perez-Vizcaino F and Esteban A (2010) Levosimendan increases portal

blood flow and attenuates intestinal intramucosal acidosis in experimental septic

shock. Shock 34:275-280.

Ghosh CC, Thamm K, Berghelli AV, Schrimpf C, Maski MR, Abid T, Milam KE,

Rajakumar A, Santel A, Kielstein JT, Ahmed A, Thickett D, Wang K, Chase M,

Donnino MW, Aird WC, Haller H, David S and Parikh SM (2015) Drug

Repurposing Screen Identifies Foxo1-Dependent Angiopoietin-2 Regulation in

Sepsis. Crit Care Med 43:e230-240.

Gibot S, Bene MC, Noel R, Massin F, Guy J, Cravoisy A, Barraud D, De Carvalho

Bittencourt M, Quenot JP, Bollaert PE, Faure G and Charles PE (2012)

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

39

Combination biomarkers to diagnose sepsis in the critically ill patient. Am J

Respir Crit Care Med 186:65-71.

Ginde AA, Blatchford PJ, Trzeciak S, Hollander JE, Birkhahn R, Otero R, Osborn TM,

Moretti E, Nguyen HB, Gunnerson KJ, Milzman D, Gaieski DF, Goyal M, Cairns

CB, Rivers EP and Shapiro NI (2014) Age-related differences in biomarkers of

acute inflammation during hospitalization for sepsis. Shock 42:99-107.

Goto T, Hussein MH, Kato S, Daoud GA, Kato T, Kakita H, Mizuno H, Imai M, Ito T,

Kato I, Suzuki S, Okada N, Togari H and Okada H (2010) Endothelin receptor

antagonist attenuates inflammatory response and prolongs the survival time in a

neonatal sepsis model. Intensive Care Med 36:2123-2139.

Group BDW (2001) Biomarkers and surrogate endpoints: preferred definitions and

conceptual framework. Clin Pharmacol Ther 69:89-95.

Gustot T (2011) Multiple organ failure in sepsis: prognosis and role of systemic

inflammatory response. Curr Opin Crit Care 17:153-159.

Han JH, Nachamkin I, Coffin SE, Gerber JS, Fuchs B, Garrigan C, Han X, Bilker WB,

Wise J, Tolomeo P, Lautenbach E, Prevention Epicenters Program of the

Centers for Disease C and Prevention (2015) Use of a Combination Biomarker

Algorithm To Identify Medical Intensive Care Unit Patients with Suspected Sepsis

at Very Low Likelihood of Bacterial Infection. Antibicrob Agents Chemother

59:6494-6500.

Hartman ME, Linde-Zwirble WT, Angus DC and Watson RS (2013) Trends in the

epidemiology of pediatric severe sepsis. Pediatr Crit Care Med 14:686-693.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

40

He X, Su F, Velissaris D, Salgado DR, de Souza Barros D, Lorent S, Taccone FS,

Vincent JL and De Backer D (2012) Administration of tetrahydrobiopterin

improves the microcirculation and outcome in an ovine model of septic shock.

Crit Care Med 40:2833-2840.

Heron M (2015) Deaths: Leading Causes for 2011. National Vital Statistics Reports

64:1-96.

Holthoff JH, Wang Z, Patil NK, Gokden N and Mayeux PR (2013) Rolipram improves

renal perfusion and function during sepsis in the mouse. J Pharmacol Exp Ther

347:357-364.

Holthoff JH, Wang Z, Seely KA, Gokden N and Mayeux PR (2012) Resveratrol

improves renal microcirculation, protects the tubular epithelium, and prolongs

survival in a mouse model of sepsis-induced acute kidney injury. Kidney Int

81:370-378.

Hotchkiss RS, Monneret G and Payen D (2013a) Immunosuppression in sepsis: a novel

understanding of the disorder and a new therapeutic approach. Lancet Infect Dis

13:260-268.

Hotchkiss RS, Monneret G and Payen D (2013b) Sepsis-induced immunosuppression:

from cellular dysfunctions to immunotherapy. Nat Rev Immunol 13:862-874.

Hotchkiss RS and Sherwood ER (2015) Getting sepsis therapy right. Science 347:1201-

1202.

Hutchins NA, Unsinger J, Hotchkiss RS and Ayala A (2014) The new normal:

immunomodulatory agents against sepsis immune suppression. Trends Mol Med

20:224-233.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

41

Ishikawa K, Bellomo R and May CN (2011) The impact of intrarenal nitric oxide

synthase inhibition on renal blood flow and function in mild and severe

hyperdynamic sepsis. Crit Care Med 39:770-776.

Iskander KN, Osuchowski MF, Stearns-Kurosawa DJ, Kurosawa S, Stepien D,

Valentine C and Remick DG (2013) Sepsis: multiple abnormalities,

heterogeneous responses, and evolving understanding. Physiol Rev 93:1247-

1288.

Iwashyna TJ, Cooke CR, Wunsch H and Kahn JM (2012) Population burden of long-

term survivorship after severe sepsis in older Americans. J Am Geriatr Soc

60:1070-1077.

