You are on page 1of 10

Advances in Sepsis

R e s e a rc h
Peter Bentzer, MD, PhDa,b,c, James A. Russell, MDa,b, Keith R. Walley, MDa,b,*

KEYWORDS
 Sepsis  Innate immune receptors  Pathogen  Clearance  Edema  Vascular leak

KEY POINTS
 Treatment strategies targeting early pathophysiological alterations in sepsis improve outcomes.
 Pathogen toxins are triggers of inflammation in sepsis, and therapies aimed at enhancing toxin
clearance represent potential complements to early antimicrobial therapy.
 The innate immune response is an early built-in host response to infecting pathogens. Treatments
aimed at modifying this response have therapeutic potential.
 Vascular leak, secondary edema formation, and hypovolemia contribute to poor outcome in sepsis;
several distinct pathophysiological mechanisms may be targeted to ameliorate this response.

INTRODUCTION Herein the authors consider aspects of the sep-


tic inflammatory response that provide novel tar-
Of the World Health Organization top 10 causes of gets for therapeutic intervention. Specifically, (1)
death, 4 fulfill the definition of sepsis (www.who. clearance of inflammatory pathogen molecules
int). Sepsis occurs when infection results in sys- from the circulation, (2) modulation of innate im-
temic inflammation1,2 and is termed severe sepsis mune receptors and intracellular signaling, as
when accompanied by new organ dysfunction. well as (3) vascular leak are considered.
The 28-day mortality rate of severe sepsis is 15%
to 30%,3–6 which increases to 20% to 60% when PATHOGENS AND PATHOGEN TOXINS
complicated by arterial vasodilation and ventricu-
lar dysfunction, leading to septic shock.7 The num- Targeting the host septic inflammatory response
ber of deaths due to severe sepsis and septic (eg, anti–tumor necrosis factor,9 activated protein
shock is greater than the number of deaths due C [APC]5) has not worked in more than 30 phase
to acute myocardial infarction, even in the Western 3 randomized controlled trials (RCTs).10–12 In
world,3 and continues to increase.4 Effective treat- contrast, simply targeting the pathogen by source
ment of severe sepsis can lead to complete resolu- control (draining the abscess, etc) and early
tion with no sequelae, whereas ineffective broad-spectrum antibiotics is very effective.
treatment is fatal or leads to long-term morbidities Kumar and colleagues13 found that for every hour
and increased long-term mortality rates; so timely delay in antibiotic administration after onset of
effective therapy is crucial.8 septic shock, mortality increased by 7%. Another

Disclosure statement: P. Bentzer has no competing interests to declare. K.R. Walley and J.A. Russell have
founded Cyon Therapeutics, which has licensed intellectual property from the University of British Columbia
chestmed.theclinics.com

related to PCSK9 in sepsis.


a
Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada;
b
Division of Critical Care Medicine, St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street,
Vancouver, British Columbia V6Z 1Y6, Canada; c Department of Anesthesiology and Intensive Care, Lund Uni-
versity, Lund SE-221 85, Sweden
* Corresponding author. St. Paul’s Hospital, University of British Columbia, 1081 Burrard Street, Vancouver,
British Columbia V6Z 1Y6, Canada.
E-mail address: keith.walley@hli.ubc.ca

Clin Chest Med 36 (2015) 521–530


http://dx.doi.org/10.1016/j.ccm.2015.05.009
0272-5231/15/$ – see front matter Ó 2015 Elsevier Inc. All rights reserved.
522 Bentzer et al

