Editorials
The Nina, the Pinta, and heart rate variability: The search for
prognostic indicators after cardiac arrest*
M
ore than 500 years ago,
Christopher Columbus set
sail west across the Atlantic
searching for better trade
routes with East Asia. Although Columbus and his team never found the route to
the Orient, they opened an age of European exploration of the Americas and
popularized use of the prevailing trade
winds for transcontinental travel (1). In
this issue of Critical Care Medicine, Tiainen and colleagues (2) set out to study
heart rate variability as a potential indicator of prognosis after cardiac arrest.
Although one parameter of heart rate
variability at 48 hours had prognostic implications for patients treated with therapeutic hypothermia, this report stands
out as the first to carefully study electrocardiographic data during therapeutic
hypothermia in patients who had already
experienced a cardiac arrest. As with Columbus, these secondary discoveries may
be the results better remembered.
Therapeutic hypothermia has evolved
from a novel intervention to a standard
treatment for cardiac arrest survivors
with encephalopathy (3– 6). Although
most deaths are due to hypoxic-ischemic
brain injury and not recurrent cardiovascular events (7, 8), concerns regarding
the safety or potential adverse events associated with hypothermia may contribute to clinician reluctance to implement
this important therapy (9, 10). Two recent reviews guiding efforts to predict
outcome after cardiac arrest do not include data from patients treated with hypothermia (11, 12), but do note the potential confounding effect from the 24
hours of hypothermia and the increased
use of sedation and neuromuscular
*See also p. 403.
Key Words: hypothermia; induced/adverse effects;
risk assessment/methods; arrhythmias, cardiac/
adverse effects; predictive value of tests
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e3181959abc
Crit Care Med 2009 Vol. 37, No. 2
blockade associated with it (13). Tiainen
and colleagues provide the best existing
data regarding adverse electrocardiographic events during hypothermia and
describe a tool that may improve prognostic efforts.
As part of the Hypothermia after Cardiac Arrest study, 70 consecutive adult
patients resuscitated from out-of-hospital
ventricular fibrillation at Helsinki University Hospital were randomly assigned
either to therapeutic hypothermia or to
normothermia (2). Twenty-four hour
electrocardiographic recordings were
performed on days 1, 2, and 14, and frequency and time domain assessments of
heart rate variability were performed.
Premature ventricular beats were increased in the hypothermia-treated group
during the first two recordings, but no
difference was noted in the occurrence of
ventricular tachycardia or fibrillation for
patients treated with hypothermia or normothermia. Defibrillation was required
by only one patient during hypothermia
(compared with two patients in the normothermic group). Two of 36 patients
had mean heart rates ⬍50 during hypothermia, and one of these received a temporary pacemaker that fired for ⬍5 minutes. In multivariate analysis, preserved
heart rate variability seemed to predict a
favorable outcome.
By careful documentation of electrocardiographic activity among cardiac arrest survivors treated with hypothermia
and normothermia, Tiainen et al have
added to previous work in neuroprognostication after cardiac arrest (14, 15). Of
perhaps greater applicability, their study
expands our understanding of the safety
of hypothermia among patients with
complex cardiac conditions. Therapeutic
hypothermia was recently shown to be
safe and beneficial in patients with cardiogenic shock treated with intra-aortic
balloon pumps and in conjunction with
percutaneous coronary intervention (16,
17). The present study confirms that ventricular arrhythmias are common, but
not more prevalent in patients receiving
hypothermia, and that bradycardia is common but rarely requires treatment. These
findings increase our understanding and
are based on data from more reliable monitoring than in existing reports.
Just as Europeans provided information facilitating further exploration of the
“new world” 500 years ago, Tiainen’s
group provides data that advances our
understanding of therapeutic hypothermia. We can only hope that many other
investigators will travel these waters to
discover better and safer ways of providing this important therapy and to furnish
us with tools to better predict outcomes.
Richard R. Riker, MD, FCCP
David B. Seder, MD
Gilles L. Fraser, PharmD, FCCM
Neurosciences Institute
Maine Medical Center
Portland, Maine
REFERENCES
1. Available at: http://earthguide.ucsd.edu/
virtualmuseum/climatechange1/08_1.shtml.
Accessed August 6, 2008
2. Tiainen M, Parikka HJ, Mäkijärvi MA, et al:
Arrhythmias and heart rate variability during
and after therapeutic hypothermia for cardiac arrest. Crit Care Med 2009: 37:403-409
3. Bernard SA, Gray TW, Buist MD, et al: Treatment of comatose survivors of out-ofhospital cardiac arrest with induced hypothermia. N Engl J Med 2002; 346:557–563
4. HACA Study Group: Mild therapeutic hypothermia to improve the neurologic outcome
after cardiac arrest. N Engl J Med 2002; 346:
549 –556
5. Anonymous: American Heart Association
2005 guidelines for CPR and ECC. Part 7.5.
Postresuscitation support. Circulation 2005;
112 (Suppl 24):IV-84 –IV-88
6. International Liaison Committee on Resuscitation: International consensus on cardiopulmonary resuscitation and emergency cardiovascular care science with treatment
recommendations. Circulation 2005; 112:III1–III-136
7. Oddo M, Ribordy V, Feihl F, et al: Early
predictors of outcome in comatose survivors
of ventricular fibrillation and non-ventricular fibrillation cardiac arrest treated with hypothermia: A prospective study. Crit Care
Med 2008; 36:2296 –2301
735
8. Seder DB, Jarrah S, Lord C, et al: Effect of a
therapeutic hypothermia protocol on mortality in out-of hospital cardiac arrest: Experiences at a tertiary care hospital. Proc ATS
2007; 4:A792
9. Holzer M, Bernard SA, Hachimi-Idrissi S, et
al: Hypothermia for neuroprotection after
cardiac arrest: Systematic review and individual patient data meta-analysis. Crit Care
Med 2005; 33:414 – 8
10. Brooks SC, Morrison LJ: Implementation of
therapeutic hypothermia guidelines for postcardiac arrest syndrome at a glacial pace: Seeking guidance from the knowledge translation literature. Resuscitation 2008; 77:286–292
11. Booth CM, Boone RH, Tomlinson G, et al: Is
this patient dead, vegetative, or severely neu-
rologically impaired? Assessing outcome for
comatose survivors of cardiac arrest. JAMA
2004; 291:870 – 879
12. Wijdicks EFM, Hijdra A, Young GB, et al:
Practice parameter: Prediction of outcome in
comatose survivors after cardiopulmonary
resuscitation (an evidence-based review).
Neurology 2006; 67:203–210
13. Sunde K, Dunlop O, Rostrup M, et al: Determination of prognosis after cardiac arrest
may be more difficult after introduction of
therapeutic hypothermia. Resuscitation
2006; 69:29 –32
14. Tiainen M, Roine RO, Pettilä V, et al: Serum
neuron-specific enolase and S-100B protein
in cardiac arrest patients treated with hypothermia. Stroke 2003; 34:2881–2886
15. Tiainen M, Kovala TT, Takkunen OS, et al:
Somatosensory and brainstem auditory
evoked potentials in cardiac arrest patients
treated with hypothermia. Crit Care Med
2005; 33:1736 –1740
16. Wolfrum S, Pierau C, Radke PW, et al: Mild
therapeutic hypothermia in patients after
out-of-hospital cardiac arrest due to acute
ST-segment elevation myocardial infarction
undergoing immediate percutaneous coronary intervention. Crit Care Med 2008; 36:
1780 –1786
17. Oddo M, Schaller MD, Feihl F, et al: From
evidence to clinical practice: Effective implementation of therapeutic hypothermia to improve patient outcome after cardiac arrest.
Crit Care Med 2006; 34:1865–1873
Hemodynamic support of shock state: Are we asking the
right questions?*
S
eptic shock is one of the most
challenging problems in the
critical care. Its mortality toll in
the United States ranges between 200,000 and 250,000, a number
comparable with myocardial infraction. Diagnosing and treating a septic shock is like
looking the stars at night: from single
bright spots, you have to reconstitute the
constellations to have the complete picture.
For the last 40 years, catecholamines
have been used routinely in shock state,
trying to restore normal or near-normal
hemodynamic parameters. The rationale
is to maintain a minimal level of blood
pressure in septic shock patients (1).
From a quasi-empirical use, more and
more knowledge has emerged on the
mechanisms and effect of these drugs.
Our understanding of shock state improves, including myocardial depression
in septic shock, the links between inflammation and coagulation, and microcirculation and cellular energetics.
Our pharmacologic tool set expanded
and include vasopressin and analogs,
phosphodiesterase inhibitors, and cal-
*See also p. 410.
Key Words: septic shock; catecholamines; outcome; sepsis bundle; quality control
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e3181959abc
736
cium sensitizers. As knowledge expand,
the picture gets more complex and the clinicians more confused.
The Quest for the Magic Bullet
In this issue of Critical Care Medicine,
Povoa et al (2) adds to the comparison of
different catecholamine in shock states.
They observed a large multicentered population of septic shock patients, the various catecholamines used, and the patient’s
outcome. They show that dopamine, norepinephrine, and dobutamine increase
mortality. Should we ban these drugs
from our pharmacopoeia? Certainly not.
First, the natural catecholamines are part
of the acute-phase response to physiologic
stress and are essential for human species.
These molecules are part of our survival kit
(3). Second, using a similar strategy, others
(4) observed that that dopamine decreases
mortality in septic shock patients and that
norepinephrine and dobutamine have no
effect. This is contrary to the conclusion in
the present study. Other cohorts have investigated this question with variable results (Table 1). One can argue that these
are not randomized control trials, but most
randomized control studies also failed to
point out a consistent effect. Third, most of
the studies do not start with patients on no
adrenergic support: when a certain level of
dopamine or norepinephrine is reached
randomize into replace by or add “A” vs.
“B.” First, the picture is blurred. Catecholamines at a usual pharmacologic
range yield to concentrations 100 times
above the physiologic concentrations.
Second, there is a significant interindividual variability in catecholamine kinetics: a fixed dose of dopamine can yield to
plasma concentrations in a 20-fold range.
Third, dopamine is the natural direct precursor of norepinephrine through betahydroxylase. During dopamine infusion
(3 g/kg/min), plasma norepinephrine
concentration increases (5).
Should We Change Our
Approach to Shock States
Until recently, hemodynamic support
of shock state was focused on restoring
normal or near-normal physiologic parameters. In the 1980s, there was even a
tendency toward supranormal physiologic goals. To easily achieve target hemodynamic parameters, having one single magical drug would make the
clinician’s life easier. There is a quest
toward the magic bullet applicable in all
patients: Is dopamine better than norepinephrine? Is vasopressin better than norepinephrine? What is the optimal target
for mean arterial blood pressure? What is
the optimal cardiac output/mixed venous
saturation? Consensus panel supported
the use of catecholamines in septic shock
with a grade E evidence (6). Clinical investigations suggest that increasing the
target mean arterial pressure (MAP) from
65 to 85 mm Hg does not change oxygenCrit Care Med 2009 Vol. 37, No. 2
Table 1. Some studies investigating the effect of vasopressors in vasodilatory shock in adults
Study
Cohort studies
Goncalves, et al (26)
Patients
Methods
406 patients requiring NE
Main Conclusions
Observational
prospective cohort
Observational
prospective cohort
Observational
prospective cohort
Observational
multicenter study
NE does not facilitate development
of MODS
NE lower mortality when
compared to Dopa
Fixed dose AVP is comparable with
titrated doses of NE or Dopa
Dopa tends to worsen mortality
NE and Dobu has no effect
137 septic shock patients all
receiving NE, some with
AVP
458 septic shock patients
Observational
prospective cohort
Patients receiving AVP plus NE
have a worse mortality than
patients receiving NE
Dopa decreases mortality. NE and
Dobu increases mortality
Randomized control trials
Martin, et al (30)
32 patients in septic shock
NE vs. Dopa
Ruokonen, et al (31)
10 patients in septic shock
Dopa alone or with NE
Marik, et al (32)
20 septic shock patients
Dopa vs. NE
Levy, et al (33)
30 patients with septic shock
Epi vs. fixed Dobu plus
NE
Malay, et al (34)
AVP vs. placebo plus NE
Seguin, et al (35)
10 trauma patients with
vasodilatory shock
22 patients in septic shock
Patel, et al (36)
24 patients in septic shock
Dunser, et al (37)
48 patients with vasodilatory
shock
Albanese, et al (38)
20 septic shock patients
Lauzier, et al (39)
23 patients in septic shock
First-line treatment: NE
versus AVP alone,
rescue by adding the
other drug
Schmoelz, et al (40)
61 patients with septic shock
NE plus dopexamine or
plus Dopa
Annane, et al (41)
330 patients with septic
shock
NE plus Dobu vs. Epi
Russell, et al (42)
778 patients in septic shock
NE vs. AVP
Martin, et al (27)
97 septic shock patients
Hall, et al (28)
150 patients receiving either
Dopa NE or AVP
462 septic shock patients
Sakr, et al (4)
Micek, et al (29)
Povoa, et al (2)
Observational
multi center study
Titrated Epi vs. fixed
Dobu plus titrated
NE
AVP vs. NE fixed dose
plus open label NE
AVP 4 U/hr vs. placebo
plus norEpi to MAP
⬎70
Terlipressin vs. NE
NE provided a better
hemodynamic profile, No
difference in outcome
Discrepancy between global and
regional blood flow. No
difference in outcome
NE improve splanchnic perfusion.
No difference in outcome
Trend toward less oliguria with NE
plus Dobu, No differences in
mortality
Trend toward less mortality in the
AVP group
No difference in outcome
Better creatinine clearance with
AVP, no difference in outcome
Better hemodynamic response
with AVP but no difference in
outcome
No differences in MAP, renal
function and outcome
No difference in outcome
Comments
Significant international
variation in
catecholamine use
AVP was used as a rescue
therapy in NE resistant
patients
Very small n, 24 hr
follow-up
4 hr follow-up
Open label. 85% of AVP
patients received
additional NE due to
MAP ⬍70 mm Hg at
1 hr
Dopexamine associated with better
renal function. No difference in
organ failures or mortality
No difference in hemodynamic,
organ failure, duration of
therapy, and mortality
Better survival with AVP in less
sick patients. No overall
mortality difference
NE, norepinephrine; AVP, arginine-vasopressin; Dobu, dobutamine; Dopa, dopamine; Epi, epinephrine; norEpi, norepinephrine; MAP, mean arterial
pressure; MODS, Multiple Organs Dysfunction Syndrome.
ation parameter and skin microcirculation (7) no renal function or outcome (8).
Our actual concept is that below a certain
MAP, blood flow is linearly dependent on
organ perfusion (1). This cutoff is probably not the same in the various organs
and probably different between different
various capillary beds of the same organ.
Crit Care Med 2009 Vol. 37, No. 2
The inlet pressure of physiologic capillaries is 20 –25 mm Hg. There are no data to
suggest that 50 mm Hg MAP is more
deleterious in term of microcirculatory
and organ perfusion than 65 with vasopressors. Sepsis induces a state of nutrient and oxygen deficiency at the cellular
level. This cellular energetic failure lead
to organ dysfunction, myocardial depression, and microcirculatory dysfunction
(Fig. 1). The myocardium is energetically
exhausted. Increasing the catecholamines
level is like whipping an exhausted horse.
With worsening cellular dysfunction, the
horse will not respond the whip—the
shock will became refractory to cat737
Figure 1. Proposed physiopathology of septic shock. Proposed physiopathologic links between sepsis,
mitochondrial energetic failure, and organ dysfunction and death. Therapeutic target is shown in the
gray boxes. Innovative therapies and new effects of therapies are shown with a question mark. iNO,
inducible nitrous oxide.
echolamines. We can change whip—as by
using vasopressin or analogs— but the
underlying problem will remain and, if
not corrected, shock will worsen and be
refractory to vasopressin. This may explain why “dopamine sensitive” patients
in septic shock have a better outcome
than “dopamine resistant” patients (9): In
dopamine-resistant shock, the cellular
energetic is failing. This may also explain
that in the Vasopressin and Septic Shock
Trial, the patients who may benefit from
the change of whip—switch to vasopressin—are those requiring the lowest doses
of norepinephrine—the not too exhausted
horses. Researchers have started to explore
the link between hemodynamic and microcirculation: Trzeciak et al (10) showed that
there is a correlation between survival and
microcirculatory disturbances. Sakr et al
(11) showed that small vessel perfusion improved over time in septic shock survivors
but not in nonsurvivors. Despite similar
hemodynamic parameters and amount of
support, patients dying after the resolution
of shock in multiple organ failure had a
lower percentage of perfused small vessels
than did survivors. This shows that the cellular dysfunction and microcirculatory disturbances can still be present despite restored hemodynamics. Following these
738
hypothesis, innovative and provocative,
strategies have been proposed in septic
shock. Spronk et al (12) administered nitroglycerine in septic shock patients and
showed an improvement in the microcirculation. De Backer et al (13) demonstrated
that dobutamine can improve but not restore microcirculatory perfusion in patients
with septic shock, independently of its inotropic effect. Going one step upstream (Fig.
1), Levy et al (14) measured intermediate
substrate metabolism in septic shock patients and pointed out a mitochondrial
dysfunction. Some have suggested that
lactate is not a waste product but a highoctane fuel and an adaptive mechanism
during shock states (15). The epinephrine-induced release of glucose and lactate from the muscles during shock may
be a survival mechanism aiming to feed a
starved horse (16). Recently, Regueira et
al (17) showed that norepinephrine was
able to increase maximal mitochondrial
respiration in the liver during an endotoxin challenge. This suggests that in
shock, the so-called cytopathic hypoxia is
related to mitochondrial substrate availability (18). Obviously, this view of septic
shock is simplistic: There are probably
two types of feedback and control sys-
tems: First, standard feedback such as
improvement of hemodynamic parameter
should be associated with improved microcirculation and cell oxygenation. Second, control of hemodynamic parameters
may help to avoid vicious circles such as
worsening of inflammatory response because of ongoing shock state (19). With
this in mind (Fig. 1), hemodynamic support alone has little chances to reverse
the complete cascade. It will be hard to
find a link between the type of catecholamine and the outcome. This will also
require that further randomized trial investigating the effect of hemodynamic
support in septic shock patients will have
to include a homogeneous patient group
and control all these other therapies.
Dobutamine and norepinephrine have
recently shown unexpected positive side
effects in shock state at the microcirculatory and mitochondrial level.
Some intensivists dream about the
multicentered international randomized
control trial comparing norepinephrine
vs. dopamine vs. vasopressin in septic
shock patients: The design would be
study drug “X” vs. “Y” in a concealed bag
titrated to MAP of 70 mm Hg. What is the
best in terms of MAP? What is the best in
terms of outcome? Maybe we should
change from “what is the best in term of
MAP?” to “what is the best for the mitochondria?” (20).
Sepsis Bundle Is a Whole
Package: Take it as a Whole
Following the fundamental study by
Rivers (21), several consensus conferences proposed a structured multisystem
approach to patients in septic shock (22).
Items of a bundle are inseparable: It is
not a flexible contract. The strength of
these bundle and outcome improvement
come from 1) a standardized approach
(23), 2) the check list effect avoiding
missed items, and 3) the speed of intervention including time points were goals
should be met (6 and 24 hours). Compliance to the surviving sepsis campaign
bundle in the SACiUCI study was also
published as a preliminary abstract (24).
The compliance to selected items of the
6-hour bundle ranges from 30% (antibiotics within 3 hours) to 80% (vasopressors after adequate fluid). Using an allor-none approach, ⬎70% of the patients
failed the 6-hour bundle. Failing the
6-hour bundle is associated with an increased mortality in the present cohort
(24). There are three strategies to assess
Crit Care Med 2009 Vol. 37, No. 2
compliance to a set of interventions/
markers (25): 1) As Povoa et al presented
as item-by-item measurement, 2) as a
composite variable i.e., four items of six,
or 3) using an all-or-none measurement.
The outcome improvement does not
come from a specific intervention or a
specific catecholamine. It is time to raise
the bar and assess the surviving sepsis
campaign recommendation as an inseparable package. We should go further than
the 6-hour limit: In parallel to data from
myocardial infarction regarding door-toballoon time, we should focus on a door
to sepsis bundle time. Despite an excellent worldwide campaign, endorsement
by a dozen of critical care societies and
organizations, practice has room for improvement. Sepsis is like myocardial infarction: its an emergency!