Iwashyna TJ, Ely EW, Smith DM and Langa KM (2010) Long-term cognitive impairment

and functional disability among survivors of severe sepsis. J Am Med Assoc

304:1787-1794.

Jacob A, Steinberg ML, Yang J, Dong W, Ji Y and Wang P (2008) Sepsis-induced

inflammation is exacerbated in an animal model of type 2 diabetes. Int J Clin Exp

Med 1:22-31.

Jaimes F, De La Rosa G, Morales C, Fortich F, Arango C, Aguirre D and Munoz A

(2009) Unfractioned heparin for treatment of sepsis: A randomized clinical trial

(The HETRASE Study). Crit Care Med 37:1185-1196.

Jeger V, Djafarzadeh S, Jakob SM and Takala J (2013) Mitochondrial function in sepsis.

Eur J Clin Invest 43:532-542.

Ji MH, Qiu LL, Tang H, Ju LS, Sun XR, Zhang H, Jia M, Zuo ZY, Shen JC and Yang JJ

(2015) Sepsis-induced selective parvalbumin interneuron phenotype loss and

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

42

cognitive impairments may be mediated by NADPH oxidase 2 activation in mice.

J Neuroinflammation 12:182.

Kalbitz M, Grailer JJ, Fattahi F, Jajou L, Herron TJ, Campbell KF, Zetoune FS,

Bosmann M, Sarma JV, Huber-Lang M, Gebhard F, Loaiza R, Valdivia HH, Jalife

J, Russell MW and Ward PA (2015) Role of extracellular histones in the

cardiomyopathy of sepsis. FASEB J.

Kaplan JM and Wong HR (2011) Biomarker discovery and development in pediatric

critical care medicine. Pediatr Crit Care Med 12:165-173.

Kapoor A, Shintani Y, Collino M, Osuchowski MF, Busch D, Patel NS, Sepodes B,

Castiglia S, Fantozzi R, Bishop-Bailey D, Mota-Filipe H, Yaqoob MM, Suzuki K,

Bahrami S, Desvergne B, Mitchell JA and Thiemermann C (2010) Protective role

of peroxisome proliferator-activated receptor-beta/delta in septic shock. Am J

Respir Crit Care Med 182:1506-1515.

Kaukonen KM, Bailey M, Pilcher D, Cooper DJ and Bellomo R (2015) Systemic

inflammatory response syndrome criteria in defining severe sepsis. N Engl J Med

372:1629-1638.

Kawai T, Adachi O, Ogawa T, Takeda K and Akira S (1999) Unresponsiveness of

MyD88-deficient mice to endotoxin. Immunity 11:115-122.

Kissoon N, Carcillo JA, Espinosa V, Argent A, Devictor D, Madden M, Singhi S, van der

Voort E, Latour J and Global Sepsis Initiative Vanguard Center C (2011) World

Federation of Pediatric Intensive Care and Critical Care Societies: Global Sepsis

Initiative. Pediatr Crit Care Med 12:494-503.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

43

Kojic D, Siegler BH, Uhle F, Lichtenstern C, Nawroth PP, Weigand MA, Hofer S and

Brenner T (2015) Are there new approaches for diagnosis, therapy guidance and

outcome prediction of sepsis? World J Exp Med 5:50-63.

Lapeyraque AL, Malina M, Fremeaux-Bacchi V, Boppel T, Kirschfink M, Oualha M,

Proulx F, Clermont MJ, Le Deist F, Niaudet P and Schaefer F (2011) Eculizumab

in severe Shiga-toxin-associated HUS. N Engl J Med 364:2561-2563.

Laszlo I, Trasy D, Molnar Z and Fazakas J (2015) Sepsis: From Pathophysiology to

Individualized Patient Care. J Immunol Res 2015:510436.

Lee MJ, Thangada S, Claffey KP, Ancellin N, Liu CH, Kluk M, Volpi M, Sha'afi RI and

Hla T (1999) Vascular endothelial cell adherens junction assembly and

morphogenesis induced by sphingosine-1-phosphate. Cell 99:301-312.

Leelahavanichkul A, Huang Y, Hu X, Zhou H, Tsuji T, Chen R, Kopp JB, Schnermann J,

Yuen PS and Star RA (2011) Chronic kidney disease worsens sepsis and sepsis-

induced acute kidney injury by releasing High Mobility Group Box Protein-1.

Kidney Int 80:1198-1211.

Levi M and van der Poll T (2013) Disseminated intravascular coagulation: a review for

the internist. Intern Emerg Med 8:23-32.

Liangos O, Perianayagam MC, Vaidya VS, Han WK, Wald R, Tighiouart H, MacKinnon

RW, Li L, Balakrishnan VS, Pereira BJ, Bonventre JV and Jaber BL (2007)

Urinary N-acetyl-beta-(D)-glucosaminidase activity and kidney injury molecule-1

level are associated with adverse outcomes in acute renal failure. J Am Soc

Nephrol 18:904-912.

