Endocrine Markers of Severity and Prognosis in Critical Illness

Transcription

Endocrine Markers of Severity and Prognosis in Critical Illness
Crit Care Clin 22 (2006) 161 – 179
Endocrine Markers of Severity and Prognosis in
Critical Illness
Eric S. Nylén, MDa,b,T, Nitin Seam, MDc,
Rahul Khosla, MDa,b
a
b
Veteran Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
George Washington University School of Medicine, 2150 Pennsylvania Avenue, NW,
Washington, DC, 20037, USA
c
National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, USA
The mortality of critically ill patients in the ICU remains unacceptably high
despite advancements in supportive care over the last few decades. However, a
recently developed endocrine-based intervention, intensive insulin therapy to
control stress hyperglycemia, has successfully reduced mortality in the critically
ill and has restored hope that outcomes can be substantially improved. Another
crucial challenge, in which endocrine marker strategy may have an important
impact, is the identification of high-risk patients. In this regard, the list of potential biomarkers is burgeoning as is the expanding concept of endocrine constituents. In this review, an attempt has been made to cast a wide net for potential
endocrine markers that confer useful information regarding severity and outcome
in critical illness.
Hormonal markers
Cortisol
Activation of the hypothalamic-pituitary-adrenal (HPA) axis with enhanced
cortisol concentrations is perhaps the best documented endocrine response to
severe systemic stress. In critical illness, cortisol exerts a regulatory role which is
T Corresponding author. Veteran Affairs Medical Center, 50 Irving Street, NW, Washington,
DC, 20422.
E-mail address: [email protected] (E.S. Nylén).
0749-0704/06/$ – see front matter D 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.ccc.2005.08.002
criticalcare.theclinics.com
162
nylÉn et al
essential to prevent immunotoxicity, and to provide metabolic substrate, promote
catecholaminergic cardiovascular action, optimize vascular tone and integrity,
and ultimately enhance survival [1,2]. Typically, serum cortisol levels can increase 5 to 10 fold in close proximity to the insult, but assessment based on single
cortisol values or following exogenous cortisol stimulation can be misleading,
especially because the optimal range of cortisol response is still a matter of
debate. Many patients with frequent hypoproteinemia have demonstrated a robust
cortisol response when assessed by serum-free cortisol assays, which more accurately reflect bioactive cortisol [3].
In general, there is a graded cortisol response to the degree of stress, such as
the type of surgery. Cortisol levels also correlate with the severity of injury [4], the
Glasgow Coma Scale [5], and the acute physiology and chronic health evaluation
(APACHE) scoring system [6]. There is often a wide, bimodal spectrum of
cortisol concentrations in the ICU and the patients at either extreme fare the
worst. An inverse correlation of cortisol response to the outcome of sepsis has
been frequently established [6–8] (Fig. 1). In a mixed ICU population, admission
cortisol varied from a mean of 27 mg/dL in survivors, to a mean of 47 mg/dL in
non-survivors, differences that predicted outcome [9]. Serial cortisol measurements with increasing values also signify worse survival [4]. Discriminatory
prognostic values incorporating cortsiol response to corticotrophin (ACTH) stimulation in septic shock has been reported. This approach, however, relies on ad100
Cortisol group 1
Cortisol group 2
Cortisol group 3
Cortisol group 4
Survival (%)
80
60
40
20
0
0
20
40
60
80
100
Days after admission
Fig. 1. Ninety-day survival (Kaplan-Meier curve) in 100 medical ICU patients who have severe sepsisseptic shock stratified according to the total serum cortisol concentration (within 48 hours of
admission). Group 1, cortisol 345 nmol/L (12.3 mg/dL); group 2, cortisol 345–552 nmol/L (12.3–
19.7 mg/dL); group 3, cortisol 552–1242 nmol/L (19.7–44.4 mg/dL); group 4, cortisol 1242 nmol/L
(44.4 mg/dL). Group 4 versus groups 1–3 P b 0.02 (Mantel-Cox log rank test). (From Sam S,
Corbridge TC, Mokhlesi B, et al. Cortisol levels and mortality in severe sepsis. Clin Endocrinol (Oxf)
2004;60:29–35; with permission.)
endocrine markers
163
renal synthetic reserves rather than on the overall integrated adrenal axis, which
includes extra-adrenal cortisol stimulation, and the response to ACTH can be low
in otherwise healthy subjects [2,10].
Dehydroepiandosterone sulfate
The adrenocortical sex steroid hormone dihydroepiandosterone (DHEA) sulfate was reported to correlate inversely with the APACHE score and to be particularly low in septic shock and non-survivors compared with multiple trauma
and control subjects [11]. Interestingly, the levels of the principal adrenal androgen, DHEA, may actually increase in sepsis [12].
Thyroid hormones
Serum thyroid hormone levels undergo predictable changes in systemic nonthyroidal illness (NTI). The initial change in the hypothalamic-pituitary-thyroid
(HPT) axis during a mild illness is a decrease in triiodothyronine (T3) production
caused by inhibition of the conversion of thyroxine (T4) to T3, with a reciprocal
increase in reverse T3 (rT3), the so-called low T3 syndrome. This decrease can
occur very rapidly [13] and both the decrease in serum T3 and rise in rT3 have
been reported to correlate with severity of illness [14,15]. With advanced severity
of illness, T4 also decreases; low T4 syndrome is often seen in adult and pediatric
subjects with chronic or severe illnesses [16]. Decreased T4 and T3 concentrations correlate with increased mortality: a total T4 level b 3 mg/dL was associated with an 84% mortality and a T4 level N 5 mg/dL had 15% mortality [17]. In
critical illness, the concentration of serum thyroid-stimulating hormone (TSH) is
often low–normal. A prospectively obtained admission TSH was lower in nonsurvivors than survivors [9]. Additional insights into the HPT axis relationship to
outcome was reported in a cohort of 451 critically ill patients who stayed in an
ICU for more than 5 days, randomized to conventional versus intensive insulin
management [18]. In addition to confirming the prognostic role of T3 and T4,
both elevated rT3 and the decreased T3 /rT3 ratio were prognostic for nonsurvivors on the day of admission. These parameters showed increased
significance by the last day (predictive area under curve receiver operating characteristics [AUROC] value 0.76 and 0.84, respectively). Interestingly, TSH
showed a significant divergence by day 5: non-survivors remained low whereas
survivors showed a more than 2-fold increase, preceding the rise in T3 and T4,
which suggests that serial monitoring may have a role. Although TSH is often
low–normal in NTI, there is a blunted response to thyrotropin releasing hormone
(TRH), which portends a worse outcome, as does low TSH levels [19,20].
Natriuretic peptides
Atrial natriuretic peptide (ANP) is synthesized in atrial myocyctes and is
released in response to atrial distension causing vasodilatory and natriuretic
164
nylÉn et al
bioactions [21]. An ANP receptor-mediated modulation of macrophage function
[22] and priming of neutrophils has been observed [23], which suggests a link to
the inflammation. ANP was increased approximately fivefold in a small study of
septic shock patients, which correlated with the peak of interleukin-6 (IL-6)[24].
N-terminal proANP levels correlated with severity of sepsis and cardiac depression [25] and death (AUROC of 0.88), were significantly greater than levels
for procalcitonin (proCT) and C-reactive protein (CRP), but were similar to
APACHE scores [26].