David Bracco, MD
Department of Anesthesia and
Critical Care, McGill University
Health Center—Anesthesia
Montreal General Hospital
Montreal, Quebec, Canada
REFERENCES
1. Leone M, Martin C: Vasopressor use in septic
shock: An update. Curr Opin Anaesthesiol
2008; 21:141–147
2. Póvoa PR, Carneiro AH, Ribeiro OS, et al:
Influence of vasopressor agent in septic
shock mortality. Results from the Portuguese community-acquired sepsis study
(SACiUCI). Crit Care Med 2009; 37:410 – 416
3. Roizen MF: Should we all have a sympathectomy at birth? Or at least preoperatively?
Anesthesiology 1988; 68:482– 484
4. Sakr Y, Reinhart K, Vincent JL, et al: Does
dopamine administration in shock influence
outcome? Results of the Sepsis Occurrence
in Acutely Ill Patients (SOAP) Study. Crit
Care Med 2006; 34:589 –597
5. Lieverse AG, Lefrandt JD, Girbes AR, et al:
The effects of different doses of dopamine
and domperidone on increases of plasma
norepinephrine induced by cold pressor test
in normal man. Hypertens Res 1995;
18(Suppl 1):S221–S224
6. Beale RJ, Hollenberg SM, Vincent JL, et al:
Vasopressor and inotropic support in septic
shock: An evidence-based review. Crit Care
Med 2004; 32:S455–S465
7. LeDoux D, Astiz ME, Carpati CM, et al: Effects of perfusion pressure on tissue perfusion in septic shock. Crit Care Med 2000;
28:2729 –2732
8. Bourgoin A, Leone M, Delmas A, et al: Increasing mean arterial pressure in patients
with septic shock: Effects on oxygen variables and renal function. Crit Care Med 2005;
33:780 –786
Crit Care Med 2009 Vol. 37, No. 2
9. Levy B, Dusang B, Annane D, et al: Cardiovascular response to dopamine and early prediction of outcome in septic shock: A prospective multiple-center study. Crit Care
Med 2005; 33:2172–2177
10. Trzeciak S, Dellinger RP, Parrillo JE, et al:
Early microcirculatory perfusion derangements in patients with severe sepsis and septic shock: Relationship to hemodynamics,
oxygen transport, and survival. Ann Emerg
Med 2007; 49:88 –98, 98.e1–2
11. Sakr Y, Dubois MJ, De Backer D, et al: Persistent microcirculatory alterations are associated with organ failure and death in patients with septic shock. Crit Care Med 2004;
32:1825–1831
12. Spronk PE, Ince C, Gardien MJ, et al: Nitroglycerin in septic shock after intravascular
volume resuscitation. Lancet 2002; 360:
1395–1396
13. De Backer D, Creteur J, Dubois MJ, et al: The
effects of dobutamine on microcirculatory
alterations in patients with septic shock are
independent of its systemic effects. Crit Care
Med 2006; 34:403– 408
14. Levy B, Sadoune LO, Gelot AM, et al: Evolution of lactate/pyruvate and arterial ketone
body ratios in the early course of catecholamine-treated septic shock. Crit Care
Med 2000; 28:114 –119
15. Matejovic M, Radermacher P, Fontaine E:
Lactate in shock: A high-octane fuel for the
heart? Intensive Care Med 2007; 33:
406 – 408
16. Levy B, Mansart A, Montemont C, et al: Myocardial lactate deprivation is associated with
decreased cardiovascular performance, decreased myocardial energetics, and early
death in endotoxic shock. Intensive Care
Med 2007; 33:495–502
17. Regueira T, Banziger B, Djafarzadeh S, et al:
Norepinephrine to increase blood pressure in
endotoxaemic pigs is associated with improved hepatic mitochondrial respiration.
Crit Care 2008; 12:R88
18. Leverve XM: Mitochondrial function and
substrate availability. Crit Care Med 2007;
35:S454 –S460
19. Rivers EP, Kruse JA, Jacobsen G, et al: The
influence of early hemodynamic optimization on biomarker patterns of severe sepsis
and septic shock. Crit Care Med 2007; 35:
2016 –2024
20. Wagner F, Radermacher P, Georgieff M, et al:
Sepsis therapy: What’s the best for the mitochondria? Crit Care 2008; 12:171
21. Rivers E, Nguyen B, Havstad S, et al: Early
goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med
2001; 345:1368 –1377
22. Dellinger RP, Levy MM, Carlet JM, et al:
Surviving Sepsis Campaign: International
guidelines for management of severe sepsis
and septic shock: 2008. Crit Care Med 2008;
36:296 –327
23. Micek ST, Roubinian N, Heuring T, et al:
Before-after study of a standardized hospital
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
order set for the management of septic
shock. Crit Care Med 2006; 34:2707–2713
Cardoso T, Carneiro A, Ribeiro O, et al: The
Portuguese network data: Compliance with
the Surviving Sepsis Campaign bundles 关Abstract 0071兴. Intensive Care Med 2006; 32:
S23
Nolan T, Berwick DM: All-or-none measurement raises the bar on performance. JAMA
2006; 295:1168 –1170
Goncalves JA Jr, Hydo LJ, Barie PS: Factors
influencing outcome of prolonged norepinephrine therapy for shock in critical surgical illness. Shock 1998; 10:231–236
Martin C, Viviand X, Leone M, et al: Effect of
norepinephrine on the outcome of septic
shock. Crit Care Med 2000; 28:2758 –2765
Hall LG, Oyen LJ, Taner CB, et al: Fixed-dose
vasopressin compared with titrated dopamine and norepinephrine as initial vasopressor therapy for septic shock. Pharmacotherapy 2004; 24:1002–1012
Micek ST, Shah P, Hollands JM, et al: Addition of vasopressin to norepinephrine as independent predictor of mortality in patients
with refractory septic shock: An observational study. Surg Infect (Larchmt) 2007;
8:189 –200
Martin C, Papazian L, Perrin G, et al: Norepinephrine or dopamine for the treatment
of hyperdynamic septic shock? Chest 1993;
103:1826 –1831
Ruokonen E, Takala J, Kari A, et al: Regional
blood flow and oxygen transport in septic
shock. Crit Care Med 1993; 21:1296 –1303
Marik PE, Mohedin M: The contrasting effects of dopamine and norepinephrine on
systemic and splanchnic oxygen utilization
in hyperdynamic sepsis. JAMA 1994; 272:
1354 –1357
Levy B, Bollaert PE, Charpentier C, et al:
Comparison of norepinephrine and dobutamine to epinephrine for hemodynamics,
lactate metabolism, and gastric tonometric
variables in septic shock: A prospective, randomized study. Intensive Care Med 1997; 23:
282–287
Malay MB, Ashton RC Jr, Landry DW, et al:
Low-dose vasopressin in the treatment of vasodilatory septic shock. J Trauma 1999; 47:
699 –703; discussion 703– 695
Seguin P, Bellissant E, Le Tulzo Y, et al:
Effects of epinephrine compared with the
combination of dobutamine and norepinephrine on gastric perfusion in septic shock.
Clin Pharmacol Ther 2002; 71:381–388
Patel BM, Chittock DR, Russell JA, et al:
Beneficial effects of short-term vasopressin
infusion during severe septic shock. Anesthesiology 2002; 96:576 –582
Dunser MW, Mayr AJ, Ulmer H, et al: Arginine vasopressin in advanced vasodilatory
shock: A prospective, randomized, controlled
study. Circulation 2003; 107:2313–2319
Albanese J, Leone M, Delmas A, et al: Terlipressin or norepinephrine in hyperdynamic
septic shock: A prospective, randomized
study. Crit Care Med 2005; 33:1897–1902
739
39. Lauzier F, Levy B, Lamarre P, et al: Vasopressin or norepinephrine in early hyperdynamic
septic shock: A randomized clinical trial. Intensive Care Med 2006; 32:1782–1789
40. Schmoelz M, Schelling G, Dunker M, et al:
Comparison of systemic and renal effects of
dopexamine and dopamine in norepinephrine-treated septic shock. J Cardiothorac
Vasc Anesth 2006; 20:173–178
41. Annane D, Vignon P, Renault A, et al: Norepinephrine plus dobutamine versus epinephrine alone for management of septic
shock: A randomised trial. Lancet 2007; 370:
676 – 684
42. Russell JA, Walley KR, Singer J, et al: Vasopressin versus norepinephrine infusion in
patients with septic shock. N Engl J Med
2008; 358:877– 887
Treatment of septic renal injury by alkaline phosphatase:
An emperor with new clothes*
I
n this issue of Critical Care Medicine, Heemskerk et al (1) describe
a clinical trial on the effect of
treatment with purified bovine intestinal alkaline phosphatase (AF) vs. placebo in a small series (n ⫽ 36) of patients
with severe sepsis or septic shock from
Gram-negative and Gram-positive microorganisms and having (impending or
manifest) acute kidney injury and failure.
Indeed, AF is capable of detoxifying endotoxin, even at physiologic concentrations,
by dephosphorylation of the lipid A moiety of lipopolysaccharide (2), and exogenous administration in animals with
Gram-negative sepsis and shock appeared
beneficial (3, 4). The Heemskerk et al (1)
trial was too small to discern a morbidity
or survival benefit of AF treatment, but,
nevertheless, the authors observed some
effect on renal function parameters. In a
small substudy (in patients not on renal
replacement therapy), in which the effect
of AF seemed even more pronounced,
protection appeared associated with less
inducible nitric oxide synthase (iNOS)derived nitric oxide (NO) production and
release of markers of the (resultant?) injury in the tubular cells and excreted in
the urine. These (selected) observations
should be regarded as preliminary, and
protection by AF of renal function in
sepsis and shock should be confirmed
in larger studies with stronger end
points, such as the need for renal replacement therapy, the speed of recovery of septic renal failure, and alike,
*See also p. 417.
Key Words: alkaline phosphatase; renal injury;
septic shock; nitric oxide synthase
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194c12c
740
which have not been studied by Heemskerk et al (1). In contrast, the authors
have focused on acute kidney injury
criteria including serum creatinine,
even in patients already on renal replacement therapy at the start. We cannot formally exclude that AF affected
tubular creatinine excretion rather
than glomerular filtration. Furthermore, the mechanisms behind this potentially beneficial and relatively specific effect on the kidney remain to be
demonstrated, because, among others,
it is unclear how AF would also benefit
patients with Gram-positive septic
shock.
Endogenous AF is ubiquitous and
abundant in epithelial cells and serves,
among others, as an ectonucleotidase, to
dephosphorylate and degrade extravascular (high energy, monoester) phosphate
compounds. There are various isoenzymes for different tissues, which, for
instance, play a role in bone turnover,
bile excretion, and placental development and function (5). Expressed in the
mucosa of the gastrointestinal tract, AF
may help to limit translocation of
harmful endotoxin from the lumen (5).
The function of the enzyme located in
the brush border of the proximal tubule
is unclear and may only partly relate to
resorption of phosphate. It is shed and
excreted in the urine in the course of
tubular injury and may be upregulated
in the kidney during (experimental)
sepsis (6, 7).
Extracellular phosphate compounds,
released by various cells and tissues particularly on hypoxia or inflammation,
may, via nucleotide signaling and purinergic receptors, affect a wide variety of
processes, involving innate immunity,
epithelial transport, and regulation of
blood flow (8 –10). This applies to both
adenosine triphosphate and its dephos-
phorylation products adenosine diphosphate, adenosine monophosphate, and
adenosine, and the kidney, but effects depend on specific receptor stimulations
and conditions (11). For instance, the
compounds are vasodilators in normal
rats but vasoconstrictors (also in the renal bed) in endotoxin-challenged animals
(12). Adenosine triphosphate and adenosine, being mediators of tubuloglomerular feedback, may also decrease renal
blood flow by afferent vasoconstriction
via A1-adenosine receptor stimulation (8,
11, 13, 14). The compounds may have
proinflammatory actions so that (specific) adenosine (receptor) antagonists
are protective, whereas, in contrast, stimulation of some (A2A) adenosine receptors
may have anti-inflammatory actions,
even in the kidney (15, 16). We also know
that inhibitors of phosphodiesterases, degrading phosphate diesters, such as cyclic
adenosine monophosphate to adenosine
can be protective in experimental acute
kidney injury after a wide variety of challenges, including endotoxemia (17). How
AF might interfere with these processes is
largely unknown. Indeed, the study by
Heemskerk et al (1) does not give insight
into the specific effect or the routing of
AF in the kidney during sepsis. Nevertheless, it suggests that iNOS upregulation
is associated, even perhaps in a causative
manner, with proximal tubular damage
and, thereby, contributes to acute kidney
injury, as observed before (18). Oxidative
stress, NO-derived peroxynitrite and subsequent mitochondrial and nuclear DNA
damage, and protein nitrosylation may
be some of the mechanisms underlying
iNOS–NO-derived toxicity. This may
also explain, at least in part, the often
presumed maintenance of renal blood
flow and the observed fall in filtration
fraction and glomerular filtration in patients with impending acute renal failCrit Care Med 2009 Vol. 37, No. 2
ure during hyperdynamic sepsis (19).
We cannot judge these mechanisms
from the data in the study by Heemskerk et al (1), and, therefore, the clinical significance of potential renal AFinduced adenosine and iNOS-derived
NO and associated benefits and harms
remain to be elucidated.
Taken together, the preliminary
study by Heemskerk et al (1), done in a
difficult-to-study patient population,
addresses important and partly novel
issues on the role of AF, acute kidney
injury, and iNOS in human septic
shock. However, many questions remain on the new clothes of the emperor
(AF) and, particularly, how they are tied
together. The interesting observations
need confirmation in a larger trial.
A. B. Johan Groeneveld, MD,
PhD, FCCP, FCCM
Department of Intensive Care,
VUMC—Intensive Care
Amsterdam, The Netherlands
Marc G. Vervloet, MD
Department of Nephrology,
VUMC—Intensive Care
Amsterdam, The Netherlands
REFERENCES
1. Heemskerk S, Masereeuw R, Moesker O, et al:
Alkaline phosphatase treatment improves renal
function in severe sepsis or septic shock patients. Crit Care Med 2009; 37:417– 423
2. Koyama I, Matsunaga T, Harada T, et al:
Alkaline phosphatases reduce toxicity of lipopolysaccharides in vivo and in vitro through
dephosphorylation. Clin Biochem 2002; 35:
455– 461
3. Verweij WR, Bentala H, Huizinga-van der
Vlag A, et al: Protection against an Escherichia coli-induced sepsis by alkaline phosphatase in mice. Shock 2004; 22:174 –179
4. van Veen SQ, van Vliet AK, Wulferink M, et
al: Bovine intestinal alkaline phosphatase attenuates the inflammatory response in secondary peritonitis in mice. Infect Immun
2005; 73:4309 – 4314
5. Millán JL: Alkaline phosphatases. Structure,
substrate specificity, and functional relatedness to other members of a large superfamily
of enzymes. Purinergic Signal 2006;
2:335–341
6. Kapojos JJ, Poelstra K, Borghuis T, et al:
Induction of glomerular alkaline phosphatase after challenge with lipopolysaccharide.
Int J Exp Path 2003; 84:135–144
7. Trof RJ, Di Maggio F, Leemreis J, et al: Biomarkers of acute renal injury and renal failure. Shock 2006; 26:245–253
8. Komlosi P, Fintha A, Bell PD: Renal cell-tocell communication via extracellular ATP.
Physiology 2005; 20:86 –90
9. Bours MJL, Swennen ELR, Di Virgilio F, et
al: Adenosine 5⬘-triphosphate and adenosine
as endogenous signaling molecules in immunity and inflammation. Pharmacol Ther
2006; 112:358 – 404
10. Vallon V: P2 receptors in the regulation of
renal transport mechanisms. Am J Physiol
Renal Physiol 2008; 294:F10 –F27
11. Guan Z, Osmond DA, Inscho EW: Purinoceptors in the kidney. Exp Biol Med 2007; 232:
715–726
12. Jolly L, March JE, Kemp PA, et al: Regional
haemodynamic responses to adenosine receptor activation vary across time following lipopolysaccharide treatment in conscious rats. Br J Pharmacol 2008; 154:
1600 –1610
13. Vallon V, Mühlbauer B, Osswald H: Adenosine and kidney function. Physiol Rev 2006;
86:901–940
14. Castrop H: Mediators of tubuloglomerular
feedback regulation of glomerular filtration:
ATP and adenosine. Acta Physiol Scand
2007; 189:3–14
15. Linden J: Molecular approach to adenosine
receptors: Receptor-mediated mechanisms of
tissue protection. Annu Rev Pharmacol Toxicol 2001; 41:775–787
16. Okusa MD: A2A adenosine receptor: A novel
therapeutic target in renal disease. Am J
Physiol Renal Physiol 2002; 282:F10 –F18
17. Wang W, Zolty E, Falk S, et al: Pentoxifylline
protects against endotoxin-induced acute renal failure in mice. Am J Physiol Renal
Physiol 2006; 291:F1090 –F1095
18. Wang W, Zolty E, Falk S, et al: Endotoxemiarelated acute kidney injury in transgenic
mice with endothelial overexpression of GTP
cyclohydrolase-1. Am J Physiol Renal
Physiol 2008; 294:F571–F576
19. Wan L, Bagshaw SM, Langenberg C, et al:
Pathophysiology of septic acute kidney injury: What do we really know? Crit Care Med
2008; 36:S198 –S203
Is target population more important than patient location when
evaluating tight glycemic control?*
C
linical trials in the intensive
care unit can be divided into
two categories. The first category includes disease- or syndrome-specific trials that examine the effect of a drug or practice on a group of
patients with that specific disease- or syndrome. Examples of this type of trial
*See also p. 424.
Key Words: tight glucose control; fibrinolysis; sepsis; insulin therapy; plasminogen activator inhibitor-1
Supported by SCCM Vision Grant (BRG); Supported
by K-23 GMO7-1399 (JES).
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318195468e
Crit Care Med 2009 Vol. 37, No. 2
would be the use of lung-protective ventilation in patients with acute lung injury
(1) or the use of steroids in patients with
septic shock (2). The second category of
trial enrolls patients with a particular severity of disease that leads to admission
to an intensive care unit. This type of
treatment-specific trial would include patients with different types of disease that
precipitate similar severity of illness and
location within an intensive care unit.
Examples of this type of trial would include the Saline versus Albumin Fluid
Evaluation trial examining the use of specific forms of volume resuscitation in
critically ill patients (3) or goal-directed
therapy in patients with a broad range of
critical illness (4). Such trials in a heterogeneous group of patients may provide
improved external validity. However,
variation in response to an investigational treatment across different disease
processes may obscure a potentially beneficial treatment effect. It is, therefore,
important to consider whether the disease process might modify the effect of
the treatment being tested when choosing which category of trial to design.
Most of the larger trials of tight glycemic control in patients with critical illness fit the second category of studies
(5– 8). These trials were performed on a
broad range of critically ill patients, including patients with a variety of surgical
and medical diagnoses, and the effects of
tight glycemic control across these groups
of patients were tested. Importantly, the
benefits of tight glycemic control seen in
741
primarily cardiovascular surgical patients
(6, 9) were not replicated in two large trials
in different patient populations (5, 7). In
addition, tight glycemic control may also
increase episodes of potentially harmful hypoglycemia. Most notably, the Efficacy of
Volume Substitution and Insulin Therapy
in Severe Sepsis and the Glucontrol studies
(7, 8) documented a potential association
between hypoglycemic episodes and worsened outcomes in critically ill patients. This
tension between tight glycemic control and
avoidance of hypoglycemic episodes highlights the importance of selecting the
proper patient population in which to test
the risk– benefit ratio of tight glycemic
control. The largest trial of tight glycemic
control, the Normoglycaemia in Intensive
Care Evaluation and Survival Using Glucose Algorithm Regulation Study
(NCT00220987), which continues to enroll
patients, also tests the effects of glucose
control in a heterogeneous group of critically ill patients.
The study by Savoli et al (10) in this
issue of Critical Care Medicine fits the
first category of studies, testing tight glycemic control in patients with sepsis. In
their prospective, multicentered study of
90 septic patients randomized to tight
glycemic control (mean glucose, 112 mg/
dL) vs. conventional glycemic control
(mean glucose, 159 mg/dL), blood plasminogen activating factor inhibitor-1
(PAI-1) activity, PAI-1 concentration, and
tissue plasminogen activator levels were
noted to decrease more rapidly over the
course of 28 days in the tight glycemic
control group. The treatment group also
had lower Sequential Organ Failure Assessment scores over the same time period.
Most of the differences between groups
were found after 10 –15 days of therapy,
when many patients had either left the intensive care unit or died. Fibrinolysis, as
measured by PAI-1 activity and concentration, was inhibited in only a subset of their
septic patients and this inhibition was associated with higher baseline Sequential
Organ Failure Assessment scores and mortality rates (10). Of note, with the exception
of interleukin-6, no significant difference in
the levels of inflammatory mediators was
found between the treatment groups.
The findings of Savoli et al (10) are consistent with previous studies showing that
742
insulin dosing may limit levels of PAI-1 in
healthy adults. Further, insulin-regulated
normoglycemia has been found to prevent
immune dysfunction and inappropriate inflammation, endothelial function, and coagulation (11). These data, coupled with
the fact that the septic state contributes to
microvascular thrombosis, tissue-factormediated thrombin activation, impaired fibrinolysis, and increased PAI-1 levels (12),
make it reasonable to propose that a potential mechanism behind the beneficial effect
of tight glycemic control via intensive insulin therapy may be due to these coagulation modulatory effects. Of note, neither
the findings of Savoli et al nor a recent
study by Langouche et al (13) demonstrate
differences in inflammatory markers between patients treated with tight glycemic
control and conventional glycemic control.
Although the study by Savoli et al
provides a biologically plausible rationale for improved outcomes in septic
patients with tight glycemic control, it
does not provide enough evidence to
change practice without having the results replicated in a larger study. The
lack of information on length of exposure to usual glycemic control before
enrollment and the unclear mechanism
that might lead to delayed inhibition of
fibrinolysis in the control group limit
the generalizability of the results.
As with the trial by Savoli et al, the
selection of a more homogeneous patient
population may permit the use of smaller
sample sizes to demonstrate a treatment
effect. In addition to large multicenter trials such as NICE–SUGAR, there room for
additional appropriately designed smaller
trials to test biologically plausible treatments in well-defined patient populations
with specific syndromes or diseases.