Liu X, Ren H and Peng D (2014) Sepsis biomarkers: an omics perspective. Front Med.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

44

Lorente JA and Marshall JC (2005) Neutralization of tumor necrosis factor in preclinical

models of sepsis. Shock 24 Suppl 1:107-119.

Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C,

Compton D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato TN

and Yancopoulos GD (1997) Angiopoietin-2, a natural antagonist for Tie2 that

disrupts in vivo angiogenesis. Science 277:55-60.

Marshall JC (2014) Why have clinical trials in sepsis failed? Trends Mol Med 20:195-

203.

Marshall JC, Reinhart K and International Sepsis F (2009) Biomarkers of sepsis. Crit

Care Med 37:2290-2298.

Martin GS (2012) Sepsis, severe sepsis and septic shock: changes in incidence,

pathogens and outcomes. Expert Rev Mol Med 10:701-706.

Martin K, Borgel D, Lerolle N, Feys HB, Trinquart L, Vanhoorelbeke K, Deckmyn H,

Legendre P, Diehl JL and Baruch D (2007) Decreased ADAMTS-13 (A

disintegrin-like and metalloprotease with thrombospondin type 1 repeats) is

associated with a poor prognosis in sepsis-induced organ failure. Crit Care Med

35:2375-2382.

Matejovic M, Ince C, Chawla LS, Blantz R, Molitoris BA, Rosner MH, Okusa MD, Kellum

JA, Ronco C and Group AXW (2015) Renal Hemodynamics in AKI: In Search of

New Treatment Targets. J Am Soc Nephrol 27:49-58.

Mathias B, Szpila BE, Moore FA, Efron PA and Moldawer LL (2015) A Review of GM-

CSF Therapy in Sepsis. Medicine (Baltimore) 94:e2044.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

45

Mayeux PR and Macmillan-Crow LA (2012) Pharmacological targets in the renal

peritubular microenvironment: Implications for therapy for sepsis-induced acute

kidney injury. Pharmacol Ther 134:139-155.

Mera S, Tatulescu D, Cismaru C, Bondor C, Slavcovici A, Zanc V, Carstina D and

Oltean M (2011) Multiplex cytokine profiling in patients with sepsis. APMIS

119:155-163.

Mikkelsen ME, Shah CV, Meyer NJ, Gaieski DF, Lyon S, Miltiades AN, Goyal M, Fuchs

BD, Bellamy SL and Christie JD (2013) The epidemiology of acute respiratory

distress syndrome in patients presenting to the emergency department with

severe sepsis. Shock 40:375-381.

Miyaji T, Hu X, Yuen PS, Muramatsu Y, Iyer S, Hewitt SM and Star RA (2003) Ethyl

pyruvate decreases sepsis-induced acute renal failure and multiple organ

damage in aged mice. Kidney Int 64:1620-1631.

Nadel S, Goldstein B, Williams MD, Dalton H, Peters M, Macias WL, Abd-Allah SA,

Levy H, Angle R, Wang D, Sundin DP, Giroir B, Sepsis REs and Organ

dysfunction in children: a gLobal perspective study g (2007) Drotrecogin alfa

(activated) in children with severe sepsis: a multicentre phase III randomised

controlled trial. Lancet 369:836-843.

Nguyen TC, Han Y, Zhou Z, Fortenberry J, Cruz MA and Carcillo JA (2014)

Thrombocytopenia-Associated Multiple Organ Failure, in Pediatric Critical Care

Medicine: Basic Science and Clinical Evidence (Wheeler W, Shanley ed).

Nguyen TC, Han YY, Kiss JE, Hall MW, Hassett AC, Jaffe R, Orr RA, Janosky J and

Carcillo JA (2008) Intensive plasma exchange increases a disintegrin and

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

46

metalloprotease with thrombospondin motifs-13 activity and reverses organ

dysfunction in children with thrombocytopenia-associated multiple organ failure.

Crit Care Med 36:2878-2887.

Noris M, Mescia F and Remuzzi G (2012) STEC-HUS, atypical HUS and TTP are all

diseases of complement activation. Nat Rev Nephrol 8:622-633.

Orde SR, Pulido JN, Masaki M, Gillespie S, Spoon JN, Kane GC and Oh JK (2014)

Outcome prediction in sepsis: speckle tracking echocardiography based

assessment of myocardial function. Crit Care 18:R149.

Ostrowski SR, Haase N, Muller RB, Moller MH, Pott FC, Perner A and Johansson PI

(2015) Association between biomarkers of endothelial injury and

hypocoagulability in patients with severe sepsis: a prospective study. Crit Care

19:191.

Osuchowski MF, Craciun FL, Schuller E, Sima C, Gyurko R and Remick DG (2010)

Untreated type 1 diabetes increases sepsis-induced mortality without inducing a

prelethal cytokine response. Shock 34:369-376.

Osuchowski MF, Remick DG, Lederer JA, Lang CH, Aasen AO, Aibiki M, Azevedo LC,

Bahrami S, Boros M, Cooney R, Cuzzocrea S, Jiang Y, Junger WG, Hirasawa H,

Hotchkiss RS, Li XA, Radermacher P, Redl H, Salomao R, Soebandrio A,

Thiemermann C, Vincent JL, Ward P, Yao YM, Yu HP, Zingarelli B and Chaudry

IH (2014) Abandon the mouse research ship? Not just yet! Shock 41:463-475.