Brain natriuretic peptide (BNP) is released primarily from ventricular cardiomyocytes in response to myocardial tension and stretch, and has overlapping
bioactions with ANP [21]. In a prospective observational study of severe sepsis
and septic shock, BNP correlated with evidence of reduced myocardial systolic
performance and elevated BNP at days 2 and 3 of the ICU stay, and was associated with an increased mortality [27]. N-terminal proBNP performed in a similar
manner and was prognostic within 1–2 days of shock; a level of N13,600 pg/mL
predicted mortality (AUROC 0.8) [28]. Despite BNP being elevated with renal
dysfunction, cardiac indices have been shown to be independent determinants
of plasma BNP levels in patients who have chronic renal failure. Although not
all studies confirm their prognostic properties [29], ongoing work suggests a role
of these peptides in various aspects of the immune system activity relevant to
critical illness.
Metabolic markers
Glucose
Stress hyperglycemia (SH), hyperglycemia in previously euglycemic patients
that corrects once the acute process resolves, is a well-recognized process in
critically ill patients. SH is the result of a complex interaction between stress
metabolism, cytokine influences, oxidative stress, and stress signaling pathways.
The net result is defective insulin signaling, insulin resistance, enhanced glycogenolysis, increased hepatic gluconeogenesis, and decreased use of glucose [30].
The influence of diabetes (DM) as an independent risk factor for coronary
artery disease (CAD) has been known for some time, but the correlation between
hyperglycemia and outcomes in patients with myocardial infarction (MI) has
only recently been recognized. In the Diabetes Mellitus Insulin-Glucose Infusion in Acute Myocardial Infarction (DIGAMI) trial, maintaining blood glucose
b 215 mg/dL in post-MI DM was associated with 25% decreased mortality at
3.4 years follow-up in the insulin-treated group [31]. The same study then
retrospectively examined 197 post-MI non-DM patients and found admission
hyperglycemia as an independent predictor of nonfatal reinfarction, hospitalization for congestive heart failure, and major cardiovascular events in the 1.5–
2.5 years of follow-up [32]. Another study of 336 patients post acute MI, found
that admission hyperglycemia independently predicted increased 1-year mortality
endocrine markers
165
in both DM and non-DM [33]. The detrimental in-hospital prognosis for both
DM and non-DM with MI, and of stress hyperglycemia, was further substantiated in a meta-analysis study [34].
Several retrospective studies of acute stroke found an association between
admission hyperglycemia and worse clinical outcome [35,36]. Admission hyperglycemia may also predispose to hemorrhagic conversion of an acute cerebral
infarct [37]. Persistent post-stroke hyperglycemia (mean glucose level N7 mmol/L;
N126 mg/dL) was also associated with worse outcome, increased final infarct
volume change, a worse NIH stroke scale, and modified Rankin Scale scores at
72 hours [38]. The increased mortality or functional recovery following ischemic
stroke in non-DM, in association with stress hyperglycemia, was substantiated
in a large review of the literature [39].
Several studies have shown improved outcomes in critically ill (mostly) surgical patients with control of blood glucose. Most notably, a prospective randomized trial of 1548 adults (13% of patients had history of DM) who received
mechanical ventilation in a surgical ICU, studied intensive insulin therapy
(achieved mean glucose was 100 mg/dL in the intensively treated group versus
158 mg/dL in the control group) versus conventional therapy and found a significant decrease in ICU mortality (the primary endpoint) as well as in-hospital
mortality with intensive insulin therapy [40]. Control of blood glucose, not insulin administration itself, was the cause of improved outcome [41]. This conclusion was supported by a prospective observational trial of 531 patients
admitted to a single ICU (88% of the patients had undergone surgery) in which
increased insulin administration was positively associated with death, regardless
of glucose level [42]. In another study, 800 consecutive patients were admitted to
a single medical-surgical ICU before the use of the insulin protocol and were
compared with 800 consecutive patients after the protocol was put in place. There
was a 29.3% decrease in hospital mortality and 10.8% decrease in length of
ICU stay after the protocol was started [43]. Moreover, in a recent prospective
study of 942 consecutive trauma patients admitted to a single SICU over 2 years,
patients with persistent hyperglycemia had increased mortality, time on the ventilator, and length of hospital stay (by univariate analysis) [44].
In the medical ICU the APACHE II can correlate with admission glucose
(Fig. 2); mortality was significantly lower in patients with a mean glucose of
4–6 mmol/L (72–108 mg/dL) compared with the group with a mean glucose level
of 6–10 mmol/L (108–180 mg/dL) [45]. However, in a prospective study of
135 consecutive non-DM patients who were admitted to a non-cardiac multidisciplinary ICU over 6 months, maximum blood glucose levels while in the
ICU correlated with ICU mortality only in the surgical, and not medical, patients [46].
Lipids
Hypocholesterolemia ensues early in the course of a variety of critical illnesses, and correlates with severity, nosocomial infections, and mortality. A
166
nylÉn et al
Fig. 2. Regression analysis of the APACHE II scores and admission serum glucose in 73 consecutive medical ICU patients who have a variety of critical illnesses (diabetics excluded) (R = 0.39;
r 2 = 0.15; Pb0.001).
U-type of association has been described between mortality and cholesterol in
large groups of hospitalized patients [47]. Admission cholesterol and lipoproteins
are typically decreased (eg, a cut- off of median total cholesterol of b120 mg/dL
predicted clinical outcome) and correlate inversely with cytokines such as IL-6
and interleukin-10 (IL-10) [48]. In another surgical ICU study of patients with
systemic inflammatory response syndrome (SIRS), cholesterol levels were low
on day 2, which persisted and were independently associated with mortality along
with the APACHE III score [49]. Lipolysis, based on plasma-free fatty acid and
glycerol concentrations, is markedly elevated in septic patients and these parameters, along with low triglycerides, correlated with clinical severity; high-density
lipoprotein (HDL) shows an inverse correlation [50]. The free glycerol concentration was additive to the APACHE score in predicting mortality. The role
of HDL is of particular interest because of its ability to bind and inactivate
lipopolysaccharide (LPS) as well as other anti-inflammatory properties. The
prognostic significance of serum lipids such as HDL may be significant because
of the salutary contribution of the lipid response (ie, increased low-density lipoprotein and HDL) to intensive insulin management and enhanced outcome [51].
In obesity, there is adipose infiltration of active macrophages and upregulation of a host of inflammatory genes, which contribute to a state of chronic
heightened inflammation and down-regulated bioenergetics. The few studies that
evaluate the impact of obesity on the severity or prognosis of critical illness are
contradictory. However, one prospective study suggested that that a body mass
index N27 kg/m2 correlated with severity, ICU mortality, and length of ICU
stay [52]. Another pediatric study noted increased weight with mortality from
sepsis [53].
endocrine markers
167
Immune markers
Cytokines
The inflammatory response syndrome is to a large extent initiated by transient
expression of cytokines such as tumor necrosis factor a (TNF-a) and IL-1ba.
These proinflammatory cytokines trigger IL-6 production furthering the inflammatory activation process, which includes the acute phase response. IL-6 concentrations are rather stable and easy to detect, and show reasonable correlation—
in contrast to TNF-a and IL-1b—to the severity of illness [54,55], the degree
of overall cytokinemia [56], stress hyperglycemia [57], the presence of adult
respiratory distress syndrome (ARDS) [58], and outcome [59–61]. IL-6 has also
served as a prototype stratification marker [62–64]. Thus, IL-6 may enhance
prognostication compared with current clinical criteria. In predicting hospital
mortality from sepsis, day 2 IL-6 (and ProCT) reached N 0.75 AUROC [65].
Interleukin-8 is a proinflammatory chemokine that stimulates migration of
neutrophils and macrophages to inflammatory sites. IL-8 increases in the early
stages of sepsis and has been correlated to IL-6 levels [66]. IL-8 is also associated with viral conditions such as dengue fever and its outcome [67]. IL-8
has been reported to be significant in sepsis and multiple organ failure (MOF)
[68] and is elevated in patients that die from lung injury [69].