less-established approach targeting the pathogen signaling. LBP more effectively transfers LPS
is to enhance clearance of toxins. Whether alive from bacterial cell wall fragments to lipoproteins
or dead, pathogens elicit an inflammatory than PLTP.
response by release of toxins such as lipopolysac- Following binding by transfer proteins, LPS is
charide (LPS) from gram-negative bacteria; the transferred to and sequestered within HDL and,
structurally similar glycolipid, lipoteichoic acid after LBP and PLTP-facilitated transfer, within
(LTA) from gram-positive bacteria; and other glyco- low-density lipoprotein (LDL) and very-low-
lipids such as phospholipomannan (PLM) from density lipoprotein (VLDL).23 LPS, LTA, and even
fungal pathogens.14 These toxins bind to innate im- the LPS lipid A analog, eritoran,24 are distributed
mune receptors (see section on Innate Immune primarily on HDL (w60%) with the remainder on
Signaling Induced by Pathogen Toxins) to trigger LDL and VLDL particles. In sepsis, this distribution
a septic inflammatory response. Accordingly, shifts from the predominant HDL carriage of LPS
clearance of pathogen toxins is an important, to increased LDL and VLDL carriage of LPS.
underrecognized, and modifiable aspect of sepsis
management that complements effective antimi- Subsequent Clearance of Pathogen Toxins
crobial therapy. from the Circulating Lipoprotein
Compartment
Pathogen Toxins Induce an Inflammatory
Pathogen toxins are then cleared primarily by he-
Response
patic uptake and excretion in bile.25 Characteriza-
Septic shock is primarily due to release of path- tion of the exact mechanisms of hepatic clearance
ogen toxins.8 The lipid A domain of LPS binds of pathogen toxins is limited but likely involves the
Toll-like receptor 4 (TLR4) expressed on many LDL receptor (LDLR). LDLR knockout mice have
cell lines, thereby inducing nuclear factor (NF)-kB increased mortality after cecal ligation and punc-
signaling and the subsequent proinflammatory ture compared with mice with intact LDLR26 sug-
and antiinflammatory cytokine response.15 LTA gesting a central LDLR role in pathogen toxin
of gram-positive bacterial cell membranes is clearance.
composed of a polyglycerolphosphate chain con- Further support for the role of LDLR in pathogen
nected by a glycolipid moiety16 and binds TLR2 toxin clearance is provided by recent observations
and TLR6 to induce NF-kB signaling.17 Fungal that PCSK9 decreases LPS clearance and in-
PLM is composed primarily of C24 hydroxy fatty creases the innate immune response to pathogen
acids linked to phytoceramide and phytosphingo- toxins.27 Circulating PCSK9, produced primarily
sine, with a hydrophilic polysaccharide domain by the liver, binds LDLR expressed on hepatocytes
consisting of mannose residues.18 PLM and and, upon internalization of the LDLR complex, tar-
related glycolipids are ligands for TLR2, TLR4, gets it for lysosomal degradation so that it is not re-
and TLR6. cycled back to the hepatocyte cell surface (Fig. 1).
Thus, increased PCSK9 activity decreases LDLR
Sequestration as the Initial Step in Limiting expression of hepatocytes, whereas reduced
the Adverse Effects of Pathogen Toxins PCSK9 activity results in increased LDLR expres-
sion and increases pathogen toxin clearance, de-
Pathogen toxins in the aqueous phase are quickly
creases cytokine inflammatory response, and
bound by transfer proteins that bind lipid moi-
improves survival from sepsis. This effect is lost
eties.19 When transfer protein availability is limited,
in LDLR knockout in mice and by an LDLR genetic
LPS incorporation into lipoproteins is reduced and
variant that interferes with binding of PCSK9 to the
LPS toxicity is increased, thus transfer proteins are
LDLR.27 Taken together, these data support the
the first step in sequestering pathogen toxins.
hypothesis that pathogen toxin clearance depends
LPS binding protein (LBP) and bactericidal
substantially on PCSK9 and the LDLR.
permeability-increasing protein are homologous
to the endogenous lipid transfer protein phospho-
How This Knowledge May Lead to Innovative
lipid transfer protein (PLTP), and all bind pathogen
Therapeutic Strategies
toxins.19 LBP and PLTP bind LPS and transfer it
from micelles or from LPS aggregates in the Mechanisms of pathogen toxin clearance may be
aqueous phase to high-density lipoprotein novel targets for treatment. PCSK9 inhibition to in-
(HDL)20,21 and other lipoproteins.22 While LBP crease pathogen toxin clearance conceivably
can bind and additionally transfer LPS to CD14, could decrease the inflammatory response and
a cofactor in subsequent signaling via TLR4, improve survival in human sepsis. PCSK9 inhibition
PLTP does not transfer LPS to CD1420 and, thus, targets the pathogen and like antibiotics, may be
does not trigger downstream inflammatory particularly effective.
Advances in Sepsis Research 523

Fig. 1. Endocytosis of the LDLR/LDL cholesterol (LDL-C) complex in the presence and absence of PCSK9.

INNATE IMMUNE SIGNALING INDUCED BY recognizes single-stranded viral RNA, and TLR9
PATHOGEN TOXINS recognizes unmethylated CpG bacterial DNA.29
Innate Immune Receptors DAMPs have been identified for many TLRs
including heat shock protein 60 (HSP60), HSP70,
Innate immune receptors, including TLRs, nucleo-
and Gp96. Endogenous ligands for TLR4 include
tide-binding oligomerization domain (NOD)-like re-
fibrinogen28 the fibronectin EDA (extra domain A)
ceptors (NLRs), and retinoic acid-inducible gene
domain, and hyaluronan. Endogenous mRNA is a
(RIG)-like receptors (RLRs), contribute to the
DAMP for TLR3.30,31
myocardial, and other organ systems’, response
to danger signals. TLRs, NLRs, and RLRs are NOD-like receptors
pattern recognition receptors that respond to NLRs can be grouped into 4 NOD receptors, which
exogenous ligands for pathogen-associated mo- induce a proinflammatory response via NF-kB,
lecular patterns (PAMPs). Potentially more impor- and 14 NLR pyrin domain-containing receptors
tant, innate immune receptors also respond to plus several NLR caspase recruitment domain-
endogenous ligands, damage-associated molecu- containing receptors,32,33 which assemble the in-
lar patterns (DAMPs). flammasome. Inflammasomes involve multiple
proteins, notably including caspases. The assem-
Toll-like receptors bled inflammasome can then be activated by
TLRs are type I transmembrane receptors consist- PAMPs and by endogenous activators (low intra-
ing of extracellular leucine-rich repeats linked to a cellular K1 levels and reactive oxygen species).
cytoplasmic Toll/interleukin-1 receptor (TIR) ho- Subsequent inflammasome signaling involves
mology domain.28 Ten or more TLRs respond to caspase-mediated cleavage leading to processing
endogenous and exogenous danger signals28 and secretion of mature interleukin (IL)-1b and IL-
(Table 1). TLRs 1, 2, 4, 5, and 6 are expressed 18.32 NLRs mediate tissue injury by their IL-1b
on the cell surface. The response of TLR4 to and IL-18 signatures.32 Compared with TLRs,
gram-negative bacterial cell wall molecules such much less is known about NLRs.
as LPS is best understood. TLR2 in concert with
TLR1 or TLR6 recognizes bacterial cell wall com- Retinoic acid-inducible gene-like receptors
ponents such as LTA, peptidoglycan, and yeast RLRs are recently discovered cytosolic RNA heli-
cell wall components.28 TLR5 binds gram- cases that act as innate immune receptors
negative bacterial flagellin. TLRs 3, 7, 8, and 9 in sensing viral dsRNA by recognizing 50 -triphos-
are expressed mainly intracellularly; their predom- phate ends, which characterize nonself RNA.34
inantly nucleic acid ligands must be internalized RLRs include RIG-1, melanoma differentiation-
into endosomes to initiate signaling.28 TLR3 rec- associated protein 5, and LGP2 (Laboratory of Ge-
ognizes double-stranded (ds) viral RNA, TLR7 netics and Physiology). RIG-1 results in a type I
524
Bentzer et al
Table 1
Summary of endogenous and exogenous TLR receptor ligands described in humans