Palsson-McDermott EM and O'Neill LA (2004) Signal transduction by the

lipopolysaccharide receptor, Toll-like receptor-4. Immunology 113:153-162.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

47

Patil NK, Parajuli N, Macmillan-Crow LA and Mayeux PR (2014) Inactivation of renal

mitochondrial respiratory complexes and manganese superoxide dismutase

during sepsis: mitochondria targeted antioxidant mitigates injury. Am J Physiol

Renal Physiol 306:F734-F743.

Pierrakos C and Vincent JL (2010) Sepsis biomarkers: a review. Crit Care 14:R15.

Portella VG, Silva-Filho JL, Landgraf SS, de Rico TB, Vieira MA, Takiya CM, Souza MC,

Henriques MG, Canetti C, Pinheiro AA, Benjamim CF and Caruso-Neves C

(2013) Sepsis-Surviving Mice Are More Susceptible to a Secondary Kidney Insult.

Crit Care Med 41:1056-1068.

Prescott HC, Langa KM, Liu V, Escobar GJ and Iwashyna TJ (2014) Increased 1-year

healthcare use in survivors of severe sepsis. Am J Respir Crit Care Med 190:62-

69.

Prowle JR and Davenport A (2015) Does early-start renal replacement therapy improve

outcomes for patients with acute kidney injury? Kidney Int 88:670-673.

Prowle JR, Molan MP, Hornsey E and Bellomo R (2012) Measurement of renal blood

flow by phase-contrast magnetic resonance imaging during septic acute kidney

injury: a pilot investigation. Crit Care Med 40:1768-1776.

Ramchandra R, Wan L, Hood SG, Frithiof R, Bellomo R and May CN (2009) Septic

shock induces distinct changes in sympathetic nerve activity to the heart and

kidney in conscious sheep. Am J Physiol Regul Integr Comp Physiol 297:R1247-

R1253.

Ranieri VM, Thompson BT, Barie PS, Dhainaut JF, Douglas IS, Finfer S, Gardlund B,

Marshall JC, Rhodes A, Artigas A, Payen D, Tenhunen J, Al-Khalidi HR,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

48

Thompson V, Janes J, Macias WL, Vangerow B, Williams MD and Group P-SS

(2012) Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med

366:2055-2064.

Rautanen A, Mills TC, Gordon AC, Hutton P, Steffens M, Nuamah R, Chiche JD, Parks

T, Chapman SJ, Davenport EE, Elliott KS, Bion J, Lichtner P, Meitinger T,

Wienker TF, Caulfield MJ, Mein C, Bloos F, Bobek I, Cotogni P, Sramek V,

Sarapuu S, Kobilay M, Ranieri VM, Rello J, Sirgo G, Weiss YG, Russwurm S,

Schneider EM, Reinhart K, Holloway PA, Knight JC, Garrard CS, Russell JA,

Walley KR, Stuber F, Hill AV, Hinds CJ and Investigators EEG (2015) Genome-

wide association study of survival from sepsis due to pneumonia: an

observational cohort study. Lancet Respiratory Medicine 3:53-60.

Remick DG, Newcomb DE, Bolgos GL and Call DR (2000) Comparison of the mortality

and inflammatory response of two models of sepsis: lipopolysaccharide vs. cecal

ligation and puncture. Shock 13:110-116.

Rittirsch D, Hoesel LM and Ward PA (2007) The disconnect between animal models of

sepsis and human sepsis. J Leukoc Biol 81:137-143.

Rittirsch D, Huber-Lang MS, Flierl MA and Ward PA (2009) Immunodesign of

experimental sepsis by cecal ligation and puncture. Nat Protoc 4:31-36.

Romanovsky A, Morgan C and Bagshaw SM (2013) Pathophysiology and management

of septic acute kidney injury. Pediatr Nephrol 29:1-12.

Rule JA, Hynan LS, Attar N, Sanders C, Korzun WJ, Lee WM and Acute Liver Failure

Study G (2015) Procalcitonin Identifies Cell Injury, Not Bacterial Infection, in

Acute Liver Failure. PLoS One 10:e0138566.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

49

Ruth A, McCracken CE, Fortenberry JD, Hall M, Simon HK and Hebbar KB (2014)

Pediatric severe sepsis: current trends and outcomes from the Pediatric Health

Information Systems database. Pediatr Crit Care Med 15:828-838.

Saito H, Maruyama I, Shimazaki S, Yamamoto Y, Aikawa N, Ohno R, Hirayama A,

Matsuda T, Asakura H, Nakashima M and Aoki N (2007) Efficacy and safety of

recombinant human soluble thrombomodulin (ART-123) in disseminated

intravascular coagulation: results of a phase III, randomized, double-blind clinical

trial. J Thromb Haemost 5:31-41.