Interleukin-10 is a potent anti-inflammatory cytokine and may be a useful
prognostic marker for organ failure in pancreatitis [70], sepsis mortality [71],
and myocarditis mortality [72,73].
Interleukin-18, previously known as the inducer of interferon-gamma, has
potent immunomodulatory actions that involve T cells. IL-18 levels increase in
those who develop SIRS, in subjects with peritonitis, and organ failure patients
[74], and can be predictive of outcome [75]. In sepsis, IL-18 correlates with
severity and, together with IL-6, is considerably higher in patients with grampositive compared with gram-negative infections [76].
C-reactive protein
C-reactive protein (CRP) is an acute phase protein that is predominantly produced and secreted by hepatocytes in response to cytokines such as IL-6. Plasma
levels of CRP are below 10 mg/L in 99% of healthy individuals and usually rise
4 to 6 hours after an infectious stimulus to reach a peak after 36 to 50 hours.
A CRP level N10 mg/dL on ICU admission was associated with high mortality [77]. In children with hematopoietic stem cell transplants, a CRP level
N10 mg/dL was predictive of poor outcome [78]. Patients who had acute myocardial infarction and CRP levels N2.23 mg/dL on admission, had a significantly higher mortality and heart failure rate [79]. A strong correlation between
the CRP to pre-albumin ratio and severity of organ dysfunction at 48 hours
and 120 hours has been noted in critically ill patients [80]. In patients who have
chronic respiratory failure, elevated CRP levels are strongly predictive of rate of
168
nylÉn et al
hospitalizations and survival [81]. Moreover, persistently high CRP concentrations in the ICU are associated with a poor outcome [82]. However, although
CRP is a very sensitive marker and is elevated in both inflammation and infection, it is non-specific, and not able to differentiate from underlying inflammatory
diseases such as auto-immune diseases, trauma, major surgeries, burns, and malignant diseases.
Calcitonin precursors and procalcitonin
High molecular fractions cross-reacting with polyclonal calcitonin antisera
was first recognized to be associated with infections in the staphylococcal toxic
shock syndrome [83]. Subsequent studies have revealed that a variety of critical
illnesses can increase the circulating concentration of several precursor peptides
to calcitonin (ie, calcitonin precursors; CTpr) including procalcitonin (ProCT)
[84]. In critical illness, the source of CTpr appears to be a diversity of nonthyroidal cells without the ability to complete cellular hormonal packaging. Although CTpr levels can be detected in normal healthy sera [85], it is markedly
(and persistently) increased by 24–36 hours following exposure to bacteria, LPS,
or proinflammatory cytokines. Similar responses also occur in severe systemic
conditions such as burns and heat stroke, and a more muted response has been
noted to viral infections, whereas the response to fungi is variable [86]. ProCT
release appears to be unimpeded and remains useful in neutropenic states [87].
Several studies show a strong correlation of CTpr with severity scores such as
APACHE [88], SOFA (sequential organ failure assessment) [89], SAPS (Simplified Acute Physiology Score) [90], and PRISM (Pediatric Risk of Mortality)
[91]. Across the spectrum of severity, from SIRS to septic shock, CTpr levels
are much less variable than IL-6 or CRP [92]. CTpr also correlates with mortality;
a cutoff value of 6 ng/mL on day 1 separated patients who died from those
who survived with 87.5% sensitivity and 45% specificity [93]; a day 2 ProCT
reached a predictive value of hospital mortality (AUROC N 0.75) [65]; a maximal
ProCT value b 2.1 ng/mL was seen in 100% of survivors [88]. Sequencial
decrease in ProCT can also suggest improved survival [94]. Peak PCT (and the
albumin level) outperformed the Euroscore (European System for Cardiac Operative Risk Evaluation) for predicting post-operative mortality following bypass
surgery [95].
Monocyte migration inhibitory factor
Monocyte migration inhibitory factor (MIF) is a pleiotropic mediator with
hormonal, cytokine, and enzymatic activities implicated in the regulation of immune and inflammatory responses. In addition to monocytes and macrophages,
MIF is produced in a variety of cells including a variety of endocrine cells.
Glucocorticoids induce the secretion of MIF and MIF, in turn, can antagonize
glucocorticoid release. It has also been shown that treatment of septic patients
with glucocorticoids can result in decreased MIF release [96]. The proinflamma-
endocrine markers
169
tory mediator role of MIF in sepsis has been established in experimental studies
[97]. In human studies, the MIF concentration is elevated in sepsis, and is
highest in those with septic shock [98]. In another study, sepsis patients had
significantly higher MIF levels compared with trauma or control patients on the
day of admission; levels which were persistently elevated [99]. MIF levels
were also significantly higher in non-survivors compared with survivors of
septic shock. Similarly, MIF was higher in subjects who had ARDS compared
with non-ARDS subjects and predicted mortality outcome better than ARDS. In
a comparative study of 25 patients with sepsis, both MIF and IL-6 were significantly higher in patients who died. In this study, an MIF level of N1100 pg/mL
had a sensitivity of 100% and specificity of 64% for a fatal outcome [100]. Higher
circulating MIF levels 6 hours post-cardiopulmonary bypass surgery were associated with worse outcome [101].
High mobility group-1
High mobility group-1 (HMG-1) is a highly conserved DNA-binding protein
with proinflammatory cytokine-like properties, which elicits a sepsis-like syndrome when administered [102]. Moreover, antibodies to HMG-1 confer delayed
protection against experimental sepsis [103]. HMG-1 is considered a ‘‘late’’
mediator and is released 8–32 hours following endotoxin exposure. Moreover,
in human sepsis, HMG-1 remained elevated, whereas cytokines such as IL-6,
IL-8, and IL-10, had returned to near normal values [104]. However, although
IL-6, IL-8, and IL-10 correlated with the SOFA score, there was no such correlation with the levels of HMG-1.
Platelet activating factor and platelet activating factor-acetyhydrolase
Platelet activating factor (PAF) and other cell membrane-derived phospholipids have been reported to be proinflammatory mediators and are elevated in
sepsis and involved in organ failure [105,106]. In hypoxic-ischemic encephalopathy, PAF levels were twice that of healthy controls and correlated with the
clinical severity [107]. PAF binding to platelets correlated to the extent, infarct
volume, and neurological impairment in stroke victims [108].
PAF is inactivated by PAF–acetylhydrolase (PAF-AH). In acute MI, trauma,
and sepsis, the PAF-AH is decreased and related to organ failure [106,109].
Soluble triggering receptor expressed on myeloid cells-1
The triggering receptor expressed on myeloid cells (TREM-1) is a member of
the immunoglobulin super family, the expression of which is up-regulated on
phagocytic cells in the presence of bacteria or fungi [110]. In noninfectious
inflammatory disorders TREM-1 is weakly expressed [111]. A soluble form of
TREM-1 (sTREM-1) is released from activated phagocytes and determination of
the level of s-TREM-1 in biological fluids as plasma [112] and bronchoalveolar
170
nylÉn et al
lavage [113] has been shown to be helpful to identify an infectious process.
Moreover, it had a favorable prognostic performance relative to ProCT and CRP
[112]. Decreasing level of sTREM-1 over time is associated with a favorable
clinical outcome [114].