B. Robert Gibson, MD
Department of Surgery
Johns Hopkins Medical
Institutions
Baltimore, MD
Jonathan E. Sevransky,
MD, MHS
Division of Pulmonary
and Critical Care
Medicine
Johns Hopkins Medical
Institutions
Baltimore, MD
REFERENCES
1. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute
lung injury and the acute respiratory distress
syndrome. The acute respiratory distress
syndrome network. N Engl J Med 2000; 342:
1301–1308
2. Sprung CL, Annane D, Keh D, et al: Hydrocortisone therapy for patients with septic
shock. N Engl J Med 2008; 358:111–124
3. Finfer S, Bellomo R, Boyce N, et al: A comparison of albumin and saline for fluid resuscitation in the intensive care unit. N Engl
J Med 2004; 350:2247–2256
4. Gattinoni L, Brazzi L, Pelosi P, et al: A trial of
goal-oriented hemodynamic therapy in critically ill patients. N Engl J Med 1995; 333:
1025–1032
5. Van den Berghe G, Wilmer A, Hermans G,
et al: Intensive insulin therapy in the
medical ICU. N Engl J Med 2006; 354:
449 – 461
6. van den Berghe G, Wouters P, Weekers F, et
al: Intensive insulin therapy in the critically
ill patients. N Engl J Med 2001; 345:
1359 –1367
7. Brunkhorst FM, Engel C, Bloos F, et al: Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med
2008; 358:125–139
8. Devos P, Preiser J, Melot C, et al: Impact of
tight glucose control by intensive insulin
therapy on ICU mortality and the rate of
hypoglycaemia: Final results of the GluControl study. Intensive Care Med 2007;
33:S189
9. Furnary AP, Zerr KJ, Grunkemeier GL, et
al: Continuous intravenous insulin infusion reduces the incidence of deep sternal
wound infection in diabetic patients after
cardiac surgical procedures. Ann Thorac
Surg 1999; 67:352–360; discussion
360 –362
10. Savoli M, Cugno M, Polli F, et al: Tight glycemic control may favor fibrinolysis in patients with sepsis. Crit Care Med 2009; 37:
424 – 431
11. Stegenga ME, van der Crabben SN, Levi M, et
al: Hyperglycemia stimulates coagulation,
whereas hyperinsulinemia impairs fibrinolysis in healthy humans. Diabetes 2006; 55:
1807–1812
12. Levi M, Ten Cate H: Disseminated intravascular coagulation. N Engl J Med 1999; 341:
586 –592
13. Langouche L, Meersseman W, Vander Perre
S, et al: Effect of insulin therapy on coagulation and fibrinolysis in medical intensive
care patients. Crit Care Med 2008; 36:
1475–1480
Crit Care Med 2009 Vol. 37, No. 2
Myocardial depression/injury in sepsis: Two sides of the
same coin?*
I
n the modern era, the concept of
reversible myocardial depression
or dysfunction was described by
Wiggers (1). He postulated the existence of a myocardial depressing factor
responsible for myocardial dysfunction in
hemorrhagic shock. During the 1960s
and 1970s, experimental studies showed
evidence of transient myocardial dysfunction in several forms of critical disease,
including hemorrhagic and septic shock
(2, 3).
Sequential studies have shown that
patients in septic shock adequately resuscitated typically displayed a high output
and low-systemic resistance hemodynamic circulatory condition, with myocardial depression despite the high output (4 –7). The presence of a normal or
even high ejection fraction does not exclude myocardial derangements. In those
patients who died, this hemodynamic
pattern persisted until death.
The initial phase of understanding
and the study of cardiovascular manifestations in sepsis and septic shock
began with the development of radionuclide cineangiography techniques (radioisotopic ventriculography) and with the
application of volumetric echocardiography
in managing the critically ill patients.
Discussions on the true involvement
of the heart in sepsis and septic shock,
regardless of hemodynamic conditions,
date back to the early 1960s (8) when
some studies already used endotoxic
shock models in animals. In the 1980s,
using nuclear medicine techniques,
Parker et al (6) demonstrated the decreased biventricular ejection fraction
in these septic patients. The connection
between clinical myocardial depression
*See also p. 441.
KEY WORDS: myocardial depression; myocardial injury; troponin; sepsis; echocardiography; activated
protein C
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194de3e
Crit Care Med 2009 Vol. 37, No. 2
and the effects of myocardial depressor
substances was described by Parillo et
al (9), in the late 1980s, by measuring
the serum levels of the substances in
these patients during the septic phase.
This study established a strong tie between in vivo and in vitro observations
of cardiac function and the activity of
myocardial depressor substances in septic shock.
In 1994, we and others (10) published
the histopathologic findings of the myocardium in 71 autopsies of patients who
met morphologic criteria of sepsis, comparing them with a control group and
observing the presence of interstitial
myocarditis in 27% of the sample, bacterial colonization in 11%, necrosis of cardiac fibers in 7%, and interstitial edema
in 28%, although this last finding did not
demonstrate a significant difference relative to controls. Furthermore, the identification of troponin in 1999 as a reliable
marker of myocardial injury in sepsis
added some new insights to this complex
disease (11).
In this issue of Critical Care Medicine,
Bouhemad et al (12) were auspicious in
bringing some light to this matter by
identifying two groups of troponinpositive septic patients whose clinical and
echocardiographic features were markedly different.
Echocardiographic abnormalities
showing increased left-ventricular dimensions and reduced ejection fraction
were detected in those patients with severe but reversible myocardial dysfunction, whereas those who were not able to
dilate, probably because of greater infiltrate of polymorphonuclear cells within
the myocardial fibers rendering the myocardium less compliant, were more prone
to ultimate death. It is now clear that we
cannot manage every septic patient the
very same way. As pointed out by the
authors, the concept of preload recruitment applies exclusively to the subgroup
with systolic left ventricular impairment
where ventricular enlargement may represent an adaptive mechanism to main-
tain cardiac output, whereas those presenting with ventricular relaxation
impairment despite showing no ventricular dilation at all and no drop in ejection
fraction exhibit worse prognosis.
More recently, John et al (13) were
able to show better outcomes in troponin-positive patients submitted to activated protein C administration.
Like cardiac patients, we can possibly
now stratify the most severe septic patients by determining troponin levels and
echocardiographic ventricular dimensions and, therefore, select the most suitable therapeutic choices and probably
achieve better outcomes in these critically ill septic patients. Further studies
should address these questions to clarify
the most controversial issues related to
this two-sided coin disease.
Constantino Fernandes, MD
Hospital Israelita Albert
Einstein—Critical Care
Department, Avenida Albert
Einstein
Sao Paulo, Brazil
REFERENCES
1. Wiggers CJ: Myocardial depression in shock. A
survey of cardiodynamic studies. Am Heart J
1947; 33:633– 650
2. Weil MH, MacLean LD, Visscher MD, et al:
Studies on circulatory changes in the dog
produced by endotoxin from gram-negative
microorganisms. J Clin Invest 1956; 35:
1191–1198
3. Solis RT, Downing SE: Effects of E. coli endotoxemia on ventricular performance. Am J
Physiol 1966; 211:307–313
4. Vincent JL, De Backer D: Inotrope/vasopressor support in sepsis-induced organ hypoperfusion. Semin Respir Crit Care Med 2001;
22:61–74
5. Ellrodt AG, Riedinger MS, Kimchi A, et al: Left
ventricular performance in septic shock: Reversible segmental and global abnormalities.
Am Heart J 1985; 110:402– 409
6. Parker MM, Shelhamer JH, Bacharach SL, et
al: Profound but reversible myocardial depression in patients with septic shock. Ann
Intern Med 1984; 100:483– 490
743
7. Reilly JM, Cunnion RE, Burch-Whitman C, et
al: A circulating myocardial depressant substance
is associated with cardiac dysfunction and peripheral hypoperfusion (lactic acidemia) in patients
with septic shock. Chest 1989; 95:1072–1080
8. Lefer AM, Martin J: Origin of myocardial
depressant factor in shock. Am J Physiol
1970; 218:1423–1427
9. Parillo JE, Burch C, Shelhamer JH, et al: A
circulating myocardial depressant substance
in humans with septic shock. Septic shock
patients with a reduced ejection fraction
have a circulating factor that depresses in
vitro myocardial cell performance. J Clin
Invest 1985; 76:1539 –1553
10. Fernandes CJ Jr, Iervolino M, Neves RA, et al:
Interstitial myocarditis in sepsis. Am J Cardiol 1994; 74:958
11. Fernandes CJ Jr, Akamine N, Knobel E: Cardiac troponin: A new serum marker of myo-
cardial injury in sepsis. Intensive Care Med
1999; 25:1165–1168
12. Bouhemad B, Nicolas-Robin A, Arbelot C, et
al: Acute left ventricular dilatation and
shock-induced myocardial dysfunction. Crit
Care Med 2009; 37:441– 447
13. John J, Awab A, Norman D, et al: Activated
protein C improves survival in severe sepsis
patients with elevated troponin. Intensive
Care Med 2007; 33:2122–2128
How long does it take to demonstrate the value of an idea?*
I
n 1991, the Consensus Conference
of the American College of Chest
Physicians and the Society of Critical Care Medicine introduced the
systemic inflammatory response syndrome, defined the presence of both infection and systemic inflammatory response syndrome as sepsis, and stated
clear definitions of severe sepsis and septic shock (1). Subsequently, infection and
sepsis appeared to have similar outcomes,
unaffected by the presence or number of
inflammatory response criteria (2). A decade after the previous Consensus Conference, the scientific community of intensivists developed the idea of a staging
system for sepsis similar to that used by
oncologist to stratify septic patients on
the basis of their predisposition, infection, host response, and concomitant organ dysfunction (PIRO) (3).
Community-acquired pneumonia
(CAP) is one of the most frequent infectious diseases. CAP refers to pneumonia
in a previously healthy person who has
acquired the infection from outside the
hospital, and therefore, it is also responsible for high costs for the society. Furthermore, the mortality associated with
CAP leading to organ dysfunction in the
patients admitted to intensive care unit
(ICU) is high. Taking the PROWESS
study as an example of trial on severe
sepsis, globally, 602 of the 1690 patients
*See also p. 456.
Key Words: pneumonia; sepsis; predisposition, infection, host response, and concomitant organ dysfunction; intensive care unit; severity of illness
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194be4f
744
(35.6%) enrolled had CAP, and 160 of
them died (26.6%) (4). Therefore, CAP is
a frequent cause of severe sepsis and
death.
To describe and compare CAP patients
involved in clinical trials, investigators
need both a clear definition of CAP and a
tool to assess the severity of illness. The
first one was given by the Infectious Diseases Society of America (5), which defined CAP as an acute infection of the
pulmonary parenchyma in a patient not
hospitalized or residing in a long-term
care facility for 14 days or more before
onset of symptoms. As far as the assessment of the severity of illness is concerned, mortality rate for CAP ranges
from 5% in hospital ward patients to 14%
in ICU patients, with a mean Simplified
Acute Physiology Score II (SAPS) of 33
(6) to 43% in those with a mean SAPS II
of 46 (7). Therefore, the assessment of the
degree of the severity of illness in ICU
patients with CAP is substantial for any
patient comparison.
In this issue of Critical Care Medicine,
Rello et al (8) present a scoring system
specific for CAP based on the PIRO concept, aimed at stratifying critically ill patients in mortality risk groups. The authors compared the performance of the
new score, named CAP PIRO, with that of
two different scores: one specific, ATSrevised criteria, and the other generic,
Acute Physiology and Chronic Health
Evaluation II. The CAP PIRO included the
presence of the following variables: comorbidities (chronic obstructive pulmonary disease, immunocompromise) and
age ⬎70 years, representing the predisposition of the PIRO concept; multilobar
opacities in chest x-ray and bacteremia,
representing the insult; shock and severe
hypoxemia, representing the response;
and acute renal failure and acute respiratory distress syndrome, as surrogate of
organ dysfunction (8). The CAP PIRO,
ranging 0 – 8, allowed stratifying the patients in the following four categories of
risk: low, 0 –2 points, with 28-day death
rate of 3.6%; mild, 3 points, with 13%
28-day mortality; high, 4 points, with
43% 28-day mortality; and very high, 5– 8
points, having a 28-day mortality of
76.3%. The discriminative ability, measured by the area under the receiver operating curve, was better for CAP PIRO
than for ATS-revised criteria and Acute
Physiology and Chronic Health Evaluation II. Not only mortality but also ICU
length of stay and duration of mechanical
ventilation were significantly different in
the four levels of risk of CAP PIRO (8).
The article by Rello et al (8) has some
major strengths. First, the study was performed on data prospectively collected by
33 Spanish ICUs, with a large sample of
529 adults. The wide odds ratio reported
for incidence of shock, severe hypoxemia,
or acute respiratory distress syndrome, as
well as the wide interquartile range of
lengths of mechanical ventilation and
ICU stay suggest differences in case-mix
and/or practices between ICUs, even in
the same country, but this finding is
common to multicenter studies. Second,
the CAP PIRO allows stratifying the patients admitted to ICU with CAP according to the risk of death at 28 days; therefore, it can be useful for researchers
involved in clinical trials on this topic
(for instance, on antibiotics). Finally, it
suggests a relationship between CAP
PIRO and healthcare resource use.
The study by Rello et al (8) has some
limitations. The authors did not collect
hospital mortality data and considered
patients discharged alive from ICU within
28 days as survivors. Some of those paCrit Care Med 2009 Vol. 37, No. 2
tients may have died in the hospital after
ICU discharge, according to a general
documented post-ICU mortality of 10.8%
(9). Even more relevant, in a group of
septic patients, hospital mortality has
been reported to be 48.3%, whereas 28day and 60-day mortalities were 44.8%
and 47.8%, respectively (10). The finding
that hospital mortality is consistently
higher than 28-day mortality in such patients suggests that a measure of longterm outcome should be associated with
CAP PIRO in clinical trials.
Unfortunately, the CAP PIRO does
not allow computing a probability of
hospital mortality. Furthermore, it was
not compared with more recent prognostic models, namely SAPS 3 Admission Score (11, 12) or Acute Physiology
and Chronic Health Evaluation IV (13),
which were not available at the time of
data collection, or SAPS II customized
for severe sepsis/septic shock (14).
Another article on the application of the
PIRO concept has been published few
months ago in Intensive Care Medicine
(15). The study was performed on the SAPS
3 Admission Score database to develop a
model predicting hospital mortality of patients ICU admitted with infection. There is
some overlap with the study of Rello et al
(8), and not surprisingly, there are predictive variables common to the two studies,
even if categorized in a different way. Despite the different target populations of the
two scores based on the PIRO concept,
namely, CAP and sepsis patients, the publication of those articles (8, 15) in such a
short-time interval suggests that 5 years is
the time lag required by the scientific com-
Crit Care Med 2009 Vol. 37, No. 2
munity to demonstrate the value of an idea
like PIRO.
Maurizia Capuzzo, MD
Department of Surgical,
Anesthetic and
Radiological Sciences
Section of Anesthesiology and
Intensive Care, University
Hospital of Ferrara
Ferrara, Italy
8.
9.
REFERENCES
1. Members of the American College of Chest
Physicians/Society of Crit Care Med Consensus Conference Committee: American College of Chest Physicians/Society of Critical
Care Medicine Consensus Conference: Definitions for sepsis and organ failure and
guidelines for the use of innovative therapies
in sepsis. Crit Care Med 1992; 20:864 – 874
2. Alberti C, Brun-Buisson C, Goodman SV, et
al: Influence of systemic inflammatory response syndrome and sepsis on outcome of
critically ill infected patients. Am J Respir
Crit Care Med 2003; 168:77– 84
3. Levy MM, Fink MP, Marshall JC, et al: 2001
SCCM/ESICM/ACCP/ATS/SIS International
sepsis definitions conference. Crit Care Med
2003; 31:1250 –1256
4. Laterre PF, Garber G, Levy H, et al: Severe
community-acquired pneumonia as a cause
of severe sepsis: Data from the PROWESS
study. Crit Care Med 2005; 33:952–961
5. Mandell LA, Bartlett JG, Dowell SF, et al:
Update of practice guidelines for the management of community-acquired pneumonia in
immunocompetent adults. Clin Infect Dis
2003; 37:1405–1433
6. Leroy O, Saux P, Bédos J-P, et al: Comparison of levofloxacin and cefotaxime combined
with ofloxacin for ICU patients with community-acquired pneumonia who do not require
vasopressors. Chest 2005; 128:172–183
7. Paganin F, Lilienthal F, Bourdin A, et al:
10.
11.
12.
13.
14.
15.
Severe community-acquired pneumonia: Assessment of microbial aetiology as mortality
factor. Eur Respir J 2004; 24:779 –785
Rello J, Rodriguez A, Lisboa T, et al: PIRO
score for community-acquired pneumonia: A
new prediction rule for assessment of severity in intensive care unit patients with community-acquired pneumonia. Crit Care Med
2009; 27:456 – 462
Azoulay E, Adrie C, De Lassence A, et al:
Determinants of postintensive care unit mortality: A prospective multicenter study. Crit
Care Med 2003; 31:428 – 432
Garnacho-Monteiro J, Garcia-Garmendia JL,
Barrero-Almodovar A, et al: Impact of adequate empirical antibiotic therapy on the
outcome of patients admitted to the intensive care unit with sepsis. Crit Care Med
2003; 31:2742–2751
Metnitz PGH, Moreno RP, Almeida E, et al:
SAPS 3—From evaluation of the patient to
evaluation of the ICU. Part 1. Objectives,
methods and cohort description. Intensive
Care Med 2005; 31:1336 –1344
Moreno RP, Metnitz PGH, Almeida E, et al:
SAPS 3—From evaluation of the patient to
evaluation of the ICU. Part 2. Development of
a prognostic model for hospital mortality at
ICU admission. Intensive Care Med 2005; 31:
1345–1355
Zimmerman JE, Kramer AA, McNair DS, et
al: Acute Physiology and Chronic Health
Evaluation (APACHE) IV: Hospital mortality
assessment for today’s critically ill patients.
Crit Care Med 2006; 34:1297–1310
Le Gall JR, Lemeshow S, Leleu G, et al:
Customized probability models for early severe sepsis in adult intensive care patients.
Intensive Care Unit Scoring Group. JAMA
1995; 273:644 – 650
Moreno RP, Metnitz B, Adler L, et al: Sepsis
mortality prediction based on predisposition,
infection and response. Intensive Care Med
2008; 34:496 –504
745
Intensive insulin therapy: The swinging pendulum of evidence*
S
ince 2001, the year of publication of the first intensive insulin trial by Van den Berghe et
al (1), hyperglycemia and intensive insulin therapy (IIT) are placed
high on the list of critical care controversies. In this trial, Van den Berghe demonstrated a survival benefit for surgical
intensive care unit (ICU) patients treated
with an intensive insulin protocol to
maintain normoglycemia (80⫺110 mg/
dL; 4.6% compared with 8.0% in-hospital
mortality). Although this was a singlecenter trial according to evidence base
medicine standards is not so high, IIT
was adopted by many and even incorporated in guidelines for sepsis patients,
such as the Surviving Sepsis Campaign,
endorsed by the Society of Critical Care
Medicine, and the European Society of
Intensive Care Medicine (2, 3). Since the
original hallmark study, several observational trials confirmed the positive effects
of normoglycemia on outcome in critically
ill patients (4, 5). However, enthusiasm
weaned after the results came out of three
other prospective randomized trials on IIT
in critically ill patients. Of these, two trials
are published (6, 7), one trial is finished,
but only presented as an abstract (http://
clinicaltrials.gov/ct/gui/show/NCT00107601).
A fourth trial, the Australian–New Zealand–
Canadian “Normoglycemia in Intensive
Care Evaluation and Survival Using
Glucose Algorithm Regulation” trial, is
still recruiting (http://www.controlledtrials.com/ISRCTN04968275). Unfortunately, the results of these trials have
made things less clear. The single-center
follow-up trial by Van den Berghe in medical ICU patients rendered ambivalent results (6). There was no difference in outcome between patients treated with IIT
and conventional insulin therapy. How-
*See also p. 463.
Key Words: intensive insulin therapy; blood glucose; outcome; critically ill; hypoglycemia; hyperglycemia
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194be79
746
ever, in the subgroup of patients admitted for 3 days or more, IIT was associated
with a survival benefit (in-hospital mortality in the conventional treated patients was
53.5% vs. 43% in IIT patients). This subgroup of patients could not be identified
beforehand. An important difference with
the previous trial of this group was that
patients in the medical ICU were more severely ill. Also, there was a much higher
incidence of hypoglycemia in patients
treated with IIT in the medical ICU, compared with the surgical ICU trial (18.7% vs.
5.1%). The German multicenter Efficacy of
Volume Substitution and Insulin Therapy
in Severe Sepsis study randomized severe
sepsis patients to IIT or conventional insulin therapy and Ringer’s lactate or a starch
solution for fluid resuscitation (7). The
study was stopped for safety reasons by the
data and safety monitoring board of the
study, after an interim analysis demonstrated an unexpected high rate of hypoglycemic events (17.0%) in patients randomized to IIT. The positive effects of IIT, as
demonstrated in the first study by Van den
Berghe et al, could not be confirmed, not
even in subanalysis. In the 537 patients
eligible for analysis, there was no difference
in a series of outcomes including 28- and
90-day mortality between patients treated
with conventional insulin therapy and IIT.
The European multicenter Glucontrol
study was also prematurely stopped after
inclusion of 1101 patients because of a
higher rate of adverse events in IIT patients.
All-cause mortality was not different between patients exposed to conventional and
IIT (17% vs. 15%, p ⫽ not significant). In
summary, the beneficial effects of IIT were
only confirmed in a subanalysis of a more
sick patient cohort, by the team that invented the therapy, and could not be reproduced in two, underpowered, multicenter
trials. In addition, there are concerns regarding the side effects of IIT. There was a
high incidence of hypoglycemia both in the
experienced hands of the Leuven team and
in the less experienced hands of the centers
that participated in the two multicenter
studies. These results raise concerns regarding the external validity of IIT. In other
words does IIT work in other, less experienced units? It also illustrates that it is not
necessarily correct to translate a therapy
that works in a specific patient cohort to
another cohort.
In this issue of Critical Care Medicine,
Bagshaw and colleagues present the results from an observational study on a
very large database of 66,184 Australian
ICU patients (8). Although this is an observational study, it adds a whole new dimension to the IIT controversy. In this study,
normoglycemia was associated with better
outcome, even after correction for other
covariates for increased mortality. In other
words, blood glucose levels in the normal
range are good, as in the original study by
Van den Berghe et al. Interestingly, the
“normal” range in this study is higher than
that used in the studies on IIT mentioned
before (5.6 – 8.69 vs. 4.4 –5.6 mmol/L). This
range was determined as the second quartile of all average blood glucose concentrations, and it corresponds remarkably well
with the conservative recommendations of
the Surviving Sepsis Campaign (blood glucose ⬍150 mg/dL or 8.25 mmol/L). An important aspect in this study is that patients
were categorized on an average blood glucose concentration during a 24-hour period, and not on the morning blood glucose, as in the Leuven studies and in the
VISEP study. This may be of importance, as
it has been demonstrated that glucose control may vary considerably in IIT patients,
during a 24-hour observation period (9).
Differences in blood glucose control during
a 24-hour period, may explain why IIT
works in the hands of Van den Berghe’s
team, and why this could not be reproduced in the VISEP and Glucontrol trials.
One may assume that Van den Berghe’s
team is more experienced and dedicated,
and therefore is able to maintain a stricter
glucose control during a 24-hour period
compared with less experienced units.