Intracellular
Signaling
Receptor Cell Types Localization Endogenous Ligands Exogenous Ligands Pathway Cellular Response
TLR1 1 TLR2 B lymphocytes Cell surface b-Defensin, biglycan, HSP, G (1) lipoproteins, LTA, MyD88 Cytokine production
Dendritic cells, monocytes/ HMGB1, hyaluronic acid fungal cell wall
macrophages components (PLM)
TLR2 1 TLR6 B lymphocytes Cell surface b-Defensin, biglycan, HSP, G (1) lipoproteins, LTA, MyD88 Cytokine production
Dendritic cells, monocytes/ HMGB1, hyaluronic acid fungal cell wall
macrophages components (PLM)
TLR3 B lymphocytes Endosomes/ mRNA Double-stranded viral TRIF Cytokine production
Dendritic cells lysosomes RNA Type 1 IFN
TLR4 B lymphocytes, dendritic cells, Cell surface b-Defensin, fibronectin, G () cell wall components MyD88/TRIF Cytokine production
monocytes/macrophages hyaluronic acid, heparan for LPS with MD2 as a Type 1 IFN
Neutrophils, intestinal sulfate, fibrinogen, coreceptor, fungal cell
epithelium HMGB1, S100, oxidized wall components (PLM
LDL and similar compounds)
TLR5 Dendritic cells, monocytes/ Cell surface — Flagellin MyD88 Cytokine production
macrophages, intestinal
epithelium
TLR7 B lymphocytes, dendritic cells, Endosomes/ ssRNA ss Viral RNA MyD88 Cytokine production
monocytes/macrophages lysosomes Type 1 IFN
TLR8 Dendritic cells, mast cells, Endosomes/ ssRNA — MyD88 Cytokine production
monocytes/macrophages lysosomes Type 1 IFN
TLR9 B lymphocytes, dendritic cells, Endosomes/ IgG chromatin complex Unmethylated CpG DNA MyD88 Cytokine production
monocytes/macrophages lysosomes Type 1 IFN

Abbreviations: G (), gram negative; G (1), gram positive; HMGB1, high-mobility group box-1 protein; HSP, heat shock protein; IgG, immunoglobulin G; MD2, myeloid differentiation
factor 2; mRNA, messenger RNA; MyD88, myeloid differentiation primary response gene 88; ss, single stranded; TRIF, TIR domain-containing adapter-inducing interferon-b; type 1 IFN,
type 1 interferon.
Advances in Sepsis Research 525