Sandquist M and Wong HR (2014) Biomarkers of sepsis and their potential value in

diagnosis, prognosis and treatment. Expert Rev Clin Immunol 10:1349-1356.

Schmidt EP, Yang Y, Janssen WJ, Gandjeva A, Perez MJ, Barthel L, Zemans RL,

Bowman JC, Koyanagi DE, Yunt ZX, Smith LP, Cheng SS, Overdier KH,

Thompson KR, Geraci MW, Douglas IS, Pearse DB and Tuder RM (2012) The

pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury

during experimental sepsis. Nat Med 18:1217-1223.

Schneider AG, Goodwin MD and Bellomo R (2013) Measurement of kidney perfusion in

critically ill patients. Crit Care 17:220.

Seely KA, Holthoff JH, Burns ST, Wang Z, Thakali KM, Gokden N, Rhee SW and

Mayeux PR (2011) Hemodynamic changes in the kidney in a pediatric rat model

of sepsis-induced acute kidney injury. Am J Physiol Renal Physiol 301:F209-

F217.

Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DR,

McDonald-Smith GP, Gao H, Hennessy L, Finnerty CC, Lopez CM, Honari S,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

50

Moore EE, Minei JP, Cuschieri J, Bankey PE, Johnson JL, Sperry J, Nathens AB,

Billiar TR, West MA, Jeschke MG, Klein MB, Gamelli RL, Gibran NS, Brownstein

BH, Miller-Graziano C, Calvano SE, Mason PH, Cobb JP, Rahme LG, Lowry SF,

Maier RV, Moldawer LL, Herndon DN, Davis RW, Xiao W and Tompkins RG

(2013) Genomic responses in mouse models poorly mimic human inflammatory

diseases. Proc Natl Acad Sci USA 110:3507-3512.

Shah BA and Padbury JF (2014) Neonatal sepsis: an old problem with new insights.

Virulence 5:170-178.

Shahul S, Gulati G, Hacker MR, Mahmood F, Canelli R, Nizamuddin J, Mahmood B,

Mueller A, Simon BA, Novack V and Talmor D (2015) Detection of Myocardial

Dysfunction in Septic Shock: A Speckle-Tracking Echocardiography Study.

Anesth Analg 121:1547-1554.

Sims CR, MacMillan-Crow LA and Mayeux PR (2014) Targeting mitochondrial oxidants

may facilitate recovery of renal function during infant sepsis. Clin Pharmacol Ther

96:662-664.

Soerensen KE, Nielsen OL, Birck MM, Soerensen DB, Leifsson PS, Jensen HE,

Aalbaek B, Kristensen AT, Wiinberg B, Kjelgaard-Hansen M, Heegaard PM and

Iburg TM (2012) The use of sequential organ failure assessment parameters in

an awake porcine model of severe Staphylococcus aureus sepsis. APMIS

120:909-921.

Souza AC, Yuen PS and Star RA (2015) Microparticles: markers and mediators of

sepsis-induced microvascular dysfunction, immunosuppression, and AKI. Kidney

Int 87:1100-1108.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

51

Srinivasan L, Kirpalani H and Cotten CM (2015) Elucidating the role of genomics in

neonatal sepsis. Semin Perinatol 39:611-616.

Starr ME, Steele AM, Saito M, Hacker BJ, Evers BM and Saito H (2014) A new cecal

slurry preparation protocol with improved long-term reproducibility for animal

models of sepsis. PLoS One 9:e115705.

Starr ME, Takahashi H, Okamura D, Zwischenberger BA, Mrazek AA, Ueda J,

Stromberg AJ, Evers BM, Esmon CT and Saito H (2015) Increased coagulation

and suppressed generation of activated protein C in aged mice during intra-

abdominal sepsis. Am J Physiol Heart Circ Physiol 308:H83-89H81.

Stearns-Kurosawa DJ, Osuchowski MF, Valentine C, Kurosawa S and Remick DG

(2011) The Pathogenesis of Sepsis. Annu Rev Pathol 6:19-48.

Stevenson EK, Rubenstein AR, Radin GT, Wiener RS and Walkey AJ (2014) Two

decades of mortality trends among patients with severe sepsis: a comparative

meta-analysis*. Crit Care Med 42:625-631.

Stubbs DJ, Yamamoto AK and Menon DK (2013) Imaging in sepsis-associated

encephalopathy--insights and opportunities. Nat Rev Neurol 9:551-561.

Suetrong B and Walley KR (2015) Lactic acidosis in sepsis: It's not all anaerobic.

Implications for diagnosis and management. Chest in press.

Sun Z, Ye H, Shen X, Chao H, Wu X and Yang J (2014) Continuous venovenous

hemofiltration versus extended daily hemofiltration in patients with septic acute

kidney injury: a retrospective cohort study. Crit Care 18:R70.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

52

Supavekin S, Zhang W, Kucherlapati R, Kaskel FJ, Moore LC and Devarajan P (2003)

Differential gene expression following early renal ischemia/reperfusion. Kidney

Int 63:1714-1724.