Endohelial markers
Nitric oxide and asymmetric dimethylarginine
Nitric oxide (NO) is a free radical formed by a group of NO synthases, which
rapidly convert l-arginine into NO and l-citrullin. Being in a gaseous state, NO
is often indirectly measured by metabolites such as nitrite and nitrate. As a potent
vasodilator, NO is implicated in the vasodilatory state of septicemia, the hypotension of which may cause organ failure. It also decreases endothelin-1 levels, a
potent vasoconstrictor. Actual NO production appears to be increased in sepsis
[115] and indirect NO measures suggest that it is elevated in SIRS, burns, sepsis,
and heatstroke. In heatstroke, excessive NO production is proportional to the
severity of illness [116]. The serum NO level has been used as a predictor of
postoperative morbidity [117].
Asymmetric dimethylarginine (ADMA), an L-arginine analog and an endogenous inhibitor of NO synthase, with its own vasoconstrictive properties, has been
shown to be associated with organ failure such as hepatic failure and also being a
strong and independent predictor of mortality [118]. In another study,
the mortality correlation to ADMA was confirmed. In addition, ADMA correlated to APACHE and several other important illness parameters [119].
Endothelin-1
This 21 amino acid peptide is a potent vasoconstrictor which stimulates the
production of endothelial NO. The serum level of endothelin is elevated in a
variety of states, especially in sepsis and septic shock, and correlates with severity and mortality [120,121]. For example, endothelin-1 levels were highest in
culture-positive neonatal sepsis, and correlated with SNAP (severity instrument
Score for Neonatal Acute Physiology) [122].
Complement markers
The complement cascade is a part of the innate immunity and its activation
has been documented in sepsis, trauma, burns, and cardiopulmonary bypass
[123]. In trauma, the complement 3a (C3a/C3) levels increase with illness severity and the level is significantly higher in non-survivors than survivors [124].
In sepsis, complement is activated by bacterial cell components as evidenced
by the increase in plasma concentration of C3a and C5a upon injection of LPS
[125]. The role of complement in combating septicemia is paradoxical, because
lysis or opsonization of invading microorganism is an important part of host
defense, but excessive complement activation can lead to harmful tissue destruction caused by severe inflammation. Selective inhibition of specific pathways
endocrine markers
171
may be more advantageous. Inhibition of C3a receptor has increased survival
during sepsis [126]. Anti-C5a antibodies alleviate symptoms of ARDS in septic
primates [127]. Cardiopulmonary bypass induces a systemic inflammatory response that contributes to clinical morbidity: two complement inhibitors, TP10
(a C3/C5 convertase inhibitor) and pexelizumab, both anti-C5 monoclonal antibodies, are in clinical trials. TP10 has been shown to limit tissue injury and
improve myocardial dysfunction during and following experimental cardiopulmonary bypass [128]. Pexelizumab was shown to reduce post cardiopulmonary
bypass graft incidence of non-Q wave myocardial infarction and mortality [129].
Coagulation markers
The activation of the coagulation cascade has been shown to be associated
with the development of MOF and poor prognosis in septic patients [130–132].
Protein C (PC) is a key molecule in the coagulation cascade and reduced
concentration is observed in septic patients and is associated with poor outcomes
[133,134]. PC is activated by binding to the endothelial PC receptor and the
thrombumodulin-thrombin complex. The activated protein C has antithrombotic,
profibrinolytic, antiapoptotic, and anti-inflammatory properties [135]. Low levels
of PC is associated with worse outcome in sepsis [136,137], and plasma levels
are also reduced in patients with acute lung injury and ARDS [138].
D-dimer (DD) formation results from the destruction of cross-linked fibrin and
is a measure of clot formation and lysis. Elevated levels of DD are associated
with increased mortality, severity of sepsis, ARDS, and MOF [139,140]. Higher
levels of DD in patients with community acquired pneumonia were shown to be
associated with APACHE scores and higher in-hospital mortality [141]. DD
levels N2000 ng/mL in patients who have a pneumonia severity index of category IV, correlated with increased risk of in-hospital death.
Coagulopathic dynamics continues to be a very important area of exploration,
many markers of which augment early prognostication [142].
Summary
The progression and outcome of critical illness is a highly complex and
heterogeneous confluence of variably definable factors, which seem to defy simplification into a few circulating endocrine markers. Attributes that would make
the current plethora of markers clinically useful include: (1) an early and timely
phase of detectability, (2) a strong discriminatory assay signal associated with the
illness, (3) a reciprocal change in the concentration with the recovery and successful treatment of the illness, (4) a performance that is synergistic with other
clinical severity and outcome instruments, and (5) a definable performance in a
variety of critical illnesses. Few of the above reviewed substances (or those
illustrated in Table 1) clearly meet all or most of these criteria. However, as
insights into the pathophysiology of many of these conditions is advanced, there
172
nylÉn et al
Table 1
Examples of additional endocrine markers of severity and outcome in critical illness
Marker
Action
ICU correlations
Reference
Soluble adhesion molecules
Adrenomedullin
sFas
Glycosides (ouabain)
sHSP70
IGFBP-1
Isoprostane
Leptin
Microalbuminuria
Thrombopoietin
Troponin
vW
Leukocyte attachment
Vasodilatory
Apoptosis signal
Inhibits Na/K ATPase
Molecular chaperones
Regulate free IGFs
Oxidative stress marker
Immunomodulator
Capillary leak
Platelet growth factor
Contractile-regulating proteins
Platelet adhesion mediator
MOF/mortality
APACHE/MOF
MOF/mortality
APACHE II/cortisol
MOF
APACHE/mortality
MOF/mortality
Mortality
SAPS/APACHE/mortality
Severity of sepsis
Mortality
Mortality
[143,144]
[145]
[146]
[147]
[148]
[149]
[150]
[151]
[152]
[153]
[154]
[155]
Abbreviations: APACHE, acute physiology and chronic health evaluation; IGF, insulin growth factor;
IGFBP-1, insulin growth factor binding protein-1; MOF, muliple organ failure; SAP, simplified acute
physiology score; sHSP70, soluble heat shock protein 70; vW, von Willebrand factor.
is certainly reason to expect that there will be excellent candidate markers with
the requisite prognostic characteristics that will have an important impact on
critical illness outcome.
References
[1] Alarifi A, Van den Berghe G, Snider Jr RH, et al. Endocrine markers and mediators in critical
illness. In: Becker KL, editor. Principles and practice of endocrinology and metabolism.
3rd edition. Philadelphia7 Lippincott, Williams Wilkins; 2001. p. 2077 – 87.
[2] Bornstein SR, Chrousos GP. The hypothalamic-pituitary-adrenal axis in stress and critical
illness. In: Becker KL, editor. Principles and practice of endocrinology and metabolism.
3rd edition. Philadelphia7 Lippincott, Williams Wilkins; 2001. p. 2087 – 94.
[3] Hamrahian AH, Tawakalitu OS, Arafah BM. Measurements of serum free cortisol in critically
ill patients. N Engl J Med 2004;350:1629 – 38.
[4] Span LF, Hermus AR, Bartelink AK, et al. Adrenocortical function: an indicator of severity
of disease and survival in chronic critically ill patients. Intensive Care Med 1992;18:93 – 6.
[5] Woolf PD. Hormonal responses to trauma. Crit Care Med 1992;20:216 – 26.
[6] Sam S, Corbridge TC, Mokhlesi B, et al. Cortisol levels and mortality in severe sepsis. Clin
Endocrinol (Oxf) 2004;60:29 – 35.
[7] Finlay WE, McKee JI. Serum cortisol levels in severely stressed patients. Lancet 1982;1:
1414 – 5.
[8] Marik P, Zaloga G. Adrenal insufficiency in the critically ill: a new look at an old problem.
Chest 2002;122:1784 – 96.
[9] Rothwell PM, Lawler PG. Prediction of outcome in intensive care patients using endocrine
parameters. Crit Care Med 1995;23:78 – 83.