interferon (IFN) response by signaling via interferon PAMP-induced danger signals with complex inflam-
regulatory factor 3 (IRF3) and IRF7. IRF3 and IRF7 matory and functional responses.38–51 In response
form homodimers and heterodimers, which form to inflammatory stimuli, cardiomyocytes express
part of the transcription enhancer complex for IFN- proinflammatory and antiinflammatory cytokines
b in the nucleus. RIG-1 also can signal via NF-kB.35 (eg, IL-6, IL-10), which initiate a local inflammatory
response, recruiting inflammatory cells necessary
Nuclear factor-kB and alternative signaling for repair, and increase expression of cell surface
pathways adhesion molecules (eg, intercellular adhesion
Activation of most TLRs by ligand binding results in molecule [ICAM-1]) allowing interaction and
dimerization, a change in conformation, and recruit- outside-in signaling from participating inflammatory
ment of the adaptor protein myeloid differentiation cells and the extracellular matrix.43,45,50–53 ICAM-1
primary response gene 88 (MyD88) to the cyto- expression and subsequent ligand binding
plasmic TIR domain.36 MyD88 recruits IRAK4 (inter- decrease cardiomyocyte contractility by involving
leukin-1 receptor-associated kinase), which, with calcium channel-binding proteins S100A8/A9.54
participation of TAK1 binding protein (TAB2) and This complex response starts with binding of path-
TRAF6, leads to activation of TGF-Beta-Activated Ki- ogen molecules to innate immune receptors.
nase (TAK1). TAK1 activates IkB kinase (IKK) leading
to liberation of NF-kB from its inhibitor, IkBa (nuclear How This Knowledge May Lead to Innovative
factor of kappa light polypeptide gene enhancer in Therapeutic Strategies
B-cells inhibitor, alpha), and subsequent transloca-
tion of NF-kB to the nucleus where it induces proin- Sepsis therapies are most beneficial if initiated
flammatory cytokine production. NF-kB is a critical early, and the innate immune response (involves
mediator of several inflammatory pathways. all tissues) is an early host response. Thus modifying
All TLRs except TLR3 signal via MyD88 to the innate immune response could ameliorate mul-
induce NF-kB activity. Increased NF-kB activity tiple organ system dysfunction. Conceivably, early
leads to different responses in different cell types, administration of therapies aimed at modulation of
although all these responses are part of the com- innate immunity may be beneficial. Thus innate im-
plex inflammatory response that PAMPs initiate. munity pathways are reasonable targets to consider
Other TLR-initiated pathways have been identi- as new therapeutic strategies for sepsis.
fied. TIR domain-containing adapter-inducing
interferon-b (TRIF), like MyD88, is an alternate VASCULAR LEAK
adaptor protein that binds to the TIR domain of Fluid Homeostasis in Sepsis
TLR3 and TLR4.37 TRIF interacts with TRAF6, Hypovolemia secondary to vascular leakage is an
RIP1, and TAK1, leading to activation of NF-kB important contributor to hemodynamic instability
and IRF3. TAK1 can also activate mitogen-acti- in sepsis and often leads to large-volume fluid
vated protein kinase kinase (MKK3/6) and MKK7, resuscitation with ensuing edema formation and
which signal via inflammatory gene transcription organ dysfunction. Observational studies show an
factors p38 and JNK (c-Jun N-terminal kinase). association between fluid overload and outcome,55
NOD1 and NOD2 also signal via NF-kB indepen- and RCTs suggest benefit from restrictive fluid
dent of MyD88. Ligation of these NLRs results in administration.56 Experimental studies showing
activation of RIP2, which interacts with IKKg to that interventions specifically targeting vascular
allow release of NF-kB and translocation into the leakage increases survival without affecting
nucleus.32,33 RIP2 alternatively signals via TAK1, markers of inflammatory response further support
TAB1, and TAB2/3 to activate the p38 MAPK that vascular leakage is a determinant of outcome
(mitogen-activated protein kinase) signaling and not simply a marker of injury severity.57
pathway. NLR inflammasomes share caspase Transvascular fluid transport depends on trans-
signaling pathways.32 capillary hydrostatic and colloid osmotic pres-
sures as well as permeability for fluid and
An Example of Septic Organ Dysfunction:
macromolecules. Extravasation of macromole-
Proinflammatory and Antiinflammatory
cules (eg, albumin) occurs by both diffusion and
Responses in the Heart
convection; thus, changes in both permeability
Cardiomyocytes’ primary function is contraction to and transcapillary hydrostatic pressure are of cen-
provide the motive force that drives cardiac output tral importance for this process. In most organs,
and generates arterial pressure. Cardiomyocytes hydrostatic and osmotic pressures are unbal-
have key additional properties, analogous to innate anced and net fluid filtration from circulation to tis-
immune antigen-presenting cells (eg, dendritic cells, sue occurs. Normally, approximately 5% of the
tissue macrophages). Cardiomyocytes respond to intravascular albumin leaves the circulation every
526 Bentzer et al

hour and is returned to the circulation via the volume in whole animal models of sepsis, and
lymph. Inflammation-induced changes in the inter- drugs approved for human use are available.
stitial matrix and lymphatic function are potentially
important contributors to hypovolemia in sepsis.
Vascular Leak and the Vasopressin Axis in
However, most studies investigating disruptions
Sepsis
in fluid homeostasis in sepsis have focused on
pathways involved in increases in permeability The initial phase of sepsis is characterized by vaso-
such as the vascular endothelial growth factor as dilatation secondary to increased production of
well as the angiopoietin and thrombin pathways.58 vasodilatory substances (nitric oxide, prostacy-
In the following discussion, 2 other mechanisms clin), decreased production of vasoconstrictors
and pathways, the sphingosine-1-phosphate (vasopressin), changed expression of vasocon-
(S1P) and vasopressin systems, are highlighted strictive receptors,59 and disruption of myogenic
as potential therapeutic targets in sepsis. Both control mechanisms responsible for autoregulation
systems have been shown to influence plasma of capillary hydrostatic pressure60 (Fig. 2). If

Fig. 2. Mechanisms involved in sepsis induced vascular leak. Vasodilation is partly due to changes in concentra-
tion of vasoconstricting agents such as vasopressin (Vaso) and decreased expression of vasopressin receptors
(V1aR) on smooth muscle cells as well as vasodilators such as nitric oxide. Precapillary arteriolar vasodilation in-
creases capillary pressure. Decreased myogenic activity causes a change in arterial pressure that is transferred
to the capillaries, the major site of vascular fluid exchange. Vascular endothelial permeability is increased
through multiple mechanisms acting on intercellular junctions of endothelial cells. For example, decreased avail-
ability of spinogosine-1-phosphate (S1P) through decreased plasma levels of apolipoprotein M (ApoM) and albu-
min (Alb) will decrease permeability via decreased stimulation of S1P1 receptors (S1P1 in the figure). Decreased
levels of activated protein C (APC) increase permeability through decreased endothelial synthesis of S1P because
of decreased activation of the endothelial protein C receptor (EPCR)/protease-activated receptor (PAR)-1 receptor
complex. S1P also increases permeability through activation of endothelial S1P3 receptors (S1P3 in the figure),
but the importance of this pathway in sepsis is unclear. Sepsis-induced inflammation disrupts the interstitial ma-
trix and transiently decreases interstitial pressure, which may contribute to early leak of fluid.
Advances in Sepsis Research 527