Higher target levels may diminish the beneficial effects of IIT, but more adequate
blood glucose control during a 24-hour period may compensate for that. Therefore,
average blood glucose may be a more adequate marker of the efficacy of IIT, and
should be considered in future studies. In
addition, higher target blood glucose concentration will probably lower the inciCrit Care Med 2009 Vol. 37, No. 2
dence of hypoglycemia and make this therapy a more safe one, even in less
experienced hands. Another interesting
finding is the U-shaped outcome curve. Patients with an average blood glucose at the
lower and high end of the spectrum do
worse compared with patients in the normal range. Going low is not always good;
this supports the hypothesis that less aggressive IIT and blood glucose control during a 24-hour period may improve outcome, even in a multicenter setting.
Finally, this study also provides us with an
explanation for the different results in the
prospective IIT studies published to date.
The effects of normal blood glucose were
most prominent in the surgical and cardiac
surgery subgroups, comparable with the
cohort in the first Leuven trial (1). The
beneficial effects were less pronounced or
even absent in sepsis patients and medical
patients, like the patients included in the
VISEP study and the second study by the
Leuven group (6, 7). Future studies on IIT
should probably look at even more specific
cohorts, such as acute respiratory distress
syndrome patients and patients admitted
after trauma.
There are several limitations to this
trial. It is an observational trial and, therefore, prone to bias, and average blood glucose measured during the first 24 hours
was used as a surrogate for blood glucose
control during the rest of the ICU stay. The
study is, therefore, more hypothesis generating than the absolute proof of evidence.
However, the multicenter setting and the
large number of patients give us robust
data. These data may help us explain results
from other studies in this field and design
new studies on this topic.
Eric A. J. Hoste, MD, PhD
Intensive Care Unit, Ghent
University Hospital
Gent, Belgium
3.
4.
5.
6.
7.
8.
REFERENCES
1. Van den Berghe G, Wouters P, Weekers F, et al:
Intensive insulin therapy in critically ill patients.
N Engl J Med 2001; 345:1359 – 1367
2. Dellinger RP, Carlet JM, Masur H, et al: Surviving Sepsis Campaign guidelines for man-
9.
agement of severe sepsis and septic shock. Crit
Care Med 2004; 32:858 – 873
Dellinger RP, Levy MM, Carlet JM, et al: Surviving Sepsis Campaign: International guidelines
for management of severe sepsis and septic
shock: 2008. Intensive Care Med 2008; 34:17– 60
Finney SJ, Zekveld C, Elia A, et al: Glucose
control and mortality in critically ill patients.
JAMA 2003; 290:2041–2047
Krinsley JS: Effect of an intensive glucose
management protocol on the mortality of critically ill adult patients. Mayo Clin Proc 2004;
79:992–1000
Van den Berghe G, Wilmer A, Hermans G, et
al: Intensive insulin therapy in the medical
ICU. N Engl J Med 2006; 354:449 – 461
Brunkhorst FM, Engel C, Bloos F, et al: Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med 2008;
358:125–139
Bagshaw SM, Egi M, George C, et al: Early
blood glucose control and mortality in critically ill patients in Australia. Crit Care Med
2009; 37:463– 470
Oeyen SG, Hoste EA, Roosens CD, et al: Adherence to and efficacy and safety of an insulin
protocol in the critically ill: A Prospective Observational Study. Am J Crit Care 2007; 16:
599 – 608
Cardiocerebral resuscitation: Few answers, more questions*
A
major concern since the advent of the modern age of cardiac resuscitation is that we
might be resuscitating patients
but leaving a large group of patients with
prolonged neurologic disability. Cobbe et
al (1) found that about 40% of initial
survivors of out-of-hospital cardiac arrest
could be discharged home without major
neurologic disability. Adrie et al (2) found
that while early death from refractory
shock occurred in 42 patients from a
group of 130 patients achieving restoration of spontaneous circulation (ROSC)
for ⬎1 hour, 60 of the remaining 88
patients died of complications related to
neurologic failure later in the hospital
course. Concern that survivors from car-
*See also p. 471.
Key Words: cardiac arrest; heart failure; cerebral
oxygenation; ventricular fibrillation; cardiocerebral
resuscitation
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194c426
Crit Care Med 2009 Vol. 37, No. 2
diac arrest could become a long-term
burden on the society was ameliorated by
studies like these showing that there was
a high rate of mortality in severely neurologically impaired patients in the period immediately after resuscitation. Cole
and Corday (3) found that only two patients (7% of the total) requiring more
than 4 minutes of advance cardiac life
support could be resuscitated and discharged alive, both with permanent brain
damage. In a study of resuscitated inhospital intensive care unit and nonintensive care unit victims of cardiac arrest,
Bell and Hodgson (4) found that although
30% and 45% of patients, respectively,
were comatose after resuscitation, most
of these patients died. Only 3% and 4.5%
of patients from these groups were discharged alive with brain damage.
No-reflow, the concept of achieving
ROSC, but subsequently suffering a fall in
cerebral blood flow, which, in turn, contributed to poor cerebral outcome, and
ultimately, the demise of the patient was
postulated by Negovsky (5) and shown to
exist in animal and clinical models. Safar
(6), challenging the prevailing emphasis on
cardiac resuscitation, noted that too many
victims achieve ROSC as demonstrated in
the studies earlier, only to die of cerebral
failure, emphasizing the need for a new
type of cardiopulmonary resuscitation
(CPR), cardiocerebral CPR, or CPCR.
Mullner et al (7) demonstrated that prearrest variables and postarrest neurologic
function influenced outcome. Adverse outcome increased with increasing age and in
patients with increasing numbers of prearrest diseases, especially congestive heart
failure and diabetes mellitus. Again, longterm survival with profound neurologic impairment was not found to be a problem
with 106 of the 121 patients (29% of the
survivors) with unfavorable neurologic recovery dead within 6 months of the initial
cardiac arrest. Age and an ejection fraction
⬍35% were also shown to be associated
with unfavorable outcomes by Bunch et al
(8) in a study of ischemic vs. nonischemic
heart disease-associated cardiac arrests.
The influence of left ventricular function
was confirmed in a study of in-hospital cardiac arrest. Gonzalez et al (9) found a 19%
survival to discharge in patients with a nor747
mal prearrest ejection fraction vs. 8% in
those with moderate to severe left ventricular dysfunction.
In this issue of Critical Care Medicine,
Skhirtladze et al (10) help explain the
influence of heart failure on neurologic
outcome from cardiac arrest in their fascinating model of reversible humancardiac arrest by studying patients having
a cardiodefibrillator tested during implantation. Although physiologic studies
have been conducted on victims of cardiac arrest, most have been on patients
with long downtimes before instrumentation. In this study, ventricular fibrillation is induced to test the cardiodefibrillator, offering the opportunity to
measure cerebral oxygenation before and
after ROSC. In this elegant study, the
authors showed that despite similar blood
pressure, heart rate, and pulse oximetry
values, patients with severe heart failure
were more likely to have clinically important cerebral desaturation detected by
near infrared spectroscopy. This was ev
ident at baseline and after brief periods of
cardiac arrest. This might explain why
patients with heart failure suffering cardiac arrest do so poorly. Cohan et al (11)
found seemingly paradoxical results in a
xenon study of cerebral blood flow in humans after cardiac arrest. In this study,
comatose patients who regained consciousness had relatively normal cerebral blood
flow before regaining consciousness,
whereas those who died without awakening
developed hyperemic cerebral blood flow
after ROSC. Inoue et al (12) also found
early hyperemia to be associated with a
poor prognosis after cardiac arrest. Conversely, Mullner et al (13) found higher
cerebral oxygen extraction to be associated
with better cerebral recovery. Different
models and different techniques do not allow us to directly compare the studies, but
the results emphasize that we need to understand more about postresuscitation cerebral blood flow and its relationship to
underlying brain ischemia before we can
know the correct approach to CPCR in the
many different situations that present to
us. It does not mean that a hyperemic flush
is not helpful after attainment of ROSC, but
emphasizes that we really do not know the
“formula” for postresuscitation care. Prearrest variables, such as heart failure, vascu-
748
lar disease, diabetes, and intra-arrest variables, such as duration of no-flow and lowflow may all need to be accounted for to
determine the appropriate approach to postROSC care for the individual patient. Optimal
cerebral resuscitation will probably turn
out to be similar to defibrillation— one approach will not fit all patients. Just as we
may need to do chest compression before
defibrillation in patients with prolonged
no-flow times, we may need to tailor our
approach differently for patients with premorbid conditions, long no-flow, or lowflow times.
Perhaps to achieve better outcomes, we
should monitor efficacy of artificial circulation with widely available, easily applied
tools, such as end-tidal CO2 (14) while maximizing cardiac output with techniques,
such as continuous chest compressionCPR. Kellum et al (15) showed that with
continuous chest compression-CPR, survival increased from 19% to 48%, compared with a control group derived from
the 3 years preceding the test period. In
addition, neurologically intact survival increased from 77% in the first period vs.
84% in the experimental period.
Unfortunately, after ⬎50 years of
CPR and advanced cardiac life support, the
majority of patients still either cannot
achieve ROSC or die of postresuscitation
disease. Basic physiologic studies in animals and humans, such as the study by
Skhirtladze et al in this issue of Critical
Care Medicine help answer small parts of
the question. In turn, these clues need to be
taken back to bench models and cell-based
studies to develop better techniques for the
key variables involved in resuscitation from
cardiac arrest: generation of cardiac output during the arrest phase, generation of
an organized rhythm, and finally functional cardiac and cerebral recovery.
Robert L. Levine, MD, FACEP, FCCM
Department of Emergency
Medicine
University of Texas School of
Medicine at Houston
The Department of Medicine
The Methodist Hospital
Houston, TX
REFERENCES
1. Cobbe SM, Dalziel K, Ford I, et al: Survival of
1476 patients initially resuscitated from out
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
of hospital cardiac arrest. BMJ 1996; 312:
1633–1637
Adrie C, Cariou A, Mourvillier B, et al: Predicting survival with good neurological recovery at hospital admission after successful
resuscitation of out-of-hospital cardiac arrest: The OHCA score. Eur Heart J 2006;
27:2840 –2845
Cole SL, Corday E: Four-minute limit for
cardiac resuscitation. JAMA 1956; 161:
1454 –1458
Bell JA, Hodgson HJF: Coma after cardiac
arrest. Brain 1974; 97:361–372
Negovsky VA: The second step in resuscitation- the treatment of the “post-resuscitation
disease.” Resuscitation 1972; 1:1–7
Safar P: Resuscitation medicine research:
Quo Vadis. Ann Emerg Med 1996; 27:
542–552
Mullner M, Sterz F, Behringer W, et al: The
influence of chronic prearrest health conditions on mortality and functional neurological recovery in cardiac arrest survivors. Am J
Med 1998; 104:369 –373
Bunch TJ, Kottke TE, Lopez-Jimenez F, et al:
A comparative analysis of short- and longterm outcomes after ventricular fibrillation
out-of-hospital cardiac arrest in patients with
ischemic and nonischemic heart disease.
Am J Cardiol 2006; 98:857– 860
Gonzalez MM, Berg RA, Madkarni VM, et al:
Left ventricular systolic function and outcome
after in-hospital cardiac arrest. Circulation
2008; 117:1864 –1872
Skhirtladze K, Birkenberg B, Mora B, et al:
Cerebral desaturation during cardiac arrest:
Its relation to arrest duration and left ventricular pump function. Crit Care Med 2009:
37:471– 475
Cohan SL, Mun SK, Petite J, et al: Cerebral
blood flow in humans following resuscitation from cardiac arrest. Stroke 1989; 20:
761–765
Inoue Y, Shiozaki T, Irisawa T, et al: Acute
cerebral blood flow variations after human
cardiac arrest assessed by stable xenon enhanced computed tomography. Curr Nuerovasc Res 2007; 4:49 –54
Mullner M, Sterz F, Domanovits H, et al:
Systemic and cerebral oxygen extraction after human cardiac arrest. Eur j Emerg Med
1996; 3:19 –24
Levine RL, Wayne MA, Miller CC: End-tidal
carbon dioxide and outcome of out-ofhospital cardiac arrest. N Eng J Med 1997;
337:301–306
Kellum MJ, Kennedy KW, Ewy GA: Cardiocerebral resuscitation improves survival of
patients with out-of-hospital cardiac arrest.
Am J Med 2006; 119:335–340
Crit Care Med 2009 Vol. 37, No. 2
Vasopressin and its copilot copeptin in sepsis and septic shock*
V
asopressin levels increase
early in septic shock because
hypotension is the most potent stimulus of increased synthesis and release of vasopressin. Indeed,
if an animal is challenged with conflicting signals to vasopressin release (such as
hypotension and hyponatremia), the animal will increase vasopressin because hypotension is a more potent stimulus than
hyponatremia is an inhibitor of vasopressin release. After the initial increase of
vasopressin levels in septic shock, vasopressin levels then decline rapidly to levels that are inappropriately low (compared with hypotensive patients who have
cardiogenic shock) (1–3). Thus, few endocrine systems are so rapidly activated (to
increase serum levels) and then are so rapidly exhausted (such that serum levels decrease) as the vasopressin axis in sepsis.
Serum levels of vasopressin—a nonapeptide—represent the interactions of
the synthesis, release, and metabolism of
vasopressin. Synthesis of preprovasopressin occurs in various nuclei of the hypothalamus. Subsequently, there is conversion to provasopressin followed by
conversion of provasopressin by subtilisin-like proprotein convertase (SPC3) to
vasopressin (4). Vasopressin is metabolized by insulin-regulated aminopeptidase also known as vasopressinase (5).
Vasopressin and copeptin levels are altered in sepsis and septic shock. In this
issue of Critical Care Medicine, Jochberger et al (6) report an observational
cohort study designed to compare serum
levels of vasopressin and copeptin of patients who had infection (n ⫽ 10), severe
sepsis (n ⫽ 22), and septic shock (n ⫽
28). Measurements of serum levels of vasopressin and copeptin were made for the
first 7 days of admission. Patients with
severe sepsis and septic shock had higher
*See also p. 476.
Key Words: copeptin; vasopressin levels; sepsis;
septic shock; cardiac surgery; survival
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194d117
Crit Care Med 2009 Vol. 37, No. 2
vasopressin (and copeptin) levels than patients who had infection. However, there
was no difference in serum vasopressin
levels between patients who had severe
sepsis vs. septic shock. This is novel information that requires thoughtful interpretation. Should the levels of vasopressin
have been higher in septic shock (than severe sepsis) because of the additional stimulus of hypotension in patients who had
septic shock? Alternatively, does this study
teach us that serum levels of vasopressin
(and copeptin) of patients who have severe
sepsis and septic shock are similar and so
those patients should be treated similarly?
Copeptin is a 39-amino acid glycopeptide that is the C terminal part of provasopressin, very similar to the C peptide of
insulin (7). Jochberger et al (6) found
that plasma vasopressin levels correlated
significantly with copeptin levels. Is the
correlation of serum levels of copeptin
and vasopressin tight enough so that
copeptin could be measured as a surrogate for vasopressin? Alternatively, does
this study teach us that serum levels of
vasopressin must be measured in patients
who have severe sepsis and septic shock
(because renal dysfunction was common
and disturbed the tight correlation of serum vasopressin and copeptin levels)?
This study extends studies of serum levels of vasopressin and copeptin. Lin et al (8)
studied patients in the emergency department and found lower vasopressin levels in
patients who went on to septic shock (3.6
pg/mL) compared with patients who had
sepsis (10.6 pg/mL) and severe sepsis (21.8
pg/mL) (Table 1). Struck et al (9) described
the rationale and methods for measurement of copeptin. Dunser et al (10) published a case report showing increased levels of copeptin and vasopressin early (first
36 hours) in septic shock, which decreased
during recovery. Jochberger et al (11)
found, as in other studies, critically ill patients had higher copeptin levels than controls. Patients who had cardiac surgery had
higher copeptin levels than patients who
had sepsis or controls. Vasopressin and
copeptin levels were highly correlated. Interestingly, the ratio of copeptin to vasopressin was higher in sepsis than after cardiac surgery suggesting altered estimation
of vasopressin by copeptin in patients who
have sepsis. In another study, Jochberger et
al (12) confirmed that critically ill patients
had higher vasopressin levels (11.9 pg/mL)
than controls (0.9 pg/mL). Patients who
had hemodynamic dysfunction had higher
vasopressin levels than patients without hemodynamic dysfunction (14.1 vs. 8.7 pg/
mL). As in previous studies, patients who
had cardiac surgery had higher vasopressin
(1) and copeptin (11, 12) levels than septic
patients. Morgentaler et al (13) reported a
sandwich assay with two antibodies for
copeptin and found higher levels in critically ill patients than healthy controls (79.5
vs. 4.2 pmol/L, respectively) with good correlation between vasopressin and copeptin
levels. Morgentaler et al (7) showed that
hemorrhagic shock in baboons quickly lead
to markedly increased serum copeptin levels (from 7.5 to 269 pM), and resuscitation
was associated with declining copeptin (to
27 pM) indicating that copeptin levels are
modulated profoundly and rapidly in hemorrhagic shock. Morgentaler et al (7) also
evaluated critically ill patients and found
increased copeptin levels in sepsis: (healthy
controls 4 pM; critically ill without sepsis
27 pM; sepsis 50 pM; severe sepsis 74 pM;
and septic shock 171 pM). Muller et al (14)
assessed copeptin levels in 545 patients
who had lower respiratory tract infection
vs. 50 healthy controls. Copeptin levels
were higher in patients than healthy controls and increased as severity of pneumonia increased. Both Morgentaler et al (7,
13) and Muller et al found higher levels of
copeptin in nonsurvivors compared with
survivors (Morgentaler et al [7], 171 vs. 87
pM; Muller et al [14], 70 vs. 24 pM).
Russell et al (3) found that vasopressin
levels were extremely low in severe septic
shock (median 3.2 pmol/L) and increased
during low-dose (0.03 U/min) vasopressin
infusion to about 74 pM (6 hours) and 98
pM (24 hours).
Lodha et al (15) found that serum vasopressin levels in pediatric septic shock are
similar to the results of Jochberger et al (6).
Vasopressin levels were increased in septic
shock (116 pg/mL) and severe sepsis (106
pg/mL), and vasopressin levels did not
change over 96 hours of evaluation. As in
Morgentaler et al (7) and Muller et al (14),
749
Table 1. Vasopressin and copeptin levels in humans. Studies of septic shock are in bold for
comparison
5.
Study (Year, Reference)
Landry 1997 (1)
Lin 2005 (8)
Jochberger 2006 (12)
Morgentaler 2006 (13)
Morgentaler 2007 (7)
Lodha 2006 (15)
Jochberger 2007 (17)
Russell 2008 (3)
Jochberger 2008 (6)
Condition
Vasopressin Levels Copeptin Levels
Septic shock
Cardiogenic shock
Septic shock
Sepsis
Severe sepsis
Sepsis
Sepsis
Critically ill (no sepsis)
Sepsis
Severe sepsis
Septic shock
Pediatric septic shock
Pediatric severe sepsis
Shock after cardiac surgery
Multiple trauma
Septic shock
Septic shock plus vasopressin infusion
(0.03 U/min) at 6 hours
Septic shock plus vasopressin infusion
(0.03 U/min) at 24 hours
Infection
Severe sepsis
Septic shock
3.1 pg/mL
22.7 pg/mL
3.6 pg/mL
10.6 pg/mL
21.8 pg/mL
11.9 pg/mL
6.
79.5 pM
27 pM
50 pM
74 pM
171 pM
116 pg/mL
106 pg/mL
19.7 pM
44.3 pM
3.2 pM
74 pM
7.
8.
9.
98 pM
10.
3.2 pMa
6.5 pMa
6.4 pMa
25 pMa
60 pMa
70 pMa
11.
a
Estimated from Figures and text of Jochberger 2008 (6).
nonsurvivors had higher vasopressin levels
than survivors (118 vs. 76 pg/mL).
Can copeptin levels be used as a surrogate
for vasopressin levels? I suggest that copeptin
levels cannot be used as a surrogate for vasopressin levels without further evaluation because of several aspects presented by Jochberger et al and other studies. Important
considerations are that the sample size was
relatively small, these are single-center studies, and the correlation between vasopressin
and copeptin levels overall was adequate, but
was not adequate if there was continuous
veno-venous hemofiltration or renal function
(statistical correlation with creatinine was
p ⫽ 0.05 and with creatinine clearance was
p ⫽ 0.06). Thus, I suggest that copeptin and
vasopressin levels did not correlate well
enough for individual patient evaluation.
To make matters worse in patients who
have septic shock, there is downregulation
of vasopressin receptors in addition to the
deficiency of vasopressin levels in sepsis
750
(16) and that further exacerbates the vasopressin deficiency of sepsis.
James A. Russell, MD
James Hogg iCAPTURE Centre for
Cardiovascular and Pulmonary
Research
St. Paul’s Hospital
Vancouver, BC, Canada
12.
REFERENCES
15.
1. Landry DW, Levin HR, Gallant EM, et al: Vasopressin deficiency contributes to the vasodilation
of septic shock. Circulation 1997; 95:1122–1125
2. Patel B, Holmes C, Russell JA, et al: Beneficial effects of short-term vasopressin infusion during severe septic shock. Anesthesiology 2002; 96:576 –582
3. Russell JA, Walley KR, Singer J, et al: Vasopressin versus norepinephrine infusion in patients
with septic shock. N Engl J Med 2008; 358:
877– 887
4. Coates L, Birch N: Differential cleavage of
13.
14.
16.