[10] Annanne D, Sebille V, Troche G, et al. A 3-level prognostic classification in septic shock
based on cortisol levels and cortisol response to corticotropin. JAMA 2000;283:1038 – 45.
[11] Beishuizen A, Thijs LG, Vermes I. Decreased levels of dehydroepiandrosterone sulphate in
severe critical illness: a sign of exhausted adrenal reserve? Crit Care 2002;6:434 – 8.
[12] Wiebke A, Hammer F, Sanning P, et al. No evidence for DHEA deficiency in severe sepsis–
a stress paradigm revisited. Abstract P3–430; p. 648.
endocrine markers
173
[13] Friberg L, Werner S, Eggertsen G, Ahnve S. Rapid down-regulation of thyroid hormones in
acute myocardial infarction: is it cardioprotective in patients with angina? Arch Intern Med
2002;162:1388 – 94.
[14] Kaptein EM, Robinson WJ, Grieb DA, et al. Peripheral serum thyroxine, triiodothyronine
and reverse triiodonine kinetics in the low thyroxine state of acute nonthyroidal illnesses.
A noncompartmental analysis. J Clin Invest 1982;69:526 – 35.
[15] Docter R, Krenning EP, de Jong M, et al. The sick euthyroid syndrome: changes in thyroid
hormone serum parameters and hormone metabolism. Clin Endocrinol (Oxf) 1993;39:499 – 518.
[16] Simpson J, Williams FLR, Delahunty C, et al. Serum thyroid hormones in preterm infants and
relationships to indices of severity of intercurrent illness. J Clin Endocrinol Metab 2005;90:
1271 – 9.
[17] Slag MF, Morley JE, Elson MK, et al. Hypothyroxinemia in critically ill patients as a predictor of high mortality. JAMA 1981;245:43 – 5.
[18] Peeters RP, Wouters PJ, von Toor H, et al. Serum rT3 and T3/rT3 are prognostic markers in
critically ill patients and are associated with post-mortem tissue deiodinase activities. J Clin
Endocrinol Metab 2005;90(8):4559 – 65.
[19] Sumita S, Ujike Y, Namiki A, et al. Suppression of the thyrotropin response to thyrotropinreleasing hormone and its association with severity of critical illness. Crit Care Med 1994;22:
1603 – 9.
[20] Kaptein EM, Weiner JM, Robinson WJ, et al. Relationship of altered thyroid hormone indices
to survival in nonthyroidal illnesses. Clin Endocrinol (Oxf) 1982;16:565 – 74.
[21] Witthaut R. Science review: natriuretic peptides in critical illness. Crit Care 2004;8:342 – 9.
[22] Kiemer AK, Hartung T, Vollmar AM. cGMP-mediated inhibition of TNF-alpha by the atrial
natriuretic peptide in murine macrophages. J Immunol 2000;165:175 – 81.
[23] Wiedemann CJ, Niedermuhlbichler M, Braunsteiner H. Priming of polymorphonuclear
neutrophils by atrial natriuretic peptide in vitro. J Clin Invest 1992;89:1580 – 6.
[24] Witthaut R, Busch C, Fraunberger P, et al. Plasma atrial natriuretic peptide and brain natriuretic
peptide are increased in septic shock: impact of interleukin-6 and sepsis-associated left ventricular dysfunction. Intensive Care Med 2003;29:1696 – 702.
[25] Mazul-Sunko B, Zarkovic N, Vrkic N, et al. Pro-atrial natriuretic peptide hormone from right
atria is correlated with cardiac depression in septic patients. J Endocrinol Invest 2001;
24(7):RC22–4.
[26] Morgenthaler NG, Struck J, Christ-Crain M, et al. Pro-atrial natriuretic peptide is a prognostic
marker in sepsis similar to the APACHE II score:an observational study. Crit Care 2005;169:
37 – 45.
[27] Charpentier J, Luyt CE, Fulla Y, et al. Brain natriuretic peptide: a marker of myocardial
dysfunction and prognosis during severe sepsis. Crit Care Med 2004;32:660 – 5.
[28] Roch A, Allardet-Servent J, Michelet P, et al. NH2 terminal pro-brain natriuretic peptide
plasma levels as an early marker of prognosis and cardiac dysfunction in septic shock patients.
Crit Care Med 2005;33:1001 – 7.
[29] Berendes E, Van Aken H, Raufhake C, et al. Differential secretion of atrial and brain natriuretic
peptide in critically ill patients. Anesth Analg 2001;93:676 – 82.
[30] Mizok BA. Alterations in fuel metabolism in critical illness: hyperglycemia. Best Practice &
Res Clin Endocrinol Metab 2001;15:533 – 51.
[31] Malmberg K. Role of insulin-glucose infusion in outcomes after acute myocardial infarction:
the diabetes and insulin-glucose infusion in acute myocardial infarction (DIGAMI) study.
Endocr Pract 2004;10:S13–6.
[32] Norhammar AM, Ryden L, Malmberg K. Admission plasma glucose. Independent risk factor
for long-term prognosis after myocardial infarction even in nondiabetic patients. Diabetes Care
1999;33:1827 – 31.
[33] Bolk J, van der Ploeg T, Cornel JH, et al. Impaired glucose metabolism predicts mortality after
a myocardial infarction. J Cardiol 2001;79:207 – 14.
[34] Capes SE, Hunt D, Malmberg K, et al. Stress hyperglycaemia and increased risk of death after
174
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
[55]
[56]
[57]
[58]
nylÉn et al
myocardial infarction in patients with and without diabetes: a systematic overview. Lancet
2000;355:773 – 8.
Moulin T, Tatu L, Crepin-Leblond T, et al. The Besancon stroke registry: an acute stroke
registry of 2500 consecutive patients. Eur Neurol 1997;38:10 – 20.
Weir CJ, Murray GD, Dyker AG, et al. Is hyperglycemia an independent predictor of poor
outcome after acute stroke? Results of a long-term follow up study. BMJ 1997;314:1303 – 6.
Kent TA, Soukup VM, Fabian RH. Heterogeneity affecting outcome from acute stroke therapy:
making reperfusion worse. Stroke 2001;32:2318 – 27.
Baird TA, Parsons MW, Phanh T, et al. Persistent post-stroke hyperglycemia is independently
associated with infarct expansion and worse clinical outcome. Stroke 2003;34:2208 – 14.
Capes SE, Hunt D, Malmberg K, et al. Stress hyperglycaemia and increased risk of death after
myocardial infarction in patients with and without diabetes: a systematic overview. Lancet
2000;355:773 – 8.
Van den Berghe G, Bouillon R, Lauwers P. Intensive insulin therapy in critically ill patients.
N Engl J Med 2001;345:1359 – 67.
Van den Berghe G, Wouters PJ, Bouillon R, et al. Outcome benefit of intensive insulin therapy
in the critically ill: insulin dose versus glycemic control. Crit Care Med 2003;31:359 – 66.
Finney SJ, Zekveld C, Elia A, et al. Glucose control and mortality in critically ill patients.
JAMA 2003;290:2041 – 7.
Krinsley JS. Effect of an intensive glucose management protocol on the mortality of critically
ill patients. Mayo Clin Proc 2004;79:977 – 88.
Bochicchio GV, Sung J, Joshi M, et al. Persistent hyperglycemia is predictive of outcome in
critically ill trauma patients. J Trauma 2005;58:921 – 4.
Ligtenberg JJ, Meijering S, Vogelzang M, et al. Relationship of baseline glucose and mortality during medical critical illness? Chest 2005;127:2283.