hypotension persists after fluid resuscitation, Sur- concentration gradient is important for mainte-
viving Sepsis guidelines61 recommend norepi- nance of normal barrier function. In plasma, about
nephrine to maintain mean arterial pressure 60% of S1P is bound to apolipoprotein M (ApoM)
above 65 mm Hg. However, norepinephrine may (in HDL particles) and about 35% is bound to
cause fluid filtration and decrease plasma volume albumin.
in inflammatory conditions.62,63 A tentative expla- Maintenance of normal endothelial barrier func-
nation of norepinephrine’s effect is increased tion by S1P is mediated by S1P bound to both
capillary pressure because of a change in the pre- ApoM and albumin.67,68 S1P synthesis and release
capillary to postcapillary resistance ratio.64 These from endothelial cells can be initiated through acti-
observations raise the question whether other va- vation of protease-activated receptor-1 by a com-
soconstrictors may have a more beneficial effect plex of APC bound to the endothelial protein C
on fluid balance. receptor.69 The therapeutic potential of S1P/
Vasopressin is recommended as a second-line S1P1 is supported by studies showing that admin-
treatment in septic shock. Vasopressin elicits vaso- istration of S1P (or structurally similar drugs such
constriction through the V1a receptor on smooth as the precursor analog FTY 720) counteracts
muscle cells and has V2-induced antidiuretic and vascular leakage and hypovolemia in experimental
procoagulant actions through stimulation of renal sepsis.70,71 Clinical application may be limited by a
and endothelial V2 receptors. Recently, animal narrow therapeutic window above which increased
studies have reported potentially important effects vascular leakage through S1PR2 (S1P receptor)
on vascular leakage of vasopressin derivatives and S1PR3 stimulation may be seen. Furthermore,
highly selective for V1a receptors (eg, selepressin). the immunomodulatory effects of nonselective
Selepressin reduced fluid requirements and pro- S1PR agonist may limit efficacy and safety. Conse-
tected against vascular leak when compared with quently, more specific S1PR1 receptor agonists or
noradrenalin or vasopressin.65,66 A preliminary alternative approaches to increase S1PR signaling
report from a phase 2 trial also suggested a dose- could be of interest. More specific agonists (FTY
dependent limitation of positive fluid balance by se- 720 (S)-phosphate, CYM 5452, SEW2871) show
lepressin compared with placebo in septic shock. promising results in models of inflammation-
The mechanisms of vascular leakage protection induced lung and renal permeability.72,73 Nonanti-
by V1a agonism in sepsis is not known. coagulant APC derivatives increase survival and
V1a receptors are generally not expressed on decease vascular leakage in sepsis models and
endothelial cells; perhaps the beneficial effect of offer an alterative approach to decease vascular
selective V1a agonism on vascular leakage is leakage through the S1PR1 pathway.74 The obser-
mediated via a more favorable precapillary to vation that ApoM is the main carrier of S1P together
postcapillary resistance ratio, decreased capillary with the observation that ApoM level is decreased
hydrostatic pressure, and less vascular leakage in sepsis suggests that S1P’s barrier protective ef-
compared with norepinephrine. Large RCTs are fects may be enhanced by increasing plasma
needed to determine whether selective V1a recep- levels of ApoM in sepsis.75
tor agonism improves outcomes of septic shock.
How This Knowledge May Lead to Innovative
Therapeutic Strategies
Sphingosine-1-Phosphate and Endothelial
Permeability Disturbances in vascular leakage, fluid homeosta-
sis with ensuing hypovolemia, and edema forma-
S1P is a lipid with multiple functions including im-
tion are important for outcome in sepsis. The
mune modulation and maintenance of endothelial
vasopressin and S1P systems are promising tar-
barrier function. S1P binds to a series of G-pro-
gets to minimize vascular leakage; selective stim-
tein-coupled receptors, S1P receptors 1–5;
ulation of V1a receptors and S1P1 receptors holds
endothelial S1P receptor 1 (S1P1) decreases
therapeutic promise. The importance of the inter-
permeability primarily through AKT-1 and Rac-1
stitial matrix and the lymphatic system for hypovo-
activation with the endothelial cytoskeleton and
lemia and edema formation in sepsis represents a
eNOS (endothelial nitric oxide synthase) as down-
new frontier for sepsis research.
stream targets. S1P2 and S1P3 increase
permeability through RHO kinase activation.
Why Have Most Sepsis Trials Failed?
Erythrocytes are the major source of S1P in
plasma, and most cell types have the ability to syn- Most pivotal phase 3 RCTs in sepsis have failed
thesize S1P. The concentration of S1P is higher in despite strong preclinical evidence and promising
plasma than in lymph, which in turn is higher than phase 2 RCTs. Why have so many RCTs
in the interstitium, and maintenance of this S1P missed the mark? There are several possible
528 Bentzer et al