17.
provasopressin by the major molecular forms
of SPC3. J Neurochem 1998; 70:1670 –1678
Wallis MG, Lankford MF, Keller SR: Vasopressin is a physiologic substrate for the insulinregulated aminopeptidase IRAP. Am J Physiol
Endocrinol Metab 2007; 293:E1092–E1102
Jochberger S, Dörler J, Luckner G, et al: The
vasopressin and copeptin response to infection, severe sepsis, and septic shock. Crit
Care Med 2009; 37:476 – 482
Morgentaler NG, Muller B, Struck J, et al:
Copeptin, a stable peptide of the arginine
vasopressin precursor, is elevated in hemorrhagic and septic shock. Shock 2007; 28:
219 –226
Lin IY, Ma HP, Lin AC, et al: Low vasopressin/norepinephrine ratio predicts septic
shock. Am J Emerg Med 2005; 23:718 –724
Struck J, Morgentaler NG, Bergmann A:
Copeptin, a stable peptide derived from the
vasopressin precursor, is elevated in serum of
sepsis patients. Peptides 2005; 26:2500 –2504
Dunser M, Mayr VD, Wenzel V, et al: Course of
vasopressin and copeptin plasma concentrations
in a patient with severe septic shock. Anaesth
Intensive Care 2006; 34:498–500
Jochberger J, Morgentaler NG, Mayr VD, et
al: Copeptin and arginine vasopressin concentrations in critically ill patients. J Clin
Endocrinol Metab 2006; 91:4381– 4386
Jochberger S, Mayr V, Luckner G, et al: Serum
vasopressin concentrations in critically ill patients. Crit Care Med 2006; 34:293–299
Morgentaler S, Struck J, Alonso C, et al: Assay
for the measurement of copeptin, a stable peptide derived from the precursor of vasopressin.
Clin Chem 2006; 52:112–119
Muller B, Morgentaler NG, Stolz P, et al:
Circulating levels of copeptin, a novel biomarker, in lower respiratory tract infections. Eur J Clin Invest 2007; 37:145–152
Lodha R, Vivekanandhan S, Sarthi M, et al:
Serial circulating vasopressin levels in children with septic shock. Pediatr Crit Care
Med 2006; 7:220 –224
Schmidt C, Hocherl K, Kurt B, et al: Role of
nuclear factor-kappaB-dependent induction
of cytokines in the regulation of vasopressin
V1A-receptors during cecal ligation and
puncture-induced circulatory failure. Crit
Care Med 2008; 36:2363–2372
Jochberger J, Mayr VD, Luckner G, et al: Vasopressin plasma concentrations in post-cardiotomy shock: A prospective, controlled trial. Intensive Care Med 2007; 33(Suppl 2): A0763
Crit Care Med 2009 Vol. 37, No. 2
Routine nursing procedures—Take care of the patient
and the splanchnic circulation!*
C
are of the critically ill not only
comprises invasive monitoring, complex pharmacologic
intervention, and eventually
organ replacement, but also requires
careful nursing of the patient. Routine
nursing procedures consist of mechanical
ventilation–associated procedures, e.g.,
endotracheal suctioning; hygiene measures, e.g., oral care and washing; diagnostics, e.g., chest radiograph and physical examinations; removal/insertion of
catheters; and physical treatment, e.g.,
physiotherapy and patient turning. It has
been well established for decades that
many of these routine nursing procedures may affect patient homoeostasis
(1– 4), in particular, when associated with
endotracheal suctioning in mechanically
ventilated patients (5–7). In fact, nearly
five decades ago, the group of Landmesser already reported a marked fall in
arterial oxygen saturation during and after endotracheal suctioning, which could
only be partly prevented by preoxygenation with an FIO2 of 1.0 (8). Increased
oxygen consumption and CO2 production
resulting from enhanced sympathetic
tone (9 –11) clearly assumes major importance in this context as a result of
increased respiratory muscle activity because of agitation or coughing episodes.
In fact, sedation and analgesia with shortacting narcotics markedly attenuated the
hemodynamic response to routine nursing procedures (11, 12). Despite this obvious and potentially important role of
nursing procedures for patient stability,
clinical data dealing with this phenomenon are scarce, and up to, now no study is
available on the effects of such procedures on hepatosplanchnic perfusion, a
*See also p. 483.
Key Words: endotracheal suctioning; chest physiotherapy; sympathetic tone; hepatic venous oxygen
saturation
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194d157
Crit Care Med 2009 Vol. 37, No. 2
region which has been identified as a
high-risk area in the pathogenesis of
multiple organ failure, in particular, in
the presence of a mismatch between metabolic demands and oxygen supply (13).
In the current issue of Critical Care
Medicine, Jakob et al (14) present a study
exploring the role of routine nursing procedures for decreases of hepatic venous
oxygen saturation, a well-accepted
marker of splanchnic perfusion and metabolism, and the relationship of these
desaturation episodes and outcome. In 36
patients with acute respiratory or circulatory failure, the majority suffering from
septic shock (59%), mixed and hepatic
venous oxygen saturations were continuously recorded during various nursing
procedures that were part of the routine
care of the patients during their intensive
care unit stay. The main finding was that
patients are repeatedly exposed to episodes of hepatic venous desaturation—
and thereby possibly to reduced splanchnic perfusion— during routine nursing
procedures, and that these episodes were
mostly mirrored, in part, only by changes
in peripheral arterial or mixed venous
oxygen saturation. In addition, the total
number of desaturation episodes, regardless of their association with procedures,
were directly related to the maximal Sequential Organ Failure Assessment score,
but not with length of stay or mortality.
What do we learn from this study? Jakob
et al confirm the well-established fact
that regional, i.e., hepatic, venous oxygen
saturation cannot necessarily be derived
from the arterial or mixed venous one, in
particular in the presence of regional oxygen supply demand dependency (13).
Furthermore, most desaturation episodes
were due to ventilator-related procedures, and endotracheal suctioning
caused the most pronounced fall in hepatic venous oxygen saturation and increase in the hepatic-mixed venous oxygen saturation gradient. As mentioned
eariler, endotracheal suctioning is well
known to impair pulmonary gas exchange, and given the high proportion of
patients with septic shock, it is not surprising that endotracheal suctioning
caused the largest decrease in hepatic venous oxygen saturation: these patients
often present with a high hepatosplanchnic oxygen extraction, and even a small
change in oxygen demand and supply
may cause large decreases in regional
venous saturation (13). Unfortunately,
Jakob et al did not mention whether
they used measures to counteract any
endotracheal suctioning–induced impairment of pulmonary gas exchange,
i.e., application of hyperoxia (6, 8) or
alveolar recruitment maneuvers (15),
or whether they performed open or
closed suction (16, 17). The latter was
demonstrated to attenuate the suctioninduced fall in arterial oxygen saturation, albeit its benefit for morbidity remains a matter of debate (18).
A very interesting finding of the study
by Jakob et al is that more than one
quarter (27%) of all observed hepatic venous desaturation episodes occurred independently of any nursing procedure.
Although only marginally discussed by
the authors, this finding may nevertheless highlight the importance of spontaneous physiologic fluctuations such as
heart rate and blood pressure variability
(19). As a consequence, regional oxygen
saturations are also likely to show spontaneous variations, and this phenomenon
may have contributed to the authors’ findings. It should be noted, however, that the
degree of heart rate variability, the currently best studied parameter of spontaneous physiologic variation, was inversely related to outcome in several subpopulations
of intensive care unit patients (20 –22). By
contrast, albeit Jakob et al did not analyze
the relation between spontaneous drops of
hepatic venous oxygen saturation and the
Sequential Organ Failure Assessment
score, the authors report that the total—
and not only of procedure associated—
number of regional venous desaturation
episodes was directly related to the severity
of the disease. As a logic consequence, they
suggest that hepatic venous desaturation
751
episodes, hence, mirror the patients’ compromised capacity if not inability to increase blood flow in response to a rise in
regional oxygen demand.
In conclusion, Jakob et al elegantly
demonstrate that routine nursing procedures may deleteriously affect the hepatosplanchnic circulation and, consequently,
visceral organ function. Furthermore,
most of it will remain undetected by systemic monitoring parameters, because
splanchnic perfusion can only be detected
by invasive procedures such as hepatic
vein catheterization. Because more than
one quarter of all recorded hepatic venous desaturations were not procedure
related, the clinical relevance of these
findings remains open. Nevertheless, in
analogy to the sepsis bundles, nursing
procedure guidelines are probably needed
to reduce the impact of these measures
on patient outcome, and Jakob et al have
the merit to highlight this often underestimated issue of critical care medicine.
Hendrik Bracht, MD
Florian Wagner, MD
Sektion Anästhesiologische
Pathophysiologie und
Verfahrensentwicklung
Universitätsklinikum
Ulm, Germany
Universitätsklinik für
Anästhesiologie
Universitätsklinikum
Ulm, Germany
Rainer Meierhenrich, MD, PhD
Universitätsklinik für
Anästhesiologie
Universitätsklinikum
Ulm, Germany
Peter Radermacher, MD, PhD
Sektion Anästhesiologische
Pathophysiologie und
Verfahrensentwicklung
Universitätsklinikum
752
Ulm, Germany
Michael Georgieff, MD, PhD
Universitätsklinik für
Anästhesiologie
Universitätsklinikum
Ulm, Germany
REFERENCES
1. Winslow EH, Clark AP, Whiote KM, et al:
Effects of a lateral turn on mixed venous
saturation and heart rate in critically ill
adults. Heart Lung 1990; 19:557–561
2. Tyler DO, Winslow EH, Clark AP, et al: Effects of a 1-minute back rub on mixed venous
oxygen saturation and heart rate in critically
ill patients. Heart Lung 1990; 19:562–565
3. Tidwell SL, Ryan WJ, Osguthorpe SG, et al:
Effects of position changes on mixed venous
oxygen saturation in patients after coronary
revascularization. Heart Lung 1990; 19:
574 –578
4. Barker M, Adams S: An evaluation of a single
chest physiotherapy treatment on mechanically ventilated patients with acute lung injury. Physiother Res Int 2002; 7:157–169
5. Walsh JM, Vanderwarf C, Hoscheit D, et al:
Unsuspected hemodynamic alterations during endotracheal suctioning. Chest 1989; 95:
162–165
6. Clark AP, Winslow EH, Tyler DO, et al: Effects of endotracheal suctioning on mixed
venous oxygen saturation and heart rate in
critically ill adults. Heart Lung 1990; 19:
552–557
7. Kinloch D: Instillation of normal saline during endotracheal suctioning. Effects on
mixed venous oxygen saturation. Am J Crit
Care 1999; 8:231–240
8. Boba A, Cincotti JJ, Piazza TE, et al: The
effects of apnea, endotracheal suction, and
oxygen insufflation, alone and in combination, upon arterial oxygen saturation in anesthetized patients. J Clin Lab Med 1959; 53:
680 – 685
9. Weissman C, Kemper M, Damask MC, et al:
Effect of routine intensive care interactions
on metabolic rate. Chest 1984; 86:815– 818
10. McCulloch KM, Ji SA, Raju TNK: Skin blood
flow changes during routine nursery procedures. Early Hum Dev 1995; 41:147–156
11. Gemma M, Tommasino C, Cerri M, et al:
Intracranial effects of endotracheal suctioning in the acute phase of head injury. J Neurosurg Anesth 2002; 14:50 –54
12. Klein P, Kemper M, Weissman C, et al: Attenuation of the hemodynamic responses to
chest physical therapy. Chest 1988; 93:
38 – 42
13. De Backer D, Creteur J, Noordally O, et al:
Does hepato-splanchnic VO2/DO2 dependency exist in critically ill septic patients?
Am J Respir Crit Care Med 1998; 157:
1219 –1225
14. Jakob SM, Parviainen I, Ruokonen E, et al:
Increased splanchnic oxygen extraction because of routine nursing procedures. Crit
Care Med 2009; 37: 483– 489
15. Maggiore SM, Lellouche F, Pigeot J, et al:
Prevention of endotracheal suctioninginduced alveolar drecruitment in acute lung
injury. Am J Respir Crit Care Med 2003;
167:1215–1224
16. Johnson KL, Kearney PA, Johnson SB, et al:
Closed versus open endotracheal suctioning:
Costs and physiologic consequences. Crit
Care Med 1994; 22:658 – 666
17. Cereda M, Villa F, Colombo E, et al: Closed
system endotracheal suctioning maintains
lung volume during volume-controlled mechanical ventilation. Intensive Care Med
2001; 27:648 – 654
18. Jongerden IP, Rovers MM, Grypdonck MH, et
al: Open and closed endotracheal suction systems in mechanically ventilated intensive
care patients: A meta-analysis. Crit Care Med
2007; 35:260 –270
19. Buchman TG, Stein PK, Goldstein B: Heart
rate variability in critical illness and critical
care. Curr Opin Crit Care 2002; 8:311–315
20. Goldstein B, Fiser DH, Kelly MM, et al: Decomplexification in critical illness and injury: Relationship between heart rate variability, severity of illness, and outcome. Crit
Care Med 1998; 26:352–357
21. Toweill D, Sonnenthal K, Kimberly B, et al:
Linear and nonlinear analysis of hemodynamic signals during sepsis and septic shock.
Crit Care Med 2000; 28:2051–2057
22. Korach M, Sharshar T, Jarrin I, et al: Cardiac
variability in critically ill adults: Influence of
sepsis. Crit Care Med 2001; 29:1380 –1385
Crit Care Med 2009 Vol. 37, No. 2
Video instruction for dispatch-assisted cardiopulmonary
resuscitation: Two steps forward and one step back!*
S
urvival from cardiac arrests is
dependent on the alignment of
the steps outlined in the revised chain of survival: early
recognition and call for help, early cardiopulmonary resuscitation (CPR), early
defibrillation, and early postresuscitation
care (1). Increasing the proportion of the
community, willing to provide bystander
CPR, would certainly facilitate “early
CPR.”
Much attention has been recently paid
to bystander CPR: its importance and
ways of reducing the barriers to its performance (2). This has been accompanied
by the reiteration by the American Heart
Association that hands-only (compression-only) CPR is an acceptable alternative when bystanders are untrained, unable, or unwilling to provide ventilation
as well as compressions (3).
Compression-only CPR appears particularly pertinent when applied to dispatch-assisted CPR. The ability to provide
instructions over the phone to a bystander is a widely established and effective practice (2, 4), especially if the instructions are simplified (such as
removing the additional instructions for
ventilation [5, 6]). If the advancing technology associated with telecommunication could be translated into a more sophisticated tool for instruction, then
dispatch-assisted bystander CPR could be
taken to a new level. Reports of some
potential benefits and limitations of such
technology are starting to appear (7–9).
In this issue of Critical Care Medicine,
Yang et al (10) present the results from
another such study.
*See also p. 490.
Key Words: cardiopulmonary resuscitation; dispatch-assist cardiopulmonary resuscitation; bystander
cardiopulmonary resuscitation; quality of cardiopulmonary resuscitation; video instruction
The author has consulted for AHA and is an Evidence Evacuation Expert for AHA.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194d2e1
Crit Care Med 2009 Vol. 37, No. 2
In a randomized study, the authors
evaluated the potential benefits of using
interactive video instruction to improve
the quality of dispatch-assisted, compression-only CPR performed on a
manikin by “bystanders” who had only
limited (if any) training in CPR. They
used an optimally placed video cell
phone in hands-free mode and provided
either voice-only instruction or “interactive voice and video demonstration
and feedback.” The addition of video communication (and, in particular, the realtime feedback) improved the rate of chest
compressions (from a median of 63 to 95.5
compressions per minute) and the depth of
compressions (from a median of 25 to 36
mm). The published literature suggests
that an increase in at least short-term survival would be expected from such improvements in compression rate (11) and
depth (12, 13). The downside of the interactive video group was a near 30-second
delay in the time till first compression, but
given the fact that a median of 0% of the
compressions in the control group was of
an “appropriate” depth (38 –51 mm [1.5–2
inches]), and that increased to a median of
20% in the interactive video group, this
delay becomes much less important.
This study provides many messages.
Clearly, the ideal positioning of the
video cell phone is crucial, and improvements could be made to the instructions and the time required to deliver them. As the technology, coverage,
and expertise with these devices are enhanced, we can expect further improvements in their effectiveness.
In summary, the study published by
Yang et al (10) has many limitations
and is obviously only a starting point
for the incorporation of some of the
more advanced functions of the modern-day communication devices. It appears that more sophisticated interactions (such as the use of video
instruction) can improve some aspects
of the quality of dispatch-assisted chest
compressions (depth and rate), but at
this stage, it comes at a cost (delayed
commencement of compressions). It
may be considered two steps forward
and one step back, but at least we appear to be making progress.
Peter Morley, MBBS, FRACP,
FANZCA, FJFICM
Royal Melbourne Hospital
University of Melbourne
Victoria, Australia
REFERENCES
1. Nolan J, Soar J, Eikeland H: The chain of
survival. Resuscitation 2006; 71:270 –271
2. Abella BS, Aufderheide TP, Eigel B, et al: Reducing barriers for implementation of bystanderinitiated cardiopulmonary resuscitation: A scientific statement from the American Heart
Association for healthcare providers, policymakers, and community leaders regarding the effectiveness of cardiopulmonary resuscitation. Circulation 2008; 117:704–709
3. Sayre MR, Berg RA, Cave DM, et al: Handsonly (compression-only) cardiopulmonary
resuscitation: A call to action for bystander
response to adults who experience out-ofhospital sudden cardiac arrest: A science advisory for the public from the American
Heart Association Emergency Cardiovascular
Care Committee. Circulation 2008; 117:
2162–2167
4. Vaillancourt C, Stiell IG, Wells GA: Understanding and improving low bystander CPR
rates: A systematic review of the literature.
CJEM 2008; 10:51– 65
5. Hallstrom A, Cobb L, Johnson E, et al: Cardiopulmonary resuscitation by chest compression alone or with mouth-to-mouth ventilation. N Engl J Med 2000; 342:1546 –1553
6. Roppolo LP, Pepe PE, Cimon N, et al: Modified cardiopulmonary resuscitation (CPR) instruction protocols for emergency medical
dispatchers: Rationale and recommendations. Resuscitation 2005; 65:203–210
7. You JS, Park S, Chung SP, et al: The era of
audiovisual dispatch using cellular phones
with video telephony. Resuscitation 2008;
76:486 – 487
8. Johnsen E, Bolle SR: To see or not to see—
Better dispatcher-assisted CPR with videocalls? A qualitative study based on simulated
trials. Resuscitation 2008; 78:320 –326
9. Yang CW, Wang HC, Chiang WC, et al: Impact of adding video communication to dispatch instructions on the quality of rescue
breathing in simulated cardiac arrests—A
753
randomized controlled study. Resuscitation
2008; 78:327–332
10. Yang C-W, Wang H-C, Chiang W-C, et al: Interactive video instruction improves the quality of
dispatcher-assisted chest compression-only cardiopulmonary resuscitation in simulated cardiac
arrests. Crit Care Med 2009; 37: 490–495
11. Abella BS, Sandbo N, Vassilatos P, et al:
Chest compression rates during cardiopulmonary resuscitation are suboptimal: A prospective study during in-hospital cardiac arrest. Circulation 2005; 111:428 – 434
12. Edelson DP, Abella BS, Kramer-Johansen J, et
al: Effects of compression depth and pre-shock
pauses predict defibrillation failure during cardiac arrest. Resuscitation 2006; 71:137–145
13. Kramer-Johansen J, Myklebust H, Wik L, et
al: Quality of out-of-hospital cardiopulmonary resuscitation with real time automated
feedback: A prospective interventional study.
Resuscitation 2006; 71:283–292
Continuous renal replacement therapy circuit contamination: New
tale of an old problem?*
D
ialysis and continuous renal
replacement therapies’ (CRRT)
technology improved tremendously since the first hemodialysis for chronic renal failure in
1960 and the first CRRT in 1977 (1, 2). As
technical challenges of dialysis treatment
were overcome and patients began to survive, problems of preparing dialysate
from tap water became apparent. Chemical and bacterial contamination of the
water used to prepare the dialysate led to
the developments of strict standards for
water used for hemodialysis by various
organizations, with slight differences (3).
Although CRRT is the main form of renal
support provided in intensive care units
worldwide (4), widely recognized standards
have not been applied to CRRT as yet.
Microbial contamination of intravenous (IV) fluids has been known for long.
Episodes of septicemia associated with IV
therapy were reported in the 1970s,
which were linked to manufactured infusion products; however, extrinsic sources
of contamination during use were also
identified in a significant proportion of IV
therapy infusion systems (5) and are considered to be a more common source of
contamination (5–7). Guidelines for the
prevention of intravascular catheterrelated infections address how to reduce
the microbial contamination of IV infusions and potential hazards to the patients (6).
*See also p. 496.
Key Words: continuous renal replacement therapy;
microbiological; contamination; biofilm; dialysate; intravenous; infusion systems
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194d485
754
Furthermore, significant advances
during the same time periods led to
guidelines for surgical site infection prophylaxis and operating room conditions
(8). The source of pathogens in many
surgical site infections is the endogenous
flora of the patient’s skin, mucous membranes, or hollow viscera; however, exogenous sources include surgical personnel, the operating room environment
(including air), instruments, and other
materials brought to the sterile field during an operation. Room air may contain
microbial-laden dust, lint, skin squames,
or respiratory droplets that can also contribute to contamination of IV infusion
equipment during use (8).
In this issue of Critical Care Medicine, Kanagasundaram et al (9) look at
the microbiological integrity of a continuous venovenous hemofiltration (a
form of CRRT) circuit using a commercially available replacement solution.
They found that of the 24 replacement
fluid cultures, 9 breached European
Pharmacopoeia standards for ultrapure
water. One of 24 endotoxin measurements breached European Pharmacopoeia standards. Internal tubing cultures were negative, but electron
microscopy revealed 13 of the 24 collected tubing samples to be contaminated with biofilm. Only 7 of the 24
studied circuits proved to be free from
microbial contamination. They found
no clear relationship between circuit
lifespan and rates of contamination, but
this should be interpreted with caution
because of their small sample size.
Kanagasundaram et al used highly
sensitive microbiological techniques to
examine replacement fluid, quantitatively
assess endotoxin, and examine tubing
specimens via electron microscopy for
biofilms. They took precautions to mini-
mize contamination during sampling;
however, they did not separately analyze
unused tubing or commercially available
solutions directly from the bags before
use or other bags of unused fluid with the
same lot numbers. They also did not evaluate room air as a potential source of
microbial contamination (9).