Christiansen C, Toft P, Jorgensen HS, et al. Hyperglycemia and mortality in critically ill
patients. Intensive Care Med 2004;30:1685 – 8.
Gui D, Spada PL, De Gaetano A, et al. Hypocholesterolemia and the risk of death in the
critically ill surgical patient. Intensive Care Med 1996;22:790 – 4.
Gordon BR, Parker TS, Levine DM, et al. Relationship of hypolipidemia to cytokine concentrations and outcomes in critically ill surgical patients. Crit Care Med 2001;29:1563 – 8.
Bonville DA, Parker TS, Levine DM, et al. The relationships of hypocholesterolemia to
cytokine concentrations and mortality in critically ill patients with systemic inflammatory
response syndrome. Surg Infect (Larchmt) 2004;5:39 – 49.
Lind L, Lithell H. Impaired glucose and lipid metabolism seen in intensive care patients is
related to severity of illness and survival. Clin Intensive Care 1994;5:100 – 5.
Messottten D, Swinnen JV, Vanderhoydonc FV, et al. Contribution of circulating lipids to
the improved outcome of critical illness by glycemic control with intensive insulin therapy.
J Clin Endocrinol Metab 2004;89:1219 – 26.
Goulenok C, Monchi M, Chiche JD, et al. Influence of overweight on ICU mortality:
a prospective study. Chest 2004;125:1441 – 5.
Blanco Quiros A, Casado Flores J, Nieto Moro M, et al. Meningococcal sepsis in pediatrics.
Parameters associated with poor outcome. An Pediatr (Barc) 2004;61:305 – 13.
Barriere SL, Lowry SF. An overview of mortality risk prediction in sepsis. Crit Care Med
1995;23:376 – 93.
Gebhard F, Pfetsch H, Steinbach G, et al. Is interleukin-6 and early marker of injury severity
following major trauma in humans? Arch Surg 2000;135:291 – 5.
Panacek EA, Kaul M. IL-6 as a marker of excessive TNF-alpha activity in sepsis. Sepsis
1999;3:65 – 73.
Wasmuth HE, Kunz D, Graf J, et al. Hyperglycemia at admission to the intensive care unit is
associated with elevated serum concentrations of interleukin-6 and reduced ex vivo secretion
of tumor necrosis factor-alpha. Crit Care Med 2004;32:1109 – 14.
Bouros D, Alexandrakis MG, Antoniou KM, et al. The clinical significance of serum and
endocrine markers
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]
175
bronchoalveolar lavage inflammatory cytokines in patients at risk for acute respiratory distress
syndrome. BMC Pulm Med 2004;4:6.
Gogos CA, Drosou E, Bassaris HP, et al. Pro- versus anti-inflammatory cytokine profile in
patients with severe sepsis: a marker for prognosis and future therapeutic options. J Infect Dis
2000;181:176 – 80.
Pinsky MR, Vincent JL, Deviere J, et al. Serum cytokine levels in human septic shock: relation
to multiple-system organ failure and mortality. Chest 1993;103:565 – 75.
Hack CE, De Groot ER, Felt-Bersma RJ, et al. Increased plasma levels of interleukin-6 in
sepsis. Blood 1989;74:1704 – 10.
Watanabe E, Hirasawa H, Oda S, et al. Extremely high interleukin-6 levels and outcome in the
critically ill are associated with tumor necrosis factor and interleukin-1 gene related polymorphism. Crit Care Med 2005;33:89 – 97.
Panacek EA, Marshall JC, Albertson TE, et al. Efficacy and safety of the monoclonal
anti-tumor necrosis factor antibody F(abV)2 fragment afelimomab in patients with severe sepsis
and elevated interleukin-6. Crit Care Med 2004;32:2173 – 82.
Oberholzer A, Souza SM, Tschoeke SK, et al. Plasma cytokine measurements augment prognostic scores as indicators of outcome in patients with severe sepsis. Shock 2005;23:488 – 93.
Pettila V, Hynnien M, Takkunen O, et al. Predictive value procalcitonin and interleukin-6 in
critically ill patients with suspected sepsis. Intensive Care Med 2002;28:1220 – 5.
Thijs LG, Hack CE. Time course of cytokine levels in sepsis. Intensive Care Med 1999;521:
S258–63.
Raghupathy R, Chaturvedi UC, Al-Sayer H, et al. Elevated levels of IL-8 in dengue hemorrhagic fever. J Med Virol 1998;56:280 – 5.
Marty C, Misset B, Tamion F, et al. Circulating interleukin-8 concentrations in patients with
multiple organ failure of septic and nonseptic origin. Crit Care Med 1994;22:673 – 9.
Parsons PE, Eisner MD, Thompson BT, et al. Lower tidal volume ventilation and plasma cytokine markers of inflammation in patients with acute lung injury. Crit Care Med 2005;33:1 – 6.
Mentula P, Kylanpaa ML, Kemppainen E, et al. Early prediction of organ failure by combined
markers in patients with acute pancreatitis. Br J Surg 2005;92:68 – 75.
Hynninen M, Pettila V, Takkunen O, et al. Predictive value of monocyte histocompatibility
leukocyte antigen-DR expression and plasma interleukin-4 and -10 levels in critically ill patients with sepsis. Shock 2003;20:1 – 4.
Fuse K, Kodama M, Okura Y, et al. Short-term prognostic value of initial serum levels of
interleukin-10 in patients with acute myocarditis. Eur J Heart Fail 2005;7:109 – 12.
Nishii M, Inomata T, Takehana H, et al. Serum levels of interleukin-10 on admission as a
prognostic predictor of human fulminant myocarditis. J Am Coll Cardiol 2004;44:1292 – 7.
Ikuta S, Ono S, Kinoshita M, et al. Interleukin-18 concentration in the peritoneal fluid correlates with the severity of peritonitis. Am J Surg 2003;185:550 – 5.
Sablotzki A, Friedrich I, Muhling J, et al. The systemic inflammatory response syndrome
following cardiac surgery: different expression of proinflammatory cytokines and procalcitonin in patients with and without multiorgan dysfunctions. Perfusion 2002;17:103 – 9.
Feezor RJ, Oberholzer C, Baker HV, et al. Molecular characterization of the acute inflammatory
response to infections with gram-negative versus gram-positive bacteria. Infect Immun 2003;
71:5803 – 13.
Suleiman M, Aronson D, Reisner SA, et al. Admission C-reactive protein levels and 30-day
mortality in patients with acute myocardial infarction. Am J Med 2003;115:695 – 701.
Bonig H, Schneider DT, Sprock I, et al. Sepsis and multi-organ failure: predictors of poor
outcome after hematopoietic stem cell transplantation in children. Bone Marrow Transplant
2000;26:S32–4.
Ridker PM, Cushman M, Stampfer MJ, et al. Plasma concentrations of C-reactive protein and
risk of developing peripheral vascular disease. Circulation 1998;97:425 – 8.
Pinilla JC, Hayes P, Laverty W, et al. The C-reactive protein to prealbumin ratio correlates with
the severity of multiple organ dysfunction. Surgery 1998;124:799 – 805.
176
nylÉn et al
[81] Cano NJ, Pichard C, Roth H, et al. C-reactive protein and body mass index predict outcome
in end-stage respiratory failure. Chest 2004;126:540 – 6.
[82] Lobo SM, Lobo FR, Bota DP, et al. C-reactive protein levels correlate with mortality and
organ failure in critically ill patients. Chest 2003;123:2043 – 9.
[83] Chesney RW, McCarron DM, Haddad JG, et al. Pathogenic mechanisms of the hypocalcemia
of the staphylococcal toxic-shock syndrome. J Lab Clin Med 1983;101:576 – 85.