explanations. First, the drug targets are enmeshed analysis of incidence, outcome, and associated
in redundant pathways that respond to drug inter- costs of care. Crit Care Med 2001;29:1303–10.
vention to maintain the disorder of sepsis. Second, 4. Angus DC, Pereira CA, Silva E. Epidemiology of se-
the preclinical models may not be appropriate in vere sepsis around the world. Endocr Metab Im-
modeling human sepsis because most are simple, mune Disord Drug Targets 2006;6:207–12.
acute models in young animals without comorbid- 5. Bernard GR, Vincent JL, Laterre PF, et al. Efficacy and
ities, the opposite of many patients with sepsis. safety of recombinant human activated protein C for
Third, the phase 2 RCTs are often small severe sepsis. N Engl J Med 2001;344:699–709.
(n 5 100–300), considerably smaller than phase 6. Vincent JL, Sakr Y, Sprung CL, et al. Sepsis in Euro-
2 RCTs in other conditions such as heart disease. pean intensive care units: results of the SOAP study.
Fourth, sepsis is a heterogeneous condition that Crit Care Med 2006;34:344–53.
may be composed of responders and nonre- 7. Russell JA, Walley KR, Singer J, et al. Vasopressin
sponders according to genotype, gene expres- versus norepinephrine infusion in patients with sep-
sion, and clinical variables. Finally, the underlying tic shock. N Engl J Med 2008;358:877–87.
mortality of sepsis has decreased to 20% to 8. Russell JA. Management of sepsis. N Engl J Med
30% because of earlier intervention with fluids, va- 2006;355:1699–713.
sopressors, and antibiotics; thus, many RCTs are 9. Morris PE, Zeno B, Bernard AC, et al. A placebo-
underpowered for these now lower mortality rates. controlled, double-blind, dose-escalation study to
So the authors suggest that future programs assess the safety, tolerability and pharmacoki-
should include more appropriate preclinical netics/pharmacodynamics of single and multiple
models, larger phase 2 RCTs, predictive bio- intravenous infusions of AZD9773 in patients with se-
markers to test a more homogeneous patient sam- vere sepsis and septic shock. Crit Care 2012;16:R31.
ple, and composite outcome variables (eg, 10. Dubois MJ, Vincent JL. Clinically-oriented therapies
measures of organ dysfunction). in sepsis: a review. J Endotoxin Res 2000;6:463–9.
11. Eichacker PQ, Parent C, Kalil A, et al. Risk and the
efficacy of antiinflammatory agents: retrospective
SUMMARY
and confirmatory studies of sepsis. Am J Respir
Sepsis is a disease with a high short- and long-term Crit Care Med 2002;166:1197–205.
mortality and morbidity. In this review, aspects of 12. Vincent JL, Sun Q, Dubois MJ. Clinical trials of
the septic inflammatory response that provide immunomodulatory therapies in severe sepsis and
promising novel targets for therapeutic interven- septic shock. Clin Infect Dis 2002;34:1084–93.
tions are considered. Increased clearance of the 13. Kumar A, Roberts D, Wood KE, et al. Duration of hy-
bacterial toxin LPS by inhibition of PCSK9 suggests potension before initiation of effective antimicrobial
that clearance of pathogen toxins complements therapy is the critical determinant of survival in hu-
source control and early broad-spectrum antibi- man septic shock. Crit Care Med 2006;34:1589–96.
otics. The innate immune response is a key compo- 14. Trinel PA, Plancke Y, Gerold P, et al. The Candida al-
nent in host defense, and signaling pathways (eg, bicans phospholipomannan is a family of glycolipids
TLRs) are potential therapeutic targets in sepsis. presenting phosphoinositolmannosides with long
The observations that positive fluid balance is linear chains of beta-1,2-linked mannose residues.
associated with poor outcome and that treatment J Biol Chem 1999;274:30520–6.
strategies to limit fluid overload improve outcome 15. Gallay P, Heumann D, Le Roy D, et al. Mode of ac-
suggest that disturbances in vascular leakage tion of anti-lipopolysaccharide-binding protein anti-
and fluid homeostasis are determinants of bodies for prevention of endotoxemic shock in
outcome in sepsis rather than simply markers of mice. Proc Natl Acad Sci U S A 1994;91:7922–6.
severity. The vasopressin and S1P pathways are 16. Reichmann NT, Grundling A. Location, synthesis
promising targets to modulate vascular leakage. and function of glycolipids and polyglycerolphos-
phate lipoteichoic acid in gram-positive bacteria of
the phylum Firmicutes. FEMS Microbiol Lett 2011;
REFERENCES
319:97–105.
1. Bone RC. The sepsis syndrome. Definition and gen- 17. Kimbrell MR, Warshakoon H, Cromer JR, et al. Com-
eral approach to management. Clin Chest Med parison of the immunostimulatory and proinflamma-
1996;17:175–81. tory activities of candidate gram-positive endotoxins,
2. Levy MM, Fink MP, Marshall JC, et al. 2001 SCCM/ lipoteichoic acid, peptidoglycan, and lipopeptides,
ESICM/ACCP/ATS/SIS international sepsis defini- in murine and human cells. Immunol Lett 2008;118:
tions conference. Crit Care Med 2003;31:1250–6. 132–41.
3. Angus DC, Linde-Zwirble WT, Lidicker J, et al. 18. Trinel PA, Maes E, Zanetta JP, et al. Candida albi-
Epidemiology of severe sepsis in the United States: cans phospholipomannan, a new member of the
Advances in Sepsis Research 529