Taccone FS, Su F, De Deyne C, Abdellhai A, Pierrakos C, He X, Donadello K, Dewitte

O, Vincent JL and De Backer D (2014) Sepsis is associated with altered cerebral

microcirculation and tissue hypoxia in experimental peritonitis. Crit Care Med

42:e114-122.

Talan DA (2015) Procalcitonin Is Not a Useful Biomarker of Sepsis. Ann Emerg Med

66:320-321.

Taylor FB, Jr., Kinasewitz GT and Lupu F (2012) Pathophysiology, staging and therapy

of severe sepsis in baboon models. J Cell Mol Med 16:672-682.

Torio CM and Andrews RM (2013) National Inpatient Hospital Costs: The Most

Expensive Conditions by Payer, 2011: Statistical Brief #160, in Healthcare Cost

and Utilization Project (HCUP) Statistical Briefs, Rockville (MD).

Tsalik EL, Willig LK, Rice BJ, van Velkinburgh JC, Mohney RP, McDunn JE, Dinwiddie

DL, Miller NA, Mayer ES, Glickman SW, Jaehne AK, Glew RH, Sopori ML, Otero

RM, Harrod KS, Cairns CB, Fowler VG, Rivers EP, Woods CW, Kingsmore SF

and Langley RJ (2015) Renal systems biology of patients with systemic

inflammatory response syndrome. Kidney Int 88:804-814.

Turnbull IR, Clark AT, Stromberg PE, Dixon DJ, Woolsey CA, Davis CG, Hotchkiss RS,

Buchman TG and Coopersmith CM (2009) Effects of aging on the

immunopathologic response to sepsis. Crit Care Med 37:1018-1023.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

53

Unsinger J, McGlynn M, Kasten KR, Hoekzema AS, Watanabe E, Muenzer JT,

McDonough JS, Tschoep J, Ferguson TA, McDunn JE, Morre M, Hildeman DA,

Caldwell CC and Hotchkiss RS (2010) IL-7 promotes T cell viability, trafficking,

and functionality and improves survival in sepsis. J Immumol 184:3768-3779.

Vincent JL, Opal SM, Marshall JC and Tracey KJ (2013) Sepsis definitions: time for

change. Lancet 381:774-775.

Wacker C, Prkno A, Brunkhorst FM and Schlattmann P (2013) Procalcitonin as a

diagnostic marker for sepsis: a systematic review and meta-analysis. Lancet

Infect Dis 13:426-435.

Wada H, Thachil J, Di Nisio M, Mathew P, Kurosawa S, Gando S, Kim HK, Nielsen JD,

Dempfle CE, Levi M, Toh CH and The Scientific Standardization Committee on

DICotISoTH (2013) Guidance for diagnosis and treatment of DIC from

harmonization of the recommendations from three guidelines. J Thromb Haemost

11:761-767.

Wald R, Adhikari NK, Smith OM, Weir MA, Pope K, Cohen A, Thorpe K, McIntyre L,

Lamontagne F, Soth M, Herridge M, Lapinsky S, Clark E, Garg AX, Hiremath S,

Klein D, Mazer CD, Richardson RM, Wilcox ME, Friedrich JO, Burns KE and

Bagshaw SM (2015) Comparison of standard and accelerated initiation of renal

replacement therapy in acute kidney injury. Kidney Int 88:897-904.

Wang B, Chen G, Zhang J, Xue J, Cao Y and Wu Y (2015a) Increased Neutrophil

Gelatinase-Associated Lipocalin is Associated with Mortality and Multiple Organ

Dysfunction Syndrome in Severe Sepsis and Septic Shock. Shock 44:234-238.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

54

Wang H and Ma S (2008) The cytokine storm and factors determining the sequence

and severity of organ dysfunction in multiple organ dysfunction syndrome. Am J

Emerg Med 26:711-715.

Wang K, Bhandari V, Giuliano JS, Jr., CS OH, Shattuck MD and Kirby M (2014)

Angiopoietin-1, angiopoietin-2 and bicarbonate as diagnostic biomarkers in

children with severe sepsis. PLoS One 9:e108461.

Wang L and Dudek SM (2009) Regulation of vascular permeability by sphingosine 1-

phosphate. Microvasc Res 77:39-45.

Wang Z, Sims CR, Patil NK, Gokden N and Mayeux PR (2015b) Pharmacologic

targeting of sphingosine-1-phosphate receptor 1 improves the renal

microcirculation during sepsis in the mouse. J Pharmacol Exp Ther 352:61-66.

Ward NS, Casserly B and Ayala A (2008) The compensatory anti-inflammatory

response syndrome (CARS) in critically ill patients. Clin Chest Med 29:617-625.

Ward PA (2012) New approaches to the study of sepsis. EMBO Mol Med 4:1234-1243.

Warren BL, Eid A, Singer P, Pillay SS, Carl P, Novak I, Chalupa P, Atherstone A,

Penzes I, Kubler A, Knaub S, Keinecke HO, Heinrichs H, Schindel F, Juers M,

Bone RC, Opal SM and KyberSept Trial Study G (2001) Caring for the critically ill

patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial.