[84] Becker KL, Nylen ES, White JC, et al. Clinical review 167: procalcitonin and the calcitonin
gene family of peptides in inflammation, infection, and sepsis: a journey from calcitonin back
to its precursors. J Clin Endocrinol Metab 2004;89:1512 – 25.
[85] Snider Jr RH, Nylen ES, Becker KL. Procalcitonin and its component peptides in systemic
inflammation: immunochemical characterization. J Investig Med 1997;45:552 – 60.
[86] Nylen ES, Snider Jr RH, Thompson KA, et al. Pneumonitis-associated hyperprocalcitoninemia. Am J Med Sci 1996;312:12 – 8.
[87] Stryjewski GR, Nylen ES, Bell MJ, et al. Interleukin-6, interleukin-8, and a rapid and sensitive assay for calcitonin precursors for the determination of bacterial sepsis in febrile neutropenic children. Pediatr Crit Care Med 2005;6(2):129 – 35.
[88] Whang KT, Steinwald PM, White JC, et al. Serum calcitonin precursors in sepsis and systemic inflammation. J Clin Endocrinol Metab 1998;83:3296 – 301.
[89] Castelli GP, Pognani C, Meisner M, et al. Procalcitonin and C-reactive protein during systemic inflammatory response syndrome, sepsis and organ dysfunction. Crit Care 2004;8:
R234–42.
[90] Wunder C, Eichelbronner O, Roewer N. Are IL-6, IL-10 and PCT plasma concentrations
reliable for outcome prediction in severe sepsis? A comparison with APACHE III and SAPS II.
Inflamm Res 2004;53:158 – 63.
[91] Van der Kaay DC, De Kleijn ED, De Rijke YB, et al. Procalcitonin as a prognostic marker
in meningococcal disease. Intensive Care Med 2002;28:1606 – 12.
[92] Muller B, Becker KL, Schachinger H, et al. Calcitonin precursors are reliable markers of
sepsis in a medical intensive care unit. Crit Care Med 2000;28:977 – 83.
[93] Clec’h C, Ferriere F, Karoubi P, et al. Diagnostic and prognostic value of procalcitonin in
patients with septic shock. Crit Care Med 2004;32:1166 – 9.
[94] Claeys R, Vinken S, Spapen H, et al. Plasma procalcitonin and C-reactive protein in acute
septic shock: clinical and biological correlates. Crit Care Med 2002;30:757 – 62.
[95] Fritz HG, Brandes H, Bredle DL, et al. Post-operative hypoalbuminaemia and procalcitonin
elevation for prediction of outcome in cardiopulmonary bypass surgery. Acta Anaesthesiol
Scand 2003;47:1276 – 83.
[96] Maxime V, Fitting C, Annane D, et al. Corticoids normalize leukocyte production of macrophage migration inhibitory factor in septic shock. Infect Dis 2005;191:138 – 44.
[97] Bernhagen J, Calandra T, Mitchell RA, et al. MIF is a pituitary-derived cytokine that potentiates lethal endotoxaemia. Nature 1993;365:756 – 9.
[98] Calandra T, Echtenacher B, Roy DL, et al. Protection from septic shock by neutralization of
macrophage migration inhibitory factor. Nat Med 2000;6:164 – 70.
[99] Beishuizen A, Thijs LG, Haanen C, et al. Macrophage migration inhibitory factor and
hypothalamo-pituitary-adrenal function during critical illness. J Clin Endocrinol Metab 2001;
86:2811 – 6.
[100] Bozza FA, Gomes RN, Jappiassu AM, et al. Macrophage migration inhibitory factor levels
correlate with fatal outcome in sepsis. Shock 2004;22:309 – 13.
[101] de Mendonca-Filho HT, Gomes RV, de Almeida Campos LA, et al. Circulating levels of
macrophage migration inhibitory factor are associated with mild pulmonary dysfunction after
cardiopulmonary bypass. Shock 2004;22:533 – 7.
[102] Wang H, Yang H, Tracey KJ. Extracellular role of HMGB1 in inflammation and sepsis.
J Intern Med 2004;255:320 – 31.
[103] Wang H, Bloom O, Zhang M, et al. HMG-1 as a late mediator of endotoxin lethality in mice.
Science 1999;285:248 – 51.
[104] Sunden-Cullberg J, Norrby-Teglund A, Rouhiainen A, et al. Persistent elevation of high
endocrine markers
[105]
[106]
[107]
[108]
[109]
[110]
[111]
[112]
[113]
[114]
[115]
[116]
[117]
[118]
[119]
[120]
[121]
[122]
[123]
[124]
[125]
[126]
177
mobility group box-1 protein (HMGB1) in patients with severe sepsis and septic shock. Crit
Care Med 2005;33:564 – 73.
Hosford D, Paubert-Braquet M, Braquet P. Platelet-activating factor and cytokine interactions
in shock. Resuscitation 1989;18:207 – 18.
Graham RM, Strahan ME, Norman KW, et al. Platelet and plasma platelet-activating factor
in sepsis and myocardial infarction. J Lipid Mediat Cell Signal 1994;9:167 – 82.
Akisu M, Kultursay N, Coker I, et al. Plasma platelet-activating factor levels in newborn
infants with and without perinatal asphyxia: is it an additional marker of perinatal asphyxia?
Acta Paediatr Jpn 1998;40:427 – 31.
Adunsky A, Hershkowitz M, Atar E, et al. Infarct volume, neurological severity and PAF
binding to platelets of patients with acute cerebral ischemic stroke. Neurol Res 1999;21:645 – 8.
Serebruany VL, Gurbel PA, Murugesan SR, et al. Depressed plasma platelet-activating factor
acetylhydrolase in patients presenting with acute myocardial infarction. Cardiology 1998;
90:127 – 30.
Bouchon A, Dietrich J, Colonna M. Inflammatory responses can be triggered by TREM-1,
a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;164:4991 – 5.
Colonna M, Facchetti F. TREM-1 (triggering receptor expressed on myeloid cells-1). A new
player in acute inflammatory responses. J Infect Dis 2003;187:S397–401.
Gibot S, Kolopp-Sarda MN, Béné MC, et al. Plasma level of a triggering receptor expressed on
myeloid cells-1: its diagnostic accuracy in patients with suspected sepsis. Ann Intern Med
2004;141:9 – 15.
Gibot S, Cravoisy A, Levy B, et al. Rapid detection of the soluble form of triggering receptor
expressed on myeloid cells-1 (TREM-1) in the diagnosis of pneumonia. N Engl J Med 2004;
350:451 – 8.
Gibot S, Cravoisy A, et al. Time-course of sTREM (soluble triggering receptor expressed on
myeloid cells)-1, procalcitonin, and C-reactive protein plasma concentrations during sepsis.
Crit Care Med 2005;33:792 – 6.
Argaman Z, Young VR, Noviski N, et al. Arginine and nitric oxide metabolism in critically ill
septic pediatric patients. Crit Care Med 2003;31:591 – 7.
Alzeer AH, Al-Arifi A, Warsy AS, et al. Nitric oxide production is enhanced in patients with
heat stroke. Intensive Care Med 1999;25:58 – 62.
van Dissel JT, Groeneveld PH, Maes B, et al. Nitric oxide: a predictor of morbidity in
postoperative patients? Lancet 1994;343:1579 – 80.
Nijveldt RJ, Teerlink T, Van Der Hoven B, et al. Asymmetrical dimethylarginine (ADMA) in
critically ill patients: high plasma ADMA concentration is an independent risk factor of
ICU mortality. Clin Nutr 2003;22:23 – 30.