fungal mannose inositol phosphoceramide family. 35. Paz S, Sun Q, Nakhaei P, et al. Induction of IRF-3
J Biol Chem 2002;277:37260–71. and IRF-7 phosphorylation following activation of
19. Azzam KM, Fessler MB. Crosstalk between reverse the RIG-I pathway. Cell Mol Biol 2006;52:17–28.
cholesterol transport and innate immunity. Trends 36. Slack JL, Schooley K, Bonnert TP, et al. Identification
Endocrinol Metab 2012;23:169–78. of two major sites in the type I interleukin-1 receptor
20. Hailman E, Albers JJ, Wolfbauer G, et al. Neutraliza- cytoplasmic region responsible for coupling to pro-
tion and transfer of lipopolysaccharide by phospho- inflammatory signaling pathways. J Biol Chem
lipid transfer protein. J Biol Chem 1996;271:12172–8. 2000;275:4670–8.
21. Gautier T, Klein A, Deckert V, et al. Effect of plasma 37. Kumar H, Kawai T, Akira S. Pathogen recognition by
phospholipid transfer protein deficiency on lethal the innate immune system. Int Rev Immunol 2011;
endotoxemia in mice. J Biol Chem 2008;283: 30:16–34.
18702–10. 38. Goddard CM, Poon BY, Klut ME, et al. Leukocyte
22. Vreugdenhil AC, Rousseau CH, Hartung T, et al. activation does not mediate myocardial leukocyte
Lipopolysaccharide (LPS)-binding protein mediates retention during endotoxemia in rabbits. Am J Phys-
LPS detoxification by chylomicrons. J Immunol iol 1998;275:H1548–57.
2003;170:1399–405. 39. Herbertson MJ, Werner HA, Goddard CM, et al. Anti-
23. Berbee JF, Havekes LM, Rensen PC. Apolipopro- tumor necrosis factor-alpha prevents decreased
teins modulate the inflammatory response to lipo- ventricular contractility in endotoxemic pigs. Am J
polysaccharide. J Endotoxin Res 2005;11:97–103. Respir Crit Care Med 1995;152:480–8.
24. Fleischer JG, Rossignol D, Francis GA, et al. Deac- 40. Herbertson MJ, Werner HA, Studer W, et al.
tivation of the lipopolysaccharide antagonist eritoran Decreased left ventricular contractility during
(E5564) by high-density lipoprotein-associated apo- porcine endotoxemia is not prevented by ibuprofen.
lipoproteins. Innate Immun 2012;18:171–8. Crit Care Med 1996;24:815–9.
25. Harris HW, Grunfeld C, Feingold KR, et al. Chylomi- 41. Herbertson MJ, Werner HA, Walley KR. Nitric oxide
crons alter the fate of endotoxin, decreasing tumor synthase inhibition partially prevents decreased LV
necrosis factor release and preventing death. contractility during endotoxemia. Am J Physiol
J Clin Invest 1993;91:1028–34. 1996;270:H1979–84.
26. Lanza-Jacoby S, Miller S, Jacob S, et al. Hyperlipo- 42. McDonald TE, Grinman MN, Carthy CM, et al. Endo-
proteinemic low-density lipoprotein receptor- toxin infusion in rats induces apoptotic and survival
deficient mice are more susceptible to sepsis than pathways in hearts. Am J Physiol Heart Circ Physiol
corresponding wild-type mice. J Endotoxin Res 2000;279:H2053–61.
2003;9:341–7. 43. Simms MG, Walley KR. Activated macrophages
27. Walley KR, Thain KR, Russell JA, et al. PCSK9 is a decrease rat cardiac myocyte contractility: impor-
critical regulator of the innate immune response tance of ICAM-1-dependent adhesion. Am J Physiol
and septic shock outcome. Sci Transl Med 2014;6: 1999;277:H253–60.
258ra143. 44. Walley KR, Hebert PC, Wakai Y, et al. Decrease in
28. Akira S, Uematsu S, Takeuchi O. Pathogen recogni- left ventricular contractility after tumor necrosis
tion and innate immunity. Cell 2006;124:783–801. factor-alpha infusion in dogs. J Appl Physiol (1985)
29. Akira S, Takeda K, Kaisho T. Toll-like receptors: crit- 1994;76:1060–7.
ical proteins linking innate and acquired immunity. 45. Davani EY, Dorscheid DR, Lee CH, et al. Novel reg-
Nat Immunol 2001;2:675–80. ulatory mechanism of cardiomyocyte contractility
30. Nakada E, Nakada TA, Walley KR, et al. mRNA in- involving ICAM-1 and the cytoskeleton. Am J Physiol
duces RANTES production in trophoblast cells via Heart Circ Physiol 2004;287:H1013–22.
TLR3 only when delivered intracellularly using lipid 46. Boyd JH, Mathur S, Wang Y, et al. Toll-like re-
membrane encapsulation. Placenta 2011;32:500–5. ceptor stimulation in cardiomyoctes decreases
31. Nakada E, Walley KR, Nakada T, et al. Toll-like contractility and initiates an NF-kappaB dependent
receptor-3 stimulation upregulates sFLT-1 produc- inflammatory response. Cardiovasc Res 2006;72:
tion by trophoblast cells. Placenta 2009;30:774–9. 384–93.
32. Petrilli V, Dostert C, Muruve DA, et al. The inflamma- 47. Boyd JH, Chau EH, Tokunanga C, et al. Fibrinogen
some: a danger sensing complex triggering innate decreases cardiomyocyte contractility through an
immunity. Curr Opin Immunol 2007;19:615–22. ICAM-1-dependent mechanism. Crit Care 2008;
33. Proell M, Riedl SJ, Fritz JH, et al. The nod-like recep- 12:R2.
tor (NLR) family: a tale of similarities and differences. 48. De Rossi M, Bernasconi P, Baggi F, et al. Cytokines
PLoS one 2008;3:e2119. and chemokines are both expressed by human
34. Hornung V, Ellegast J, Kim S, et al. 5’-Triphosphate myoblasts: possible relevance for the immune path-
RNA is the ligand for RIG-I. Science 2006;314: ogenesis of muscle inflammation. Int Immunol 2000;
994–7. 12:1329–35.
530 Bentzer et al