J Am Med Assoc 286:1869-1878.

Weiss SL, Fitzgerald JC, Pappachan J, Wheeler D, Jaramillo-Bustamante JC, Salloo A,

Singhi SC, Erickson S, Roy JA, Bush JL, Nadkarni VM, Thomas NJ, Sepsis

Prevalence O, Therapies Study I, the Pediatric Acute Lung I and Sepsis

Investigators N (2015) Global epidemiology of pediatric severe sepsis: the sepsis

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

55

prevalence, outcomes, and therapies study. Am J Respir Crit Care Med

191:1147-1157.

Wepler M, Hafner S, Scheuerle A, Reize M, Groger M, Wagner F, Simon F, Matallo J,

Gottschalch F, Seifritz A, Stahl B, Matejovic M, Kapoor A, Moller P, Calzia E,

Georgieff M, Wachter U, Vogt JA, Thiemermann C, Radermacher P and McCook

O (2013) Effects of the PPAR-beta/delta agonist GW0742 during resuscitated

porcine septic shock. Intensive Care Med Exp 1:28.

Wheeler DS, Wong HR and Zingarelli B (2011) Pediatric Sepsis - Part I: "Children are

not small adults!". Open Inflamm J 4:4-15.

Winkler MS, Nierhaus A, Holzmann M, Mudersbach E, Bauer A, Robbe L, Zahrte C,

Geffken M, Peine S, Schwedhelm E, Daum G, Kluge S and Zoellner C (2015)

Decreased serum concentrations of sphingosine-1-phosphate in sepsis. Crit

Care 19:372.

Wong HR, Cvijanovich NZ, Anas N, Allen GL, Thomas NJ, Bigham MT, Weiss SL,

Fitzgerald J, Checchia PA, Meyer K, Shanley TP, Quasney M, Hall M, Gedeit R,

Freishtat RJ, Nowak J, Raj SS, Gertz S, Dawson E, Howard K, Harmon K, Lahni

P, Frank E, Hart KW and Lindsell CJ (2015a) A Multibiomarker-Based Model for

Estimating the Risk of Septic Acute Kidney Injury. Crit Care Med 43:1646-1653.

Wong HR, Cvijanovich NZ, Anas N, Allen GL, Thomas NJ, Bigham MT, Weiss SL,

Fitzgerald J, Checchia PA, Meyer K, Shanley TP, Quasney M, Hall M, Gedeit R,

Freishtat RJ, Nowak J, Shekhar RS, Gertz S, Dawson E, Howard K, Harmon K,

Beckman E, Frank E and Lindsell CJ (2015b) Developing a clinically feasible

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

56

personalized medicine approach to pediatric septic shock. Am J Respir Crit Care

Med 191:309-315.

Wong HR, Weiss SL, Giuliano JS, Jr., Wainwright MS, Cvijanovich NZ, Thomas NJ,

Allen GL, Anas N, Bigham MT, Hall M, Freishtat RJ, Sen A, Meyer K, Checchia

PA, Shanley TP, Nowak J, Quasney M, Chopra A, Fitzgerald JC, Gedeit R,

Banschbach S, Beckman E, Lahni P, Hart K and Lindsell CJ (2014) Testing the

prognostic accuracy of the updated pediatric sepsis biomarker risk model. PLoS

One 9:e86242.

Wood M, Song A, Maslove D, Ferri C, Howes D, Muscedere J and Boyd JG ((in press))

Brain Tissue Oxygenation in Patients with Septic Shock: a Feasibility Study. Can

J Neurol Sci.

Wu L, Gokden N and Mayeux PR (2007) Evidence for the role of reactive nitrogen

species in polymicrobial sepsis-induced renal peritubular capillary dysfunction

and tubular injury. J Am Soc Nephrol 18:1807-1815.

Wynn JL, Scumpia PO, Delano MJ, O'Malley KA, Ungaro R, Abouhamze A and

Moldawer KL (2007) Increased mortality and altered immunity in neonatal sepsis

produced by generalized peritonitis. Shock 28:675-683.

Wynn JL, Scumpia PO, Winfield RD, Delano MJ, Kelly-Scumpia K, Barker T, Ungaro R,

Levy O and Moldawer LL (2008) Defective innate immunity predisposes murine

neonates to poor sepsis outcome but is reversed by TLR agonists. Blood

112:1750-1758.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

57

Xiao Z, Wilson C, Robertson HL, Roberts DJ, Ball CG, Jenne CN and Kirkpatrick AW

(2015) Inflammatory mediators in intra-abdominal sepsis or injury - a scoping

review. Crit Care 19:373.

Yao X, Carlson D, Sun Y, Ma L, Wolf SE, Minei JP and Zang QS (2015) Mitochondrial

ROS Induces Cardiac Inflammation via a Pathway through mtDNA Damage in a

Pneumonia-Related Sepsis Model. PLoS One 10:e0139416.

Yini S, Heng Z, Xin A and Xiaochun M (2015) Effect of unfractionated heparin on

endothelial glycocalyx in a septic shock model. Acta Anaesthesiol Scand 59:160-

169.