CRRT systems involve a roller blood
pump device with additional pumps to
provide the circulation of dialysate and
replacement fluids, a dialysis filter, dialysate and replacement fluids, and the necessary infusion tubing systems, all of
which are usually incorporated in a disposable cartridge or assembled separately
depending on the device used (2). The continuous venovenous hemofiltration system
that Kanagasundaram et al used was an
integrated, single-use circuitry with a commercially available replacement fluid (as
usual in the current practice). This CRRT
system is essentially a combination of a
dialysis filter and a needleless infusion system. Thus, their findings should be interpreted in the same context. Indeed, their
results are not surprising and are reminiscent of data on “in use contamination of IV
fluids” (5).
There are four potential routes of
access for the organisms to colonize an
intravascular device and subsequently
cause blood stream infections: invasion
of the skin insertion site, contamination of the catheter hub, hematogenous
spread from a distant site of infection,
or rarely infusion of contaminated fluid
through the device. The first two
sources are the most important. Surface colonization of the tubing, probably originating from the skin, predominates in short-term catheters (in place
less than 10 days), such as peripheral IV
catheters, nontunneled central venous
catheters, and arterial catheters. InCrit Care Med 2009 Vol. 37, No. 2
traluminal colonization due to hub
contamination increases over time and
becomes predominant after 30 days of
long-term devices (tunneled central venous catheters, peripherally inserted
central catheters, and subcutaneous ports)
(6, 7, 10). Stopcocks (used for injection of
medications, administration of IV infusions, and collection of blood samples) represent a potential portal of entry for microorganisms into vascular access catheters
and IV fluids (6).
Catheter material/structure and virulence of microorganisms are important
factors along with biofilm formation (6,
7, 10 –12). Biofilm is a living community
of microorganisms, embedded in their
extracellular polymeric matrix and other
components deposited from bodily fluids
adherent to the catheter surface. Biofilm
can develop within a few hours of device
placement (both inside and outside the
lumen in the case of a catheter). Biofilms
have major medical significance. Biofilms
are less susceptible to the effects of antimicrobial agents and host defenses, making the treatment of infections very difficult (7, 10 –12).
Where do we go from here? Can we
blame the dialysate/replacement fluids
for the contamination? This can only be
answered by specifically culturing and
assaying these fluids before, and during, inline use. More important, we
should implement all preventive measures as recommended by current
guidelines to prevent catheter-related
infections and surgical site infections
first. Educating all personnel involved
is critical. Achieving operating conditions in intensive care unit is not feasible, but maximum protection during
CRRT set up and disconnection of the
Crit Care Med 2009 Vol. 37, No. 2
circuit can be used by nursing staff. We
may need to use new antiseptic barrier
caps with 2% chlorhexidine gluconate
in alcohol rather than routine alcohol
swabs for disinfection of needleless
catheter connectors and access ports
(13). We should begin culturing CRRT
fluids when the patients develop fever
or evidence of sepsis. Because biofilms
can form rapidly on the intraluminal
surfaces of IV tubing, we should probably adopt shorter changing periods
(i.e., 24 hours) for the CRRT circuit
filter and tubing, especially because
their cost decreased over time, and
maintaining patency is also an issue. For
the same token, we should perhaps begin
routine use of catheter lock solutions with
antibiotics and chelating agents that are
promising in eliminating biofilms in the
catheter lumens (7, 10, 14). Clearly, more
work needs to be done in this area, but this
is a step forward.
4.
5.
6.
7.
8.
9.
ACKNOWLEDGMENTS
I thank Dr. Matthew Levison for his valuable input and discussions about the topic.
Bulent Cuhaci, MD
Department of Medicine, Drexel
University College of
Medicine
Philadelphia, PA
REFERENCES
1. Blagg CR: The early history of dialysis for
chronic renal failure in the United States: A
view from Seattle. Am J Kidney Dis 2007;
49:482– 496
2. Dirkes SM: Continuous renal replacement
therapy: Dialytic therapy for acute renal failure in intensive care. Nephrol Nurs J 2000;
27: 581–592
3. Ward R: Worldwide water standards for he-
10.
11.
12.
13.
14.
modialysis. Haemodialysis Int 2007; 11:
S18 –S25
Uchino S, Bellomo R, Morimatsu H, et al:
Continuous renal replacement therapy: A
worldwide practice survey. Intensive Care
Med 2007; 33:1563–1570
Maki DG, Anderson RL, Shulman JA: In-Use
contamination of intravenous infusion fluid.
Appl Microbiol 1974; 28:778 –784
Centers for Disease Control and Prevention.
Guidelines for the prevention of intravascular catheter-related infections. MMWR Morb
Mortal Wkly Rep 2002; 51(RR-10): 1–32
Trautner BW, Darouiche RO: Catheterassociated infections. Pathogenesis affects
prevention. Arch Intern Med 2004; 164:
842– 850
Mangram AJ, Horan TC, Pearson ML: Guideline for prevention of surgical site infection,
1999. The Hospital Infection Control Practices Advisory Committee. Infect control
Hosp Epidemiol 1999; 20:247–278
Moore I, Bhat R, Hoenich NA, et al: A microbiological survey of bicarbonate-based replacement circuits in continuous venovenous hemofiltration. Crit Care Med 2009;
37:496 –500
Sullivan R, Samuel V, Le C, et al: Hemodialysis vascular catheter-related bacteremia.
Am J Med Sci 2007; 334:458 – 465
Passerini L, Lan K, Costerton J, et al: Biofilms on indwelling vascular catheters. Crit
Care Med 1992; 20:665– 673
Dolan RM: Biofilm: Microbial life on surfaces. Emerging infectious diseases. 2002;
8:881– 890
Menyhay SZ, Maki DG: Disinfection of
needleless catheter connectors and access
ports with alcohol may not prevent microbial
entry: The promise of a novel antisepticbarrier cap. Infect Control Hosp Epidemiol
2006; 27:23–27
Raad II, Fang X, Keutgen XM, et al: The role
of chelators in preventing biofilm formation
and catheter-related bloodstream infections.
Curr Opin Infect Dis 2008; 21:385–392
755
Arginine pharmacokinetics: Not a new paradigm but the old
pharmacology*
I
n the last 15 yrs, an increasing
number of articles have dealt with
the hypothesis that critically ill
patients in the catabolic state will
develop nutrient deficiencies and immune dysfunction. They usually show
low plasmatic levels of glutamine and arginine despite adequate nutritional support and are considered conditionally essential in this setting (1, 2). Both have
been empirically added, alone or in combination with other substrates, to standard diets trying to demonstrate their
beneficial effects on laboratory, immunologic, and clinical parameters in comparison with standard diets. These formulas
have been included in the wide concept of
immunonutrition with variable success
in terms of clinical outcomes but with
entertained controversies (3–5). Furthermore, this concept has been questioned,
and now the new paradigm of pharmaconutrition has been introduced (6). In fact,
what we must do is rethink the research
we are doing and, if we will use arginine
or glutamine like a drug, we are urged to
apply the principles of the pharmacology
to get accurate and reproducible results.
In this issue of Critical Care Medicine,
Loi et al (7) present a good example of
pharmacologic study. They administered
two different isocaloric and isonitrogenous diets, one standard and the other
enriched with arginine, to observe the
changes in plasmatic levels of glutamine
in the first 90 minutes of diet administration, once the patients were fed enterally and with a washout period of 3 hours.
The authors found a significant increase
of plasmatic levels of arginine and glutamine in patients treated with the arginine-containing diet. Furthermore, the
areas under the curve between arginine
*See also p. 501.
Key Words: arginine; glutamine; pharmacokinetics;
protein metabolism
The author has received honoraria from Fresenius
Rabl 900 and Novariis 600.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194d522
756
and proline and arginine and ornithine
showed a good correlation. Interestingly,
both groups of patients had similar nitrogen balance or proteolysis. This study
confirms in humans that there is a common fate between arginine and glutamine
through ornithine in the liver, as the
authors suggested in a previous experimental study (8). Nevertheless, this study
has some limitations. First, the study diet
contains high amounts of omega-3 fatty
acids, vitamins C and E, and selenium.
We do not know if changes in interleukin
production mediated for these fatty acids
or a higher level of antioxidants can modify glutamine consumption by immune
cells or for glutathione production. Besides, the washout period seems to be
very short and a longer fasting period like
8 or 12 hours will be a better approach
(9). Second, the study population has in
fact two subgroups: one group of patients
with cancer underwent an elective surgical procedure and another group of patients with septic shock and pancreatitis
underwent an emergent surgical procedure. Although the interaction between
the two groups of patients is not significant, it makes the overall population heterogeneous enough to extrapolate these
results to different populations without a
separate analysis of the data.
Nevertheless, Loi et al present a solid
pharmacokinetic study that demonstrates
that arginine delivered enterally increases the levels of plasmatic glutamine.
Arginine and glutamine regulate the immune response through different effects
(10, 11). Furthermore, glutamine and arginine share complex metabolic pathways
well explained in a recent review of this
journal (12). Arginine is metabolized by
arginase and produces urea, and ornithine and the last one gives citrulline or
glutamine and proline, as demonstrated
by Loi et al. Arginine is also transformed
to citrulline and nitric oxide by inducible
nitric oxide synthase. The temporal
switch of arginine as a substrate for the
inducible nitric oxide synthase to ornithine–proline axis is regulated by inflam-
matory cytokines and by the arginine metabolites themselves (13). On the other
hand, glutamine is a primary fuel for enterocytes and for gluconeogenesis in the
liver, is a precursor of pyrimidine and
purine, is incorporated to glutathione,
and serves as a precursor for the de novo
production of arginine through the citrulline–arginine intestinal–renal pathway
(14). Close to the complexity of these
metabolic pathways, we should consider
gene expression of different isoenzymes,
different patterns of tissue expression,
and the route of administration.
Glutamine seems to be beneficial in
critically ill patients, whereas arginine
supplementation is still under debate. Administering high doses of these amino
acids needs further research looking at
the dose, the way of administration, and
the physiologic and clinical effects like
other drugs. Maybe this is an unrealistic
approach, but it is clear that both amino
acids exert regulatory functions different
from being only nutrients. And finally, if
arginine can produce an excessive and
sometimes deleterious amount of nitric
oxide, and therefore is an adverse event,
why not use glutamine alone?
Teodoro Grau, MD, PhD
Servicio de Medicina Intensiva
Hospital Universitario Doce de
Octubre
Madrid, Spain
REFERENCES
1. Lacey JM, Wilmore DW: Is glutamine a conditionally essential amino acid? Nutr Rev
1990; 48:297–309
2. Luiking YL, Poeze M, Dejong CH, et al: Sepsis: An arginine deficiency state. Crit Care
Med 2004; 32:2135–2145
3. Heyland DK, Novak F, Drover JW, et al:
Should immunonutrition become routine in
critically ill patients? JAMA 2001; 286:22–29
4. Consensus recommendations from the US
summit on immune-enhancing enteral therapy. JPEN J Parenter Enteral Nutr 2001;
25(Suppl):S61–S62
5. Montejo JC, Zarazaga A, Lopez-Martinez J, et
al; for the Spanish Society of Intensive Care
Medicine and Coronary Units: Immunonutrition in the intensive care unit. A systematic
Crit Care Med 2009 Vol. 37, No. 2
review and consensus statement. Clin Nutr
2003; 22:221–233
6. Jones NE, Heyland DK: Pharmaconutrition:
A new emerging paradigm. Curr Opin Gastroenterol 2008; 24:215–222
7. Löi C, Zazzo J-F, Delpierre E, et al: Increasing
plasma glutamine in postoperative patients fed an
arginine-rich immune-enhancing diet—A pharmacokinetic randomized controlled study. Crit
Care Med 2009; 37:501–509
8. Moynard C, Belabed L, Gupta S, et al: Arginine-enriched diet limits plasma and muscle
depletion in head-injured rats. Nutrition
2006; 22:1039 –1044
9. Jackson NC, Carroll PV, Russell-Jones DL, et al:
The metabolic consequences of critical illness:
Acute effects on glutamine and protein metabolism. Am J Physiol 1999; 276:E163–E170
10. Melis GC, ter Wengel N, Boelens PG, et al:
Glutamine: Recent developments in research
on the clinical significance of glutamine. Curr
Opin Clin Nutr Metab Care 2004; 7:59 –70
11. Popovic PJ, Zeh JH III, Ochoa JB: Arginine and
immunity. J Nutr 2007; 137:1681S–1686S
12. Vermeulen MAR, van de Poll MCG, LighartMelis G, et al: Specific amino acids in the
critically ill patient-exogenous glutamine/
arginine: A common denominator. Crit
Care Med 2007; 35(Suppl):S568 –S576
13. Satriano J: Arginine pathways and the inflammatory response: Interregulation of nitric oxide and polyamines [review]. Amino
Acids 2004; 26:321–329
14. Neu J, DeMarco V, Li N: Glutamine: Clinical
applications and mechanisms of action. Curr
Opin Clin Nutr Metab Care 2002; 5:69 –75
Meaning of pulse pressure variation during cardiac surgery:
Everything is open*
F
luid management is of major
importance in critically ill patients and in surgery patients.
Static preload indicators, like
cardiac filling pressures, are of poor value
to assess fluid responsiveness (1–3).
Functional hemodynamic parameters,
like arterial pulse pressure variation
(PPV) and pulse contour stroke volume
variation (SVV), have gained wide popularity as predictors of fluid responsiveness
in mechanically ventilated patients (4). It
must be remembered that the mechanical insufflation decreases preload and increases afterload of the right ventricle.
The right ventricular preload reduction is
secondary to the decrease of the venous
return pressure gradient related to the
inspiratory increase in intrathoracic pressure. The increase in right ventricular
afterload is related to the inspiratory increase in transpulmonary pressure (alveolar pressure minus intrathoracic pressure). The reduction in preload and the
increase in afterload of the right ventricle
both lead to a decrease in right ventricular stroke volume, which, in turn, leads
to a decrease in left ventricular filling
after a phase lag of 2–3 heart beats related
to the long blood pulmonary transit time.
The resulting left ventricular preload re-
*See also p. 510.
Key Words: open chest surgery; cardiac surgery;
pulse pressure variation; stroke volume variation; fluid
responsiveness; cardiac preload
The author has consulted for Pulsion Medical
Systems.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194d575
Crit Care Med 2009 Vol. 37, No. 2
duction may induce a decrease in the left
ventricular stroke volume, which is minimal during the expiratory period.
Interestingly, the cyclic changes in
right ventricular preload induced by mechanical ventilation, result in greater cyclic changes in right ventricular stroke
volume when the right ventricle operates
on the steep rather than on the flat portion of the Frank–Starling curve. The cyclic changes in right ventricular stroke
volume and, hence, in left ventricular
preload result in greater cyclic changes in
left ventricular stroke volume when the
left ventricle operates on the steep portion of the Frank–Starling curve. Thus,
the magnitude of the respiratory changes
in left ventricular stroke volume should
be an indicator of biventricular preload
responsiveness and, hence, of fluid responsiveness. In this regard, PPV (5–7)
and SVV (7–9) have been demonstrated as
good predictors of fluid responsiveness in
various clinical settings. Two obvious
conditions are required to adequetely interpret such dynamic parameters: controlled positive pressure ventilation with
no spontaneous breathing activity (10,
11) and absence of cardiac arrhythmias
(10). Because changes in intrathoracic
pressure and transpulmonary pressure
are the major determinants of the cyclic
cardiovascular consequences of positive
pressure ventilation, a sufficiently hightidal volume (⬎7 mL/kg) is obviously also
required for well interpreting functional
dynamic parameters (12). These three
conditions are generally met in anesthetized patients undergoing surgery. Nevertheless, under open chest conditions, respiratory changes in intrathoracic
pressure are less pronounced so that respiratory changes in stroke volume or in
pulse pressure are expected to be low
even in fluid responsive patients. In this
issue of Critical Care Medicine, de Waal
et al (13) have tested this hypothesis by
evaluating the ability of PPV and SVV to
predict fluid responsiveness under both
open and closed chest conditions in patients undergoing coronary artery bypass
graft surgery. First, they confirmed that
in closed chest conditions (tidal volume:
8 mL/kg), PPV and SVV were better predictors of fluid responsiveness than static
measures of preload, such as central venous pressure or global end-diastolic volume. Second, they showed that neither
PPV and SVV nor static preload indicators
were able to predict accurately fluid responsiveness under open chest conditions, although the number of nonresponders was very low (3 of 18 patients).
In half of the fluid responders, PPV was
⬍10%, a value in agreement with the
authors’ hypothesis (13). It is likely that
the attenuation of cyclic changes in intrathoracic pressure in open chest conditions accounted for these findings. It
must be stressed, however, that in the
other half of the fluid responders, PPV
was higher than 11% (up to 22%). Surprisingly, the authors did not highlight
these striking results. One could postulate that under open chest conditions, a
high PPV (or SVV) is secondary to the
cyclic changes in transpulmonary pressure, which are potentially large in the
absence of significant changes in intrathoracic pressure. A marked inspiratory increase in transpulmonary pressure
could decrease right ventricular stroke
757
volume at inspiration through an increase in the resistance of intra-alveolar
microvessels. Thus, even if the pulmonary vasculature was in West’s zone 3
conditions at expiration in these patients, the marked increase in transpulmonary pressure had probably produced
zone 2 conditions at inspiration in some
of them (14). It could then be postulated
that fluid infusion by increasing the pulmonary venous pressure attenuated the
transfer from zone 3 to zone 2 at inspiration and, hence, the inspiratory decrease in right ventricular stroke volume.
Although this hypothesis is speculative, it
is supported by the fact that PPV (and
SVV) decreased while stroke volume increased after fluid infusion in this subgroup of patients. Similar findings were
previously reported in open chest surgery
patients ventilated with a tidal volume of
10 mL/kg (15). Although they enrolled a
limited number of patients, de Waal et al
(13) have thus brought evidence that the
presence of high PPV (or SVV) is indicative of fluid responsiveness under both
closed and open chest conditions. However, under open chest conditions, other
tools are still required to diagnose the
origin of hemodynamic instability because the presence of low PPV and SVV
cannot preclude a positive hemodynamic
response to fluid.
Jean-Louis Teboul, MD, PhD
Service de Réanimation Médicale
Centre Hospitalo-Universitaire de
Bicêtre
Assistance Publique-Hôpitaux de
Paris
EA 4046 Université Paris-Sud
Le Kremlin-Bicêtre, France
REFERENCES
1. Kumar A, Anel R, Bunnell, et al: Pulmonary
artery occlusion pressure and central venous
pressure fail to predict ventricular filling volume, cardiac performance, or the response to
volume infusion in normal subjects. Crit
Care Med 2004; 32:691– 699
2. Osman D, Ridel C, Ray P, et al: Cardiac filling
pressures are not appropriate to predict hemodynamic response to volume challenge.
Crit Care Med 2007; 35:64 – 68
3. Marik PE, Baram M, Vahid B: Does central
venous pressure predict fluid responsiveness? A
systematic review of the literature and the tale
of seven mares. Chest 2008; 134:172–178
4. Michard F, Teboul JL: Predicting fluid responsiveness in ICU patients: A critical analysis of
the evidence. Chest 2002; 121:2000 –2008
5. Michard F, Boussat S, Chemla D, et al: Relation between respiratory changes in arterial
pulse pressure and fluid responsiveness in
septic patients with acute circulatory failure.
Am J Respir Crit Care Med 2000; 162:
134 –138
6. Kramer A, Zygun D, Hawes H, et al: Pulse
pressure variation predicts fluid responsiveness following coronary artery bypass surgery. Chest 2004; 126:1563–1568
7. Preisman S, Kogan S, Berkenstadt H, et al:
Predicting fluid responsiveness in patients
undergoing cardiac surgery: Functional haemodynamic parameters including the Respi-
8.
9.
10.
11.
12.
13.
14.
15.
ratory Systolic Variation Test and static preload indicators. Br J Anaesth 2005; 95:
746 –755
Berkenstadt H, Margalit N, Hadani M, et al:
Stroke volume variation as a predictor of
fluid responsiveness in patients undergoing
brain surgery. Anesth Analg 2001; 92:
984 –989
Reuter DA, Felbinger TW, Schmidt C, et al:
Stroke volume variations for assessment of
cardiac responsiveness to volume loading in
mechanically ventilated patients after cardiac
surgery. Intensive Care Med 2002; 28:
392–398
Monnet X, Rienzo M, Osman D, et al: Passive
leg raising predicts fluid responsiveness in
the critically ill. Crit Care Med 2006; 34:
1402–1407
Heenen S, De Backer D, Vincent JL: How can
the response to volume expansion in patients
with spontaneous respiratory movements be
predicted? Crit Care 2006; 10:R102
De Backer D, Heenen S, Piagnerelli M, et al:
Pulse pressure variations to predict fluid responsiveness: Influence of tidal volume. Intensive Care Med 2005; 31:517–523
de Waal EEC, Rex S, Kruitwagen CLJJ, et al:
Dynamic preload indicators fail to predict fluid
responsiveness in open-chest conditions. Crit
Care Med 2009; 37:510 –515
Jardin F, Delorme G, Hardy A, et al: Reevaluation of hemodynamic consequences of
positive pressure ventilation: Emphasis on
cyclic right ventricular afterloading by mechanical lung inflation. Anesthesiology 1990;
72:966 –970
Reuter DA, Goepfert MS, Goresch T, et al:
Assessing fluid responsiveness during open
chest conditions. Br J Anaesth 2005; 94:
318 –323
Antioxidant therapy: Reducing malaria severity?*
E
ach year, infection with Plasmodium falciparum causes
300 – 600 million illnesses
worldwide (1). A significant
number of patients will progress to severe
malaria with organ dysfunction, which is
more common in the immunologically
*See also p. 516.
Key Words: oxidative stress; cytoadherence;
Plasmodium falciparum; endothelium; artesunate;
N-acetylcysteine; lactate; glucose-6-phosphate dehydrogenase deficiency
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194d5de
758
naïve: young children and travelers or
other adults with only sporadic exposure
to infected mosquitoes. Mortality from
severe malaria ranges from 5% to 40%,
and death often occurs shortly after hospital admission. Despite improved survival with artesunate treatment (2), mortality remains high in the first 24 hours
of hospitalization, and therefore, adjunctive therapies are still urgently needed.
Neither hypovolemia, cardiogenic
shock, nor vasodilatory shock is necessary
for organ dysfunction in severe malaria (3).
The impairment of tissue perfusion occurs
within microvessels, where parasitized red
blood cells (RBCs) adhere to endothelial
cells and circulating immune cells via spe-
cific interactions between the parasiteencoded adherence ligand P. falciparum
erythrocyte membrane protein 1 on the
RBC surface and the host-encoded adhesion receptors cluster of differentiation 36
and intercellular adhesion molecule-1 (4).
Adherence is associated with inflammatory cytokine secretion, adhesion molecule expression, tissue factor display,
and platelet aggregation (5)—findings
that are evident on postmortem examination of patients with severe malaria
(6). Microcirculatory dysfunction may
be worsened as nitric oxide is scavenged
by cell-free hemoglobin released during
hemolysis and by reactive oxygen species (7).
Crit Care Med 2009 Vol. 37, No. 2
In severe malaria, reactive oxygen species are generated by parasite hemoglobin metabolism in RBCs, nicotinamide
adenine dinucleotide phosphate oxidase
in phagocytes, and nitric oxide synthase
when the substrate arginine or cofactor
tetrahydrobiopterin is lacking. Plasma
hemoglobin released from lysed RBCs
may also catalyze the generation of reactive oxygen species. Patients with severe
malaria have increased reactive oxygen
species products in urine (8), decreased
alpha-tocopherol in RBC membranes (9),
and decreased deformability of RBCs under shear stress. Decreased deformability
of RBCs is associated with mortality (10);
this was the impetus for the study by
Charunwatthana et al (8), who hypothesized that decreased deformability impedes
the transit of RBCs through capillaries, impairing oxygen delivery. Because N-acetylcysteine (NAC) restores normal deformability of RBCs in vitro by repleting glutathione
reserves (11), treatment with NAC was expected to improve RBC deformability and
oxygen delivery in vivo.
However, oxidant stress in RBCs could
offer some compensatory benefits to a
patient with severe malaria. First, oxidative stress is a fundamental mechanism
for killing phagocytosed pathogens, including P. falciparum. Treatment with a
potent antioxidant might conceivably
protect parasites from the oxidative burst
of phagocytes and could antagonize the
oxidation-mediated antimalarial effects of
artesunate. In vitro, the parasiticidal effect of artesunate is reduced when given
simultaneously with NAC, but is unaffected when NAC is given 2 hours after
artesunate (12). In the present study,
Charunwatthana et al administered NAC
2 hours after the first dose of artesunate
and measured parasitemia every 6 hours
to see whether NAC would impair parasite killing by artesunate.
Second, oxidative stress in RBCs may
play a role in weakening adherence interactions between the parasitized RBCs and
the microvascular endothelium. This is
best illustrated by the classic protective effect of sickle cell trait against severe malaria. Infected RBCs from patients with
sickle cell trait show aberrant display of the
parasite-encoded adhesion ligand P. falciparum erythrocyte membrane protein 1
and reduced strength of adhesion to microvascular endothelial cells and blood monocytes in vitro (13). This phenotype may be
reproduced in RBCs with glucose-6-phosphate dehydrogenase deficiency or other
states of oxidative stress.
Crit Care Med 2009 Vol. 37, No. 2
Third, although oxidative stress in infected RBCs might be beneficial, oxidative stress in endothelial cells causes adhesion molecule expression, apoptosis,
and capillary leak. Endothelial cell injury
by adherent parasitized RBCs is ameliorated by superoxide dismutase, ascorbic
acid, or tocopherol in vitro, highlighting
the potential therapeutic benefit of antioxidants as endothelial cell protectors (14).
So what will be the effect of a systemic
antioxidant in severe malaria? In the study
by Charunwatthana et al (8), adults with
severe malaria were treated with artesunate
and randomized to receive NAC (dosed as
for acetaminophen overdose) or placebo.
NAC had no effect on the primary or secondary end points of the study: lactate
clearance time, coma recovery time, parasite clearance time, fever clearance time, or
mortality. End products of oxidative stress
(F2-isoprostanes) were found to be elevated
in the urine of patients with severe malaria
compared with uncomplicated malaria, but
were unchanged by NAC treatment. Although red cell deformability was lower in
fatal cases than in survivors of severe malaria, it was not changed by NAC treatment.
Why was NAC ineffective? In a pilot
study (15), 30 patients were randomized
to NAC or placebo, and at 24 hours a
greater proportion of patients in the
NAC-treated arm had normal lactate levels compared with the placebo arm (10 of
15 vs. 3 of 15, p ⫽ 0.01); however, the
placebo group had higher baseline parasitemia and bilirubin and lower Glasgow
coma scores than the treatment group.
Baseline differences in disease severity
could have overestimated the benefit, if
any, of NAC treatment. As a consequence,
the current study may have been underpowered to detect the true effect of NAC
treatment on severe malaria. Antimalarial
treatment was also different in the current
study: NAC was given 2 hours after artesunate, a pro-oxidant drug, compared with
NAC being given simultaneously with quinine in the pilot study. Furthermore, genetic polymorphisms that influence RBC
oxidative stress (such as glucose-6-phosphate dehydrogenase deficiency) were not
accounted for, but could have modified the
response to antioxidant therapy.
In hindsight, it is unclear what effect
NAC should have had on severe malaria.
Oxidative stress is considered beneficial
for parasite killing and for weakening the
adherence between infected RBCs and endothelial cells or monocytes. On the other
hand, oxidative stress causes endothelial injury and impairs RBC deformability. The
results of the current study are neutral.
Does this close the book on antioxidants for
severe malaria? No, but future strategies
might specifically target antioxidants to the
endothelium while enhancing oxidative
stress in infected RBCs.
On second thought, combining a systemic antioxidant NAC with an RBCspecific pro-oxidant artesunate may have
accomplished just that.
Hans C. Ackerman, MD, DPhil, MSc
Laboratory of Malaria and Vector
Research
National Institute of Allergy and
Infectious Diseases
Rockville, MD
Critical Care Medicine
Department
National Institutes of Health
Clinical Center
Bethesda, MD
Steven D. Beaudry, BS
Rick M. Fairhurst, MD, PhD
Laboratory of Malaria and Vector
Research
National Institute of Allergy and
Infectious Diseases
Rockville, MD
REFERENCES
1. Snow RW, Guerra CA, Noor AM, et al: The
global distribution of clinical episodes of
Plasmodium falciparum malaria. Nature
2005; 434:214 –217
2. Dondorp A, Nosten F, Stepniewska K, et al:
Artesunate versus quinine for treatment of
severe falciparum malaria: A randomised
trial. Lancet 2005; 366:717–725
3. Planche T, Onanga M, Schwenk A, et al:
Assessment of volume depletion in children
with malaria. PLoS Med 2004; 1:e18
4. Ockenhouse CF, Ho M, Tandon NN, et al:
Molecular basis of sequestration in severe
and uncomplicated Plasmodium falciparum
malaria: Differential adhesion of infected
erythrocytes to CD36 and ICAM-1. J Infect
Dis 1991; 164:163–169
5. Francischetti IM, Seydel KB, Monteiro RQ:
Blood coagulation, inflammation, and malaria. Microcirculation 2008; 15:81–107
6. Taylor TE, Fu WJ, Carr RA, et al: Differentiating the pathologies of cerebral malaria by
postmortem parasite counts. Nat Med 2004;
10:143–145
7. Gramaglia I, Sobolewski P, Meays D, et al:
Low nitric oxide bioavailability contributes
to the genesis of experimental cerebral malaria. Nat Med 2006; 12:1417–1422
8. Charunwatthana P, Faiz MA, Ruangveerayut
R, et al: N-acetylcysteine as adjunctive treatment in severe malaria: A randomized double
blinded placebo controlled-clinical trial. Crit
Care Med 2009; 37:516 –522
759
9. Griffiths MJ, Ndungu F, Baird KL, et al: Oxidative stress and erythrocyte damage in Kenyan
children with severe Plasmodium falciparum
malaria. Br J Haematol 2001; 113:486 – 491
10. Dondorp A, Angus BJ, Hardeman MR, et al:
Prognostic significance of reduced red blood
cell deformability in severe falciparum malaria. Am J Trop Med Hyg 1997; 57:507–511
11. Nuchsongsin F, Chotivanich K, Charunwatthana P, et al: Effects of malaria heme prod-
ucts on red blood cell deformability. Am J
Trop Med Hyg 2007; 77:617– 622
12. Arreesrisom P, Dondorp AM, Looareesuwan S,
et al: Suppressive effects of the antioxidant N-acetylcysteine on the antimalarial activity of artesunate. Parasitol
Int 2007; 56:221–226
13. Cholera R, Brittain NJ, Gillrie MR, et al:
Impaired cytoadherence of Plasmodium falciparum-infected erythrocytes containing
sickle hemoglobin. Proc Natl Acad Sci USA
2008; 105:991–996
14. Taoufiq Z, Pino P, Dugas N, et al: Transient
supplementation of superoxide dismutase
protects endothelial cells against Plasmodium falciparum-induced oxidative stress.
Mol Biochem Parasitol 2006; 150:166 –173
15. Watt G, Jongsakul K, Ruangvirayuth R: A pilot
study of N-acetylcysteine as adjunctive therapy
for severe malaria. QJM 2002; 95:285–290
Is responsiveness to family wishes an expression of professional
transcendence?*
T
he authors are to be praised
for proactively exploring the
concept of involving family
wishes in the decision to admit
to the intensive care unit. Participation in a
shared decision-making model is endorsed
by the Society of Critical Care Medicine (1).
In this issue of Critical Care Medicine, Escher et al (2) determine from the results of
surveys administered to physicians in Switzerland that older physicians and those
with self-determined knowledge of ethics
were more likely to admit a hypothetical
young woman with uremic syndrome in
the intensive care unit. Physicians were
also more likely to admit if the family stated
that they wanted everything done vs. “spare
useless suffering.”
As a point of discussion, for those engaged in providing critical care to the ill
and injured, the manner in which the
phrase “spare useless suffering” was used in
this study prompts a gutteral reaction to
exclaim that critical care does not uniformly produce “useless suffering”! If a few
days of intensive treatment and tests produce a dramatic effect on symptomotology
vs. weeks of protracted untreated illness on
the ward, which is suffering? One would
wonder how lay people would react. Do
they know that when they use the phrase
“do everything” that the physician interprets that as “admit to the intensive care
unit” and when they use the phrase “do not
*See also p. 528.
Key Words: ethics; end of life; transcendence;
decision-making; family; critical care
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e3181959e27
760
let her suffer” that they may actually be
pointing the physician in the direction of
withholding care or treatment? It would be
interesting to reconstruct the scenarios to
evaluate whether the terms are congruent
with the wishes of the general public, as
posited in the basic assumptions in the
research design of Escher et al.
With that said, the proposition that older
physicians and those with greater knowledge
in ethics responded in a manner more congruent with family wishes is intriguing. Do
the results point more toward the concept of
professional-transcendence? Transcendence
is defined as, the ability to expand selfboundaries intrapersonally (through increased awareness of one’s beliefs and values),
interpersonally (by reaching out to others),
and temporally (integrating the past and future into the present), and may be measured
by valid and reliable tools (3). Transcendence
is associated with experiences that increase
awareness of human vulnerability and mortality (4). Do those physicians with more life
experience, who have witnessed the suffering
of others over longer periods of time, and are
in tune to the vulnerabilities of our existence
on earth extend beyond themselves and listen
more attentively and empathetically to others? As suggested by Weaver et al (5), ethical
sensitivity may have an effect on decision
making and is related to professional transcendence. Do physicians who are less paternalistic in their views about decision making
have greater professional transcendence? If
the study by Escher et al (2) were extended to
explore a situation where the nurse had suggested that we “spare useless suffering,”
would these same older doctors with greater
confidence in their skills related to ethics
have listened and not admitted to the intensive care unit? The question left to ponder is
whether age and experience provide us with a
greater self-perceived understanding of ethics
and a higher level of professional transcendence.
A future study could be one centered on
measuring self-transcendence in physicians
and the relationship between decisionmaking models used in the decision to admit
and/or collaborate with team members. One
question could be, “Is there a relationship
between degree of self-transcendence in physicians and use of the shared decision making
vs. paternalistic model of decision making.”
Another could be, “Does the degree of selftranscendence in physicians relate to collaboration in the workplace?”
Judy E. Davidson, DNP, RN, FCCM
Department of Advanced Practice
Nursing and Research
Scripps Mercy Hospital
San Diego, CA
Beth Palmer, DNP, RN
Veterans Administration Medical
Center
San Diego, CA
REFERENCES
1. Davidson JE, Powers K, Hedayat KM, et al:
Clinical practice guidelines for support of the
family in the patient-centered ICU. American
College of Critical Care Task Force:
2004 –2005. Crit Care Med 2007; 35:605– 622
2. Escher M, Perneger TV, Heldegger CP, et al:
Admission of incompetent patients to intensive care: Doctors’ responsiveness to family
wishes. Crit Care Med 2009; 37:528 –532
3. Reed P: Self-transcendence and mental health
in oldest-old adults. Nurs Res 1991; 40:1–7
4. Reed P: The theory of self-transcendence. In:
Middle Range Theory for Nursing. Smith M,
Liehr P (Eds), New York, Springer, 2003
5. Weaver K, Morse J, Mitcham C: Ethical sensitivity in professional practice: Concept analysis. J Adv Nurs 2008; 62:607– 618
Crit Care Med 2009 Vol. 37, No. 2
Diuresis in renal failure: Treat the patient, not the urine output*
W
hen critically ill patients
develop acute kidney injury (AKI), other than
treating the underlying
cause, there are very limited therapeutic
options that have been proven to improve
renal recovery. When we observe a drop in
urine output associated with AKI, a natural
reaction is to administer agents that will
increase the urine output, such as loop
diuretics (e.g., furosemide or bumetanide)
and dopamine. But even if these therapies
increase urine output, will they provide any
meaningful benefit to the patient in terms
of morbidity and mortality? During multidisciplinary rounds in the intensive care
unit, I frequently observe that when nurses
contact an attending physician because of a
low urine output and rising serum creatinine, an almost reflex reaction is to “give
furosemide.” This is rather analogous to
ordering aspirin for a headache without
analyzing the cause of the problem. In the
case of diuretics, if the cause of the decrease
in urine output is hypovolemia, clearly, diuretics are not an appropriate choice.
The literature regarding dopamine in
AKI is clear: although it may increase urine
output, there is no benefit in any measure
of morbidity or mortality, and it should not
be used (1–3). The data regarding the use of
loop diuretics in AKI were recently summarized by Bagshaw et al (4). In brief, although loop diuretics may have some theoretical benefits in AKI, such as flushing
debris from the kidney tubules and maintaining fluid, electrolyte, and acid– base homeostasis, clinical trials have not shown
any benefit in terms of renal recovery or
any measure of morbidity or mortality. One
observational trial actually showed an increase in mortality with loop diuretics (5).
As summarized by Bagshaw et al (4), there
are many limitations with published clini-
*See also p. 533.
Key Words: dopamine; furosemide; bumetanide;
diuretics; hemofiltration; acute kidney injury; renal replacement therapy
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194de98
Crit Care Med 2009 Vol. 37, No. 2
cal trials, such as lack of generalizability to
modern intensive care unit patients, not
including intensive care unit patients, confounding cointerventions such as dopamine and mannitol, delayed or late intervention, and bolus diuretic administration
without specific therapeutic end points. Despite lack of data showing benefit, a recent
survey suggests that the majority of physicians use diuretics in patients with AKI but
do not believe that diuretics can reduce
mortality or improve renal recovery (6).
This survey also found that the majority of
physicians would be willing to participate
in randomized clinical trials of loop diuretics in AKI. Clearly, more randomized controlled clinical trials are needed.
In this issue of Critical Care Medicine,
Van Der Voort et al (7) report the results
of a randomized, double-blind, placebocontrolled trial in the specific setting of
intensive care unit patients with AKI after
hemofiltration. The purpose was to determine whether giving furosemide after hemofiltration to increase urine output
would provide any benefit on recovery of
renal function in terms of creatinine
clearance and duration of continuous renal replacement therapy. Not surprisingly, there was no significant effect on
creatinine clearance or renal recovery despite the fact that furosemide significantly increased sodium excretion and
urine output as compared with placebo.
In fact, there was a slight trend favoring
placebo in terms of renal recovery (92%
in the furosemide group vs. 100% in the
placebo group). The limitations of the
trial are clearly stated by the authors: it
was a small trial (71 patients), the furosemide patients had a higher baseline
Sepsis-Related Organ Failure Score and
were slightly older than patients in the
placebo group, and the fact that measured creatinine clearance was used to
assess renal function, which may not accurately reflect glomerular filtration rate
in critically ill patients.
When considering the main mechanism of action of loop diuretics, the results are not surprising. Loop diuretics
inhibit sodium and water resorption from
the loop of Henle and do not improve
glomerular filtration. In fact, some data
suggest that furosemide may actually decrease glomerular filtration rate (8). Conversely, animal models suggest other
mechanisms that may be beneficial, such
as attenuation of ischemia-related gene
expression (9), but the clinical significance of this effect in humans is unknown.
Despite the limitations of the trial, the
conclusions seem clear. Routine use of
furosemide after hemofiltration does not
improve renal recovery. Despite the need
for more randomized controlled trials,
the current literature regarding loop diuretics in AKI does not support routine
use. As summarized by Venkataraman
and Kellum (10), diuretics and dopamine
are ineffective in preventing AKI or improving outcomes once AKI occurs. For
now, we need to attempt to prevent renal
failure with strategies such as adequate
hydration, maintaining an adequate
mean arterial pressure, and minimizing
nephrotoxin exposure (10).
If a patient with AKI has a clinical
indication for fluid removal such as pulmonary edema, loop diuretics may be a
reasonable therapeutic approach, but the
bottom line is to treat the patient not the
urine output.
Brad E. Cooper, PharmD, FCCM
Hamlot Medical Center
Erie, PA
REFERENCES
1. ANZICS Clinical Trials Group: Low-dose dopamine in patients with early renal dysfunction: A placebo-controlled randomized trial.
Lancet 2000; 356:2139 –2143
2. Friedrich JO, Adhikari N, Herridge MS, et al:
Meta-analysis: Low-dose dopamine increases
urine output but does not prevent renal dysfunction or death. Ann Intern Med 2005;
142:510 –524
3. Kellum JA, Decker JM: Use of dopamine in
acute renal failure: A meta-analysis. Crit
Care Med 2001; 29:1526 –1531
4. Bagshaw SM, Bellomo R, Kellum JA: Oliguria, volume overload, and loop diuretics. Crit
Care Med 2008; 36:S172–S178
5. Meta RL, Pascual MT, Soroko S, et al: Diuretics, mortality, and nonrecovery of renal function in acute renal failure. JAMA 2002; 288:
2547–2553
6. Bagshaw SM, Delaney A, Jones D, et al: Di-
761
uretics in the management of acute kidney
injury: A multinational survey. Contrib
Nephrol 2007; 156:236 –249
7. van der Voort PHJ, Boerma EC, Koopmans M,
et al: Furosemide does not improve renal recovery after hemofiltration for acute renal fail-
ure in critically ill patients: A double blind
randomized controlled trial. Crit Care Med
2009; 37: 533–538
8. Trivedi H, Dresser T, Aggarwal K: Acute effect of
furosemide on glomerular filtration rate in diastolic dysfunction. Ren Fail 2007; 29:985–989
9. Aravindan N, Shaw A: Effect of furosemide infusion on renal hemodynamics and angiogenesis gene expression in acute renal ischemia/
reperfusion. Ren Fail 2006; 28:25–35
10. Venkataraman R, Kellum JA: Prevention of
acute renal failure. Chest 2007; 131:300 –308
Prevention of pulmonary dysfunction after cardiac surgery by a
vital capacity maneuver: Is it so simple?*
R
espiratory dysfunction is a frequently encountered complication in postoperative intensive care unit (ICU) patients.
After cardiac surgery, pulmonary dysfunction is a well-known encumbering
postoperative issue. Hence, this can result in a significantly increased mortality
up to 25% and an attributable morbidity
with an importantly prolonged stay in the
ICU.
In this issue of Critical Care Medicine,
Shim et al (1) tackled pulmonary impairment after an off-pump cardiac surgery.
Respiratory failure after cardiac surgery
with cardiopulmonary bypass results in
factors leading to major pulmonary complications: atelectasis is present to a
much larger extent than induced by
solely anesthesia or sternotomy (2), stasis
of secretions, due to insufficient coughing capacity, inflammation and infection.
In conjunction with advanced age, preexistent lung disease, impaired cardiac performance, and secondary prolonged duration of mechanical ventilation (3, 4),
these risk factors intensify respiratory
failure. Furthermore, coronary bypass
surgery with and without extracorporeal
circulation results in a dramatic impairment of respiratory system mechanics
(5), not in the least hampered by an intraoperative positive fluid balance (5, 6).
All these complications result in hypoxemia and gas exchange impairment, commonly observed after cardiac surgery.
Hachenberg et al (7) demonstrated a
*See also p. 539.
Key Words: pulmonary dysfunction; cardiac surgery;
shunting; vital capacity maneuver; respiratory failure
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194dee3
762
nearly doubled intrapulmonary shunting
during and after cardiac surgery, with
subsequent augmented extravascular
lung water and further collapse (8). Although multifactorial in nature, pulmonary dysfunction seems to be mostly related to intrapulmonary right-to-left
shunt.
Several investigators applied different
techniques to eliminate or diminish the
impaired gas exchange after cardiac surgery. Recruitment maneuvers, such as continuous positive airway pressure, bilevel
positive airway pressure, or vital capacity
maneuver (VCM), at the end of the cardiopulmonary bypass have been used, to alleviate atelectasis and shunting.
Continuous positive airway pressure
with varying levels of airway pressure has
lead to conflicting results: either decreased or improved lung function was
shown, although moderate continuous
positive airway pressure appeared to result in the optimalization of postbypass
pulmonary dysfunction (9). Bilevel positive airway pressure was observed to be
beneficial in general anesthesia to prevent ventilation-perfusion mismatch and
to improve oxygenation (10).