Siroen MP, van Leeuwen PA, Nijveldt RJ, et al. Modulation of asymmetric dimethylarginine in
critically ill patients receiving intensive insulin treatment: a possible explanation of reduced
morbidity and mortality? Crit Care Med 2005;33:504 – 10.
Brauner JS, Rohde LE, Clausell N. Circulating endothelin-1 and tumor necrosis factoralpha: early predictors of mortality in patients with septic shock. Intensive Care Med 2000;26:
305 – 13.
Pittet JF, Morel DR, Hemsen A, et al. Elevated plasma endothelin-1 concentrations are associated with the severity of illness in patients with sepsis. Ann Surg 1991;213:261 – 4.
Figueras-Aloy J, Gomez-Lopez L, Rodriguez-Miguelez JM, et al. Plasma endothelin-1 manifestations of neonatal sepsis. J Perinat Med 2004;32:522 – 6.
Bengtsson A. Cascade system activation in shock. Acta Anaesthes Scand 1993;98:S7–10.
Redl H, Schlag G, Bahrami S, et al. Experimental and clinical evidence of leukocyte activation in trauma and sepsis. Prog Clin Biol Res 1994;388:221 – 45.
Heideman M, Norder-Hansson B, Bengtson A, et al. Terminal complement complexes and
anaphylatoxins in septic and ischemic patients. Arch Surg 1988;123:188 – 92.
Kildsgaard J, Hollmann TJ, Matthews KW, et al. Cutting edge: targeted disruption of the C3a
receptor gene demonstrates a novel protective anti-inflammatory role for C3a in endotoxinshock. J Immunol 2000;165:5406 – 9.
178
nylÉn et al
[127] Stevens JH, O’Hanley P, Shapiro JM, et al. Effects of anti-C5a antibodies on the adult respiratory distress syndrome in septic primates. J Clin Invest 1986;77:1812 – 6.
[128] Lazar HL, Bao Y, Gaudiani J, et al. Total complement inhibition: an effective strategy to limit
ischemic injury during coronary revascularization on cardiopulmonary bypass. Circulation
1999;100:1438 – 42.
[129] Fleisig AJ, Verrier ED. Pexelizumab–a C5 complement inhibitor for use in both acute
myocardial infarction and cardiac surgery with cardiopulmonary bypass. Expert Opin Biol Ther
2005;5:833 – 9.
[130] Lorente JA, Garcia-Frade LJ, Landin L, de Pablo R, Torrado C, Renes E, et al. Time course of
hemostatic abnormalities in sepsis and its relation to outcome. Chest 1993;103:1536 – 42.
[131] McManus ML, Churchwell KB. Coagulopathy as a predictor of outcome in meningococcal
sepsis and the systemic inflammatory response syndrome with purpura. Crit Care Med 1993;
21:706 – 11.
[132] Kinasewitz GT, Yan SB, Basson B, et al. Universal changes in biomarkers of coagulation
and inflammation occur in patients with severe sepsis, regardless of causative micro-organism.
Crit Care 2004;8:R82–90.
[133] Esmon CT. The PC pathway. Chest 2003;124:26S – 32S.
[134] Mesters RM, Helterbrand J, Utterback BG, et al. Prognostic value of PC concentrations in
neutropenic patients at high risk of severe septic complications. Crit Care Med 2000;28:
2209 – 16.
[135] Grinnell BW, Joyce D. Recombinant human aPC. A system modulator of vascular function
for treatment of severe sepsis. Crit Care Med 2001;29(7):S53–61.
[136] Mesters RM, Helterbrand J, Utterback BG, et al. Prognostic value of protein C concentrations
in neutropenic patients at high risk of severe septic complications. Crit Care Med 2000;28:
2209 – 16.
[137] Yan SB, Helterbrand JD, Hartman DL, et al. Low levels of protein C are associated with poor
outcome in severe sepsis. Chest 2001;120:915 – 22.
[138] Matthay MA, Ware LB. Plasma protein C levels in patients with acute lung injury: prognostic
significance. Crit Care Med 2004;32:S229–32.
[139] Kollef MH, Eisenberg PR, Shannon W. A rapid assay for detection of circulating D-dimer
is associated with clinical outcomes among critically ill patients. Crit Care Med 1998;26:
1054 – 60.
[140] Iba T, Kidokoro A, Fukunaga M, et al. Association between the severity of sepsis and the
changes in hemostatic molecular markers and vascular endothelial damage markers. Shock
2005;23:25 – 9.
[141] Querol-Ribelles JM, Tenias JM, et al. Plasma d-Dimer levels correlate with outcomes in
patients with community acquired pneumonia. Chest 2004;126:1087 – 92.
[142] Dhainaut JF, Shorr AF, Macias WL, et al. Dynamic evolution of coagulopathy in the first day of
severe sepsis: relationship with mortality and organ failure. Crit Care Med 2005;33:341 – 8.
[143] Boldt J, Wollbruck M, Kuhn D, et al. Do plasma levels of circulating soluble adhesion
molecules differ between surviving and nonsurviving critically ill patients? Chest 1995;107:
787 – 92.
[144] Cummings CJ, Sessler CN, Beall LD, et al. Soluble E-selectin levels in sepsis and critical
illness. Correlation with infection and hemodynamic dysfunction. Am J Respir Crit Care Med
1997;156:431 – 7.
[145] Ueda S, Nishio K, Minamino N, et al. Increased plasma levels of adrenomedullin in patients
with systemic inflammatory response syndrome. Am J Respir Crit Care Med 1999;160:132 – 6.
[146] Papathanassoglou ED, Moynihan JA, Vermillion DL, et al. Soluble fat levels correlate with
multiple organ dysfunction severity, survival and nitrate levels, but not with cellular apoptotic
markers in critically ill patients. Shock 2000;14:107 – 12.
[147] Berendes E, Cullen P, Van Aken H, et al. Endogenous glycosides in critically ill patients. Crit
Care Med 2003;31:1331 – 7.
[148] Kimura F, Itoh H, Ambiru S, et al. Circulating heat-shock protein 70 is associated with postoperative infection and organ dysfunction after liver resection. Am J Surg 2004;187:777 – 84.
endocrine markers
179
[149] Mesotten D, Delhanty PJ, Vanderhoydonc F, et al. Regulation of insulin-like growth factor
binding protein-1 during protracted critical illness. J Clin Endocrinol Metab 2002;87:5516 – 23.
[150] Mishra V, Baines M, Wenstone R, et al. Markers of oxidative damage, antioxidant status
and clinical outcome in critically ill patients. Ann Clin Biochem 2005;42:269 – 76.
[151] Arnalich F, Lopez J, Codoceo R, et al. Relationship of plasma leptin to plasma cytokines
and human survival in sepsis and septic shock. J Infect Dis 1999;180:908 – 11.
[152] Gosling P, Brudney S, McGrath L, et al. Mortality prediction at admission to intensive care:
a comparison of microalbuminuria with acute physiology scores after 24 hours. Crit Care Med
2003;31:98 – 103.
[153] Zakynthinos SG, Papanikolaou S, Theodoridis T, et al. Sepsis severity is the major determinant
of circulating thrombopoietin levels in septic patients. Crit Care Med 2004;32:1004 – 10.
[154] Wright RS, Williams BA, Cramner H, et al. Elevations of cardiac troponin I are associated with
increased short term mortality in noncardiac critically ill emergency department patients. Am J
Cardiol 2002;90(6):634 – 6.
[155] Kayal S, Jais JP, Aguini N, et al. Elevated circulating E-selectin intercellular adhesion molecule 1, and von Willebrand factor in patients with severe infection. Am J Respir Crit Care Med
1998;157:776 – 84.