Zang QS, Sadek H, Maass DL, Martinez B, Ma L, Kilgore JA, Williams NS, Frantz DE,

Wigginton JG, Nwariaku FE, Wolf SE and Minei JP (2012) Specific inhibition of

mitochondrial oxidative stress suppresses inflammation and improves cardiac

function in a rat pneumonia-related sepsis model. Am J Physiol Heart Circ

Physiol 302:H1847-H1859.

Zanotti-Cavazzoni SL and Goldfarb RD (2009) Animal models of sepsis. Crit Care Clin

25:703-719.

Zeng Y, Adamson RH, Curry FR and Tarbell JM (2014) Sphingosine-1-phosphate

protects endothelial glycocalyx by inhibiting syndecan-1 shedding. Am J Physiol

Heart Circ Physiol 306:H363-H372.

Zhao X, Chen YX and Li CS (2015) Predictive value of the complement system for

sepsis-induced disseminated intravascular coagulation in septic patients in

emergency department. J Crit Care 30:290-295.

Ziaja M (2013) Septic encephalopathy. Curr Neurol Neurosci Rep 13:383.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

58

Ziegler T, Horstkotte J, Schwab C, Pfetsch V, Weinmann K, Dietzel S, Rohwedder I,

Hinkel R, Gross L, Lee S, Hu J, Soehnlein O, Franz WM, Sperandio M, Pohl U,

Thomas M, Weber C, Augustin HG, Fassler R, Deutsch U and Kupatt C (2013)

Angiopoietin 2 mediates microvascular and hemodynamic alterations in sepsis. J

Clin Invest 123:3436-3445.

Zonneveld R, Martinelli R, Shapiro NI, Kuijpers TW, Plotz FB and Carman CV (2014)

Soluble adhesion molecules as markers for sepsis and the potential

pathophysiological discrepancy in neonates, children and adults. Crit Care

18:204.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

59

Footnotes

This work was supported by the National Institutes of Health National Institute of

Diabetes and Digestive Kidney Diseases [F31 DK104533]; and National Institutes of

Health Institute of General Medical Sciences [R01 GM106419, GM112806]; the National

Institutes of Health National Center for Advancing Translational Sciences Clinical and

Translational Science Award [Grant UL1-TR000039].

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

JPET #230797

60

Figure Legends

Fig. 1. Conceptual framework for the use of discussed biomarkers in combination to

direct the development, validation and use of targeted therapies.

Fig. 2. Age, genetics, environmental factors, and comorbidities contribute to the

diversity of septic patients, with the young and the elderly populations being the most

susceptible to sepsis. As the systemic inflammatory response syndrome (SIRS) and

sepsis progress to severe sepsis with multiple organ dysfunction syndrome (MODS),

biomarkers could be used to guide targeted therapy that would halt the progression of

sepsis to increase survival and decrease morbidity.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

Inflammatory Biomarkers E-, L-, P-selectin ICAM-1, VCAM-1 CRP, PCT TNF-α IL-1, IL-6, IL-8, IL-27 HLA-DR PD-1 Lymphopenia Increased percentage of T

regulatory cells

Endothelial Biomarkers Syndecan-1 Heparan Sulfate Ang-1, Ang-2 Tie-2 S1P Microparticles

Coagulation Biomarkers Prothrombin Time Activated Partial

Thromboplastin Time D-dimer Platelet Count VWF Ultralarge VWF Multimers Fibrinogen ADAMTS-13

Organ-Directed and Physiological Biomarkers Brain:

Changes in EEG Cognitive Impairment Perfusion (NIRS)

Heart: Myocardial Dysfunction

(Echocardiography) Troponin

Kidney:

Perfusion (Ultrasound, MRI) Creatinine Clearance NGAL, KIM-1, NAG,

Liver:

ALT, AST Lung:

Ratio of partial pressure of O2 to fractional inspired O2

Microcirculation:

Blood Lactate Sublingual Perfusion Mixed venous O2 saturation

Tissue Injury: MMP-8, Extracellular Histones

Targeted Therapies Immunomodulation

Endothelium Microvasculature

Coagulation Organ-specific

Figure 1

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on February 8, 2016 as DOI: 10.1124/jpet.115.230797

at ASPE

T Journals on July 12, 2018

jpet.aspetjournals.orgD

ownloaded from

Definitions

Systemic Inflammatory Response Syndrome (SIRS): Two or more of the following:

•  fever or hypothermia •  tachycardia •  tachypnea •  leukocytosis or leukopenia

Sepsis: SIRS with a known infection

Severe Sepsis: Sepsis with 1 organ dysfunction or multiple organ dysfunction syndrome (MODS)

Sepsis

Severe Sepsis

SIRS

Death

Decreased Morbidity

Young (< 1 yr) Immature Immune System and Organ Development

Old (> 65 yrs) Weakened Immune System

and Comorbidities

Increased Survival

Biomarker-DirectedTherapy

Biomarker-DirectedTherapy

Biomarker-DirectedTherapy

Figure 2