With VCM, the lungs are inflated to a
peak airway pressure of 40 cm H2O for 15
seconds. With respect to on-pump cardiac
surgery, several investigators clearly demonstrated a beneficial effect of VCM on
postoperative extubation times. However,
the influence on pulmonary gas exchange
in the ICU remains controversial. Magnusson et al (11) assessed the effects of VCM to
prevent atelectasis after cardiopulmonary
bypass in a study on pigs. Control pigs
developed extensive atelectasis, whereas
VCM-treated pigs showed a significantly
smaller proportion of atelectasis, the least
in those animals ventilated with lower oxygen concentrations. Repetition within 6
hours seemed to provide no additional ad-
vantages based on both laboratory and
computed tomographic data (12).
In a human study, Murphy et al (13) could
not demonstrate any improvement of gas exchange in the ICU after an on-pump cardiac
surgery between control and VCM-supported
patients, whereas the latter were extubated
significantly earlier, after 6.5 ⫾ 2.1 hours vs.
9.4 ⫾ 4.2 hours. In a subset of cardiac surgical patients with cardiopulmonary bypass,
Tschernko et al (14) also showed a reduction
of intrapulmonary shunting using VCM after
termination of cardiopulmonary bypass, although shunting increased considerably after
extubation. Finally, Minkovich et al (15) performed VCM twice: both before cessation of
cardiopulmonary bypass (35 cm H2O—15
seconds) and after admission in the ICU (30
cm H2O—5 seconds). In contrast to the data
on animals presented by Magnusson et al
(11), VCM resulted in an improved and prolonged arterial oxygenation for 24 hours in
patients. No data, however, were provided
concerning the timing of extubation.
With respect to off-pump cardiac surgery, less information is available. In the
previously referred study, Tschernko et al
(14) could not show any effect of VCM on
the duration of mechanical ventilation
between patients operated on bypass
(control), those supported with VCM, and
off-pump cardiac surgery patients. In addition, they showed an increased shunting after extubation, although much less
than in those patients with cardiopulmonary bypass and with direct impact on
length of stay in the ICU and in the hospital, compared with control and onpump cardiac surgical patients.
The findings of Shim et al (1) now
clearly demonstrate that VCM leads to
earlier extubation in off-pump cardiac
surgery patients albeit without demonstrable effects on respiratory mechanics,
length of stay in the ICU nor in the hospital. Although strict extubation criteria
Crit Care Med 2009 Vol. 37, No. 2
were followed, extubation remains a
rather arbitrary factor. Many interfering
issues, as listed earlier, interfere, not in
the least an appropriate sensorium. In
this study, patients who were not ready
for extubation were sedated with midazolam. At least duration and level of sedation can be a major point of discussion.
Also, the value of VCM in this particular study could be questioned as there
was no need at all to increase FIO2
throughout the study, even in non-VCMsupported patients, in whom shunting
and atelectasis could have been expected
throughout a 4-hour-lasting procedure.
Another point of discussion is the timing
of VCM: Shim et al performed the VCM
immediately after sternotomy. As anesthesia may help the development of atelectasis, could there be a supplementary
beneficial effect of a VCM before closure
of the sternum? Finally, in none of the
patients was respiratory infection a cause
for prolonged mechanical ventilation, although the importance of this issue cannot be denied (16, 17). From larger studies, it could be expected that ⫾20% of
cardiac surgical patients are prone to tracheobronchial or pulmonary infection,
associated with prolonged stay in the hospital, multiple organ dysfunction, and increased hospital mortality (16, 18).
With respect to prevention of atelectasis and decreased intrapulmonary
shunting after cardiac surgery, the last
word has not been said. Altering the gas
mixture from FIO2 1.0 to 30% oxygen in
nitrogen during induction of anesthesia
could avoid early atelectasis and shunting (19). In view of the multifactorial
nature of this problem, further investigations are needed to ascertain a simple
approach of pulmonary dysfunction after cardiac surgery.
Jan Poelaert, MD, PhD
Department of Anesthesiology
and Perioperative Medicine
Crit Care Med 2009 Vol. 37, No. 2
Acute and Chronic Paintherapy
University Hospital of the Flemish
Free University of Brussels
Brussels, Belgium
Carl Roosens, MD
Department of Intensive Care
Medicine
Ghent University Hospital
Ghent, Belgium
10.
11.
REFERENCES
1. Shim JK, Chun DH, Choi YS, et al: Effect of
early vital capacity maneuver on respiratory
variables during multivessel off-pump coronary artery bypass graft surgery. Crit Care
Med 2009; 37:539 –544
2. Magnusson L, Zemgulis V, Wicky S, et al:
Atelectasis is a major cause of hypoxemia and
shunt after cardiopulmonary bypass: An experimental study. Anesthesiology 1997; 87:
1153–1163
3. Nozawa E, Azeka E, Ignez ZM, et al: Factors
associated with failure of weaning from longterm mechanical ventilation after cardiac
surgery. Int Heart J 2005; 46:819 – 831
4. Paulus S, Lehot JJ, Bastien O, et al: Enoximone and acute left ventricular failure during weaning from mechanical ventilation after cardiac surgery. Crit Care Med 1994; 22:
74 – 80
5. Roosens C, Heerman J, De Somer F, et al:
Effects of off-pump coronary surgery on the
mechanics of the respiratory system, lung,
and chest wall: Comparison with extracorporeal circulation. Crit Care Med 2002; 30:
2430 –2437
6. Ranieri VM, Vitale N, Grasso S, et al: Timecourse of impairment of respiratory mechanics after cardiac surgery and cardiopulmonary bypass. Crit Care Med 1999; 27:
1454 –1460
7. Hachenberg T, Tenling A, Nystrom SO, et al:
Ventilation-perfusion inequality in patients
undergoing cardiac surgery. Anesthesiology
1994; 80:509 –519
8. Hachenberg T, Tenling A, Rothen HU, et al:
Thoracic intravascular and extravascular
fluid volumes in cardiac surgical patients.
Anesthesiology 1993; 79:976 –984
9. Boldt J, King D, Scheld HH, et al: Lung
12.
13.
14.
15.
16.
17.
18.
19.
management during cardiopulmonary bypass: Influence on extravascular lung water.
J Cardiothorac Anesth 1990; 4:73–79
Yu G, Yang K, Baker AB, et al: The effect of
bi-level positive airway pressure mechanical
ventilation on gas exchange during general
anaesthesia. Br J Anaesth 2006; 96:522–532
Magnusson L, Zemgulis V, Tenling A, et al:
Use of a vital capacity maneuver to prevent
atelectasis after cardiopulmonary bypass: An
experimental study. Anesthesiology 1998; 88:
134 –142
Magnusson L, Wicky S, Tyden H, et al: Repeated vital capacity maneuvers after cardiopulmonary bypass: Effects on lung function
in a pig model. Br J Anaesth 1998; 80:
682– 684
Murphy GS, Szokol JW, Curran RD, et al:
Influence of a vital capacity maneuver on
pulmonary gas exchange after cardiopulmonary bypass. J Cardiothorac Vasc Anesth
2001; 15:336 –340
Tschernko EM, Bambazek A, Wisser W, et al:
Intrapulmonary shunt after cardiopulmonary bypass: The use of vital capacity maneuvers versus off-pump coronary artery bypass
grafting. J Thorac Cardiovasc Surg 2002;
124:732–738
Minkovich L, Djaiani G, Katznelson R, et al:
Effects of alveolar recruitment on arterial
oxygenation in patients after cardiac surgery:
A prospective, randomized, controlled clinical trial. J Cardiothorac Vasc Anesth 2007;
21:375–378
Kollef MH, Sharpless L, Vlasnik J, et al: The
impact of nosocomial infections on patient
outcomes following cardiac surgery. Chest
1997; 112:666 – 675
Kollef MH, Skubas NJ, Sundt TM: A randomized clinical trial of continuous aspiration of
subglottic secretions in cardiac surgery patients. Chest 1999; 116:1339 –1346
Bouza E, Perez A, Munoz P, et al: Ventilatorassociated pneumonia after heart surgery: A prospective analysis and the value of surveillance.
Crit Care Med 2003; 31:1964–1970
Rothen HU, Sporre B, Engberg G, et al: Atelectasis and pulmonary shunting during induction of general anaesthesia—Can they be
avoided? Acta Anaesthesiol Scand 1996; 40:
524 –529
763
And the winner is: Regional citrate anticoagulation*
A
cute kidney injury requiring
renal replacement therapy
(RRT) is an independent mortality risk factor in intensive
care patients. Most likely, the unwanted
consequences of both acute kidney injury
and RRT contribute to this increased
mortality. Continuous RRT is often the
procedure of choice in these patients.
One requirement for continuous RRT
having potential deleterious consequences is systemic anticoagulation. Although anticoagulation exposes patients
to the risk of bleeding, its absence may
result in clotting of the circuit and less
effective treatment. Worldwide, systemic
unfractionated heparin is the most common anticoagulant (1). In the last two
decades, various methods of regional anticoagulation using citrate (RCA) have
been described (2, 3); however, its use
worldwide is limited. Several factors are
responsible for this, including concerns
about the safety of RCA, the metabolic
complexity, the costs and logistics of obtaining custom-made solutions, and the
lack of uniformity in therapeutic approaches.
Citrate causes anticoagulation in the
extracorporeal circuit by chelating ionized calcium (iCa). The citrate– calcium
complex is partly filtered and partly diluted by the total blood volume. In the
systemic circulation, citrate is rapidly
metabolized via the Krebs cycle in the
liver and other tissues yielding bicarbonate and releasing iCa. The extracorporeal
loss of calcium is substituted. Citrate
possibly also inhibits the calciummediated activation of inflammatory
cells in the extracorporeal circulation
(4, 5). A potential safety risk with RCA
is the occurrence of metabolic disturbances, entailing the frequent monitoring of acid– base status, plasma electro-
*See also p. 545.
Key Words: citrate; heparin; acute kidney injury;
continuous renal replacement therapy
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194df2e
764
lytes, and calcium levels. The infusion
of hypertonic trisodium citrate may
lead to hypernatremia and/or alkalosis,
unless adapted replacement solutions
are used. Citrate toxicity may occur in
patients insufficiently metabolizing citrate (e.g., severe liver failure, decreased muscle perfusion). Systemic citrate levels are not routinely measured
in clinical practice, but accumulation is
easily detected by an increased total
calcium concentration/iCa ratio (6).
In this edition of Critical Care Medicine,
Oudemans-van Straaten et al compare the
safety and efficacy of RCA with the systemic
low-molecular weight heparin nadroparin
(7). To date, this is the largest randomized
controlled trial on RCA. Patients with acute
kidney injury requiring RRT and no contraindications for systemic nadroparin
and/or RCA were included. Of the 215 randomized patients, 200 patients received
continuous veno-venous hemofiltration
(CVVH) per protocol (97 RCA and 103 nadroparin). The study was powered on adverse events necessitating discontinuation
of study medication. Primary outcomes
were safety and efficacy. Discontinuation
was required in two patients of the RCA
group (citrate accumulation, clotting) and
in 20 patients in the nadroparin group
(bleeding, thrombocypenia) (p ⬍ 0.001).
Unfortunately, anti-Xa levels were not measured and this might have reduced bleeding. Nevertheless, the differences in bleeding and transfusion were not statistically
significant between the groups, possibly because nadroparin was discontinued when
bleeding occurred, and patients with a high
risk of bleeding were excluded. Circuit survival was not significantly different between
groups. These results contrast with the
findings of previous randomized controlled
trials (Table 1). Although previous studies
were small and underpowered, some differences among studies are noteworthy. The
present study infused fixed doses of nadroparin and citrate, whereas previous
studies titrated unfractionated heparin and
citrate to achieve a doubling of the activated partial thromboplastin time and a
circuit iCa of ⬍0.35 mmol/L. Routine circuit disconnection affects circuit survival.
Circuits were routinely disconnected after
72 hours in the studies by Oudemans-van
Straaten et al and Betjes et al (8), but not in
the studies by Monchi et al (9) and Kutsogiannis et al (10). Notably, in the present
study RCA did not result in serious metabolic disturbances confirming previous
findings (2, 3, 8 –12). In the present study,
RCA even resulted in less metabolic alkalosis (p ⫽ 0.001) compared with nadroparin;
however, the RCA protocol aimed at normal pH using two replacement fluids, and
this was not the case in the nadroparin
group.
A remarkable finding in the present
study is the survival benefit of the RCA
group. Three-month mortality on intention-to-treat was 48% (RCA) and 63% (nadroparin) (p ⫽ 0.03). Three-month mortality per protocol was 45% (RCA) and 62%
(nadroparin) (p ⫽ 0.02). There were no
significant differences in CVVH-timing and
CVVH-dose between groups. Post hoc analysis showed that RCA was particularly beneficial in surgical patients, patients with
sepsis, patients with severe organ failure,
and relatively younger patients. As suggested by the authors, one can hypothesize
that RCA is favorable, nadroparin unfavorable, or both. Although the patients in the
nadroparin group tended to bleed more,
the differences in bleeding and transfusion
between groups were not statistically significant and could not explain the RCA survival benefit. The incidence of metabolic
alkalosis was higher in the nadroparin
group. Alkalemia has hemodynamic consequences as a result of direct effect and by
means of reducing iCa. In the present
study, however, iCa was lower in the RCA
group. Furthermore, metabolic alkalosis
during CVVH was not related to mortality
at univariate analysis, supporting the recent findings of Demirjian et al (13). The
authors speculate that this reduction in
mortality is from RCA blocking inflammation. This is certainly a plausible explanation based on the results of previous studies
on patients undergoing chronic dialysis (4,
5, 14). These studies demonstrated that dialysis-induced polymorphonuclear cell degranulation is primarily iCa dependent and
is abolished during RCA dialysis (4, 5). In
addition, Gabutti et al (14) showed that
RCA dialysis had a favorable effect on interCrit Care Med 2009 Vol. 37, No. 2
Table 1. Randomized controlled trials comparing the safety and efficacy of regional citrate anticoagulation and systemic heparins
Author
Study Design
(Number of
Patients)a
Anticoagulation
Monitoring
CRRT
Modality
Filter
Oudemans-van
RCA 97, LMWH Fixed dose LMWH Postdilution CTA 1.9 m2
Straaten et al (7)
103
and RCA
CVVH
Monchi et al (9)
Betjes et al (8)
Kutsogiannis
et al (10)
Cross-over
Circuit iCa ⬍0.3
Postdilution PS 1.9 m2
RCA-UFH 8,
mmo/L, APTT
CVVH
UFH-RCA 12
60–80 sec
RCA 21, UFH
Circuit iCa 0.25– Postdilution CTA 1.9 m2
27
0.3 mmo/L,
CVVH
APTT 50–70
sec
RCA 16, UFH
Circuit iCa 0.25– Predilution AN 69 1.0 m2
14
35 mmo/L, PTT
CVVHDF
45–65 sec
QB
(mL/min)
QUF
(mL/hr)
Citrate
(mmol/L
QB)
220
2000–4000
3
150
35 mL/kg/hr
4.3
150
1500
125
1000, QD
1000
mL/hr
3
3.3
Median
Circuit
Survival (hr)
Major
Bleeding
Events
Units of PRC
per Day of
CRRT
RCA 27,
RCA 6,
RCA 0.27,
LMWH 26
LMWH 16
LMWH 0.36
(p ⫽ 0.68)b
(p ⫽ 0.08)
(p ⫽ 0.31)
RCA 70,
RCA 0,
RCA 0.2,
UFH 40
UFH 1
UFH 1
(p ⫽ 0.007)
(p ⫽ NR)
(p ⫽ 0.0008)
RCA 36,
RCA 0,
RCA 0.43,
UFH 38.4
UFH 10
UFH 0.88
(p ⫽ NS)b
(p ⬍ 0.01)
(p ⫽ 0.01)
RCA 124.5,
RCA 0,
UFH 38.3
UFH 7
(p ⫽ 0.001)
(p ⫽ NR)
RCA 0.17,
UFH 0.33
(p ⫽ 0.13)
RCA, regional citrate anticoagulation; LMWH, low molecular weight heparin; UFH, unfractionated heparin; iCa, ionized calcium; (A)PTT, (activated)
partial thromboplastin time; CVVH, continuous veno-venous hemofiltration; CVVHDF, continuous veno-venous hemodiafiltration; CTA, cellulose triacetate; PS, polysulfone; AN69, polyacrilonitrile; QB, blood flow; QUF, ultrafiltrate flow; QD, dialysate flow; NS, not significant; NR, not reported; PRC, packed
red cell; CRRT, continuous renal replacement therapy.
a
Inclusion criteria, patients without contraindications for citrate or systemic heparin; bRoutine circuit disconnection after 72 hrs.
leukin-1 release. Unfortunately, the
present study does not provide data concerning biocompatibility and inflammatory
markers to support the theory.
In summary, the study of Oudemansvan Straaten et al provides convincing evidence that RCA is as effective as, yet safer
than nadroparin. The study has some limitations, including being a single-center
study and using low-molecular weight heparin instead of unfractionated heparin. Furthermore, it is clear that the study was
designed to evaluate the safety and efficacy
of RCA and not survival. Another large
Dutch multicenter study is underway, randomizing patients into CVVH with RCA or
unfractionated heparin (www.clinicaltrials.
gov/ct2/show/NCT00209378). Primary outcome measures of this study are mortality,
circuit survival, and bleeding complications, whereas laboratory markers of inflammation, endothelial dysfunction, and
coagulation parameters are secondary end
points. If this upcoming study confirms the
results of Oudemans-van Straaten et al,
RCA will definitely be the number one anticoagulation for continuous RRT.
Catherine S. C. Bouman, MD, PhD
Department of Intensive Care
Academic Medical Center
University of Amsterdam
Amsterdam, The Netherlands
Crit Care Med 2009 Vol. 37, No. 2
REFERENCES
1. Uchino S, Bellomo R, Morimatsu H, et al:
Continuous renal replacement therapy: A
worldwide practice survey. The beginning
and ending supportive therapy for the kidney
(B.E.S.T. kidney) investigators. Intensive
Care Med 2007; 33:1563–1570
2. Egi M, Naka T, Bellomo R, et al: A comparison of two citrate anticoagulation regimens
for continuous veno-venous hemofiltration.
Int J Artif Organs 2005; 28:1211–1218
3. Tolwani AJ, Prendergast MB, Speer RR, et al:
A practical citrate anticoagulation continuous venovenous hemodiafiltration protocol
for metabolic control and high solute clearance. Clin J Am Soc Nephrol 2006; 1:79 – 87
4. Bohler J, Schollmeyer P, Dressel B, et al:
Reduction of granulocyte activation during
hemodialysis with regional citrate anticoagulation: Dissociation of complement activation and neutropenia from neutrophil degranulation. J Am Soc Nephrol 1996;
7:234 –241
5. Bos JC, Grooteman MP, van Houte AJ, et al:
Low polymorphonuclear cell degranulation
during citrate anticoagulation: A comparison
between citrate and heparin dialysis. Nephrol
Dial Transplant 1997; 12:1387–1393
6. Hetzel GR, Taskaya G, Sucker C, et al: Citrate
plasma levels in patients under regional anticoagulation in continuous venovenous hemofiltration. Am J Kidney Dis 2006; 48:
806 – 811
7. Oudemans-van Straaten HM, Bosman RJ,
Koopmans M, et al: Citrate anticoagulation
for continuous venovenous hemofiltration.
Crit Care Med 2009; 37:545–552
8. Betjes MG, van Oosterom D, van Agteren M,
et al: Regional citrate versus heparin anticoagulation during venovenous hemofiltration
in patients at low risk for bleeding: Similar
hemofilter survival but significantly less
bleeding. J Nephrol 2007; 20:602– 608
9. Monchi M, Berghmans D, Ledoux D, et al:
Citrate vs. heparin for anticoagulation in
continuous venovenous hemofiltration: A
prospective randomized study. Intensive
Care Med 2004; 30:260 –265
10. Kutsogiannis DJ, Gibney RT, Stollery D, et al:
Regional citrate versus systemic heparin anticoagulation for continuous renal replacement in critically ill patients. Kidney Int
2005; 67:2361–2367
11. Bagshaw SM, Laupland KB, Boiteau PJ, et
al: Is regional citrate superior to systemic
heparin anticoagulation for continuous renal replacement therapy? A prospective observational study in an adult regional critical care system. J Crit Care 2005; 20:
155–161
12. Mehta RL, McDonald BR, Aguilar MM, et al:
Regional citrate anticoagulation for continuous arteriovenous hemodialysis in critically
ill patients. Kidney Int 1990; 38:976 –981
13. Demirjian S, Teo BW, Paganini EP: Alkalemia during continuous renal replacement
therapy and mortality in critically ill patients. Crit Care Med 2008; 36:1513–1517
14. Gabutti L, Ferrari N, Mombelli G, et al: The
favorable effect of regional citrate anticoagulation on interleukin-1beta release is dissociated from both coagulation and complement activation. J Nephrol 2004; 17:
819 – 825
765
New biomarkers of acute kidney injury: Promise for the future but
beware the lure of novelty*
A
t last, a much needed consensus definition of acute kidney
injury (AKI) has emerged,
which should not only facilitate comparative research and audit but
also recognize the adverse impact of even
modest disease (1). However, its reliance
on rises in serum creatinine—the traditional biomarker of renal dysfunction—
does carry the risk of missed therapeutic
opportunity because of the time lag between the initiating insult and the diagnostic elevation (2). The interpretation of
changes in serum creatinine is also complicated by factors such as diet, muscle
mass, and the use of certain drugs and
supplements, as well as by the presence of
prerenal dysfunction when no cellular injury has actually occurred. It is these
limitations that have stimulated an ongoing and intensive evaluation of a variety
of alternative early biomarkers of AKI.
When examining the evolving literature, it is crucial to bear in mind what the
potential candidate biomarker is actually
reflecting; cystatin C, for instance, is a
measure of renal functional status (a
“quick creatinine”) whereas others, such
as neutrophil gelatinase–associated lipocalin (NGAL) and urinary interleukin18, are products of the pathophysiologic
processes that underlie AKI (3) and indicate active renal damage (a “troponin of