49. Finkel MS, Oddis CV, Jacob TD, et al. Negative 63. Nygren A, Redfors B, Thoren A, et al. Norepineph-
inotropic effects of cytokines on the heart mediated rine causes a pressure-dependent plasma volume
by nitric oxide. Science 1992;257:387–9. decrease in clinical vasodilatory shock. Acta anaes-
50. Hattori Y, Kasai K. Induction of mRNAs for ICAM-1, thesiologica Scand 2010;54:814–20.
VCAM-1, and ELAM-1 in cultured rat cardiac myo- 64. Minnear FL, Barrie PS, Malik AB. Effects of epineph-
cytes and myocardium in vivo. Biochem Mol Biol rine and norepinephrine infusion on lung fluid bal-
Int 1997;41:979–86. ance in sheep. J Appl Physiol Respir Environ
51. Niessen HW, Krijnen PA, Visser CA, et al. Intercel- Exerc Physiol 1981;50:1353–7.
lular adhesion molecule-1 in the heart. Ann N Y 65. Maybauer MO, Maybauer DM, Enkhbaatar P, et al.
Acad Sci 2002;973:573–85. The selective vasopressin type 1a receptor agonist
52. Raeburn CD, Dinarello CA, Zimmerman MA, et al. selepressin (FE 202158) blocks vascular leak in ovine
Neutralization of IL-18 attenuates lipopolysaccharide- severe sepsis*. Crit Care Med 2014;42:e525–33.
induced myocardial dysfunction. Am J Physiol Heart
66. Rehberg S, Ertmer C, Vincent JL, et al. Role of selec-
Circ Physiol 2002;283:H650–7.
tive V1a receptor agonism in ovine septic shock. Crit
53. Raeburn CD, Calkins CM, Zimmerman MA, et al.
Care Med 2011;39:119–25.
ICAM-1 and VCAM-1 mediate endotoxemic myocar-
dial dysfunction independent of neutrophil accumu- 67. Adamson RH, Clark JF, Radeva M, et al. Albumin
lation. Am J Physiol Regul Integr Comp Physiol modulates S1P delivery from red blood cells in
2002;283:R477–86. perfused microvessels: mechanism of the protein ef-
54. Boyd JH, Kan B, Roberts H, et al. S100A8 and fect. Am J Physiol Heart Circ Physiol 2014;306:
S100A9 mediate endotoxin-induced cardiomyocyte H1011–7.
dysfunction via the receptor for advanced glycation 68. Christoffersen C, Obinata H, Kumaraswamy SB, et al.
end products. Circ Res 2008;102:1239–46. Endothelium-protective sphingosine-1-phosphate
55. Boyd JH, Forbes J, Nakada TA, et al. Fluid resusci- provided by HDL-associated apolipoprotein M.
tation in septic shock: a positive fluid balance and Proc Natl Acad Sci U S A 2011;108:9613–8.
elevated central venous pressure are associated 69. Feistritzer C, Riewald M. Endothelial barrier protec-
with increased mortality. Crit Care Med 2011;39: tion by activated protein C through PAR1-
259–65. dependent sphingosine 1-phosphate receptor-1
56. National Heart, Lung, and Blood Institute Acute Res- crossactivation. Blood 2005;105:3178–84.
piratory Distress Syndrome (ARDS) Clinical Trials 70. Lundblad C, Axelberg H, Grande PO. Treatment
Network, Wiedemann HP, Wheeler AP, et al. Com- with the sphingosine-1-phosphate analogue FTY
parison of two fluid-management strategies in acute 720 reduces loss of plasma volume during experi-
lung injury. N Engl J Med 2006;354:2564–75. mental sepsis in the rat. Acta anaesthesiologica
57. London NR, Zhu W, Bozza FA, et al. Targeting Scand 2013;57:713–8.
Robo4-dependent slit signaling to survive the cyto- 71. Peng X, Hassoun PM, Sammani S, et al. Protective
kine storm in sepsis and influenza. Sci Transl Med effects of sphingosine 1-phosphate in murine
2010;2:23ra19. endotoxin-induced inflammatory lung injury. Am J
58. Opal SM, van der Poll T. Endothelial barrier dysfunc- Respir Crit Care Med 2004;169:1245–51.
tion in septic shock. J Intern Med 2015;277:277–93.
72. Wang L, Sammani S, Moreno-Vinasco L, et al.
59. Schmidt C, Hocherl K, Kurt B, et al. Role of nuclear
FTY720 (s)-phosphonate preserves sphingosine 1-
factor-kappaB-dependent induction of cytokines in
phosphate receptor 1 expression and exhibits supe-
the regulation of vasopressin V1A-receptors during
rior barrier protection to FTY720 in acute lung injury.
cecal ligation and puncture-induced circulatory fail-
Crit Care Med 2014;42:e189–99.
ure. Crit Care Med 2008;36:2363–72.
60. Favory R, Poissy J, Alves I, et al. Activated protein C 73. Wang Z, Sims CR, Patil NK, et al. Pharmacologic tar-
improves macrovascular and microvascular reac- geting of sphingosine-1-phosphate receptor 1 im-
tivity in human severe sepsis and septic shock. proves the renal microcirculation during sepsis in
Shock 2013;40:512–8. the mouse. J Pharmacol Exp Ther 2015;352:61–6.
61. Dellinger RP, Levy MM, Rhodes A, et al. Surviving 74. Kerschen EJ, Fernandez JA, Cooley BC, et al. Endo-
sepsis campaign: international guidelines for man- toxemia and sepsis mortality reduction by non-
agement of severe sepsis and septic shock. Critical anticoagulant activated protein C. J Exp Med
Care Medicine 2012;41(2):580–637. 2007;204:2439–48.
62. Dubniks M, Persson J, Grande PO. Effect of blood 75. Kumaraswamy SB, Linder A, Akesson P, et al.
pressure on plasma volume loss in the rat under Decreased plasma concentrations of apolipoprotein
increased permeability. Intensive Care Med 2007; M in sepsis and systemic inflammatory response
33:2192–8. syndromes. Crit Care 2012;16:R60.

You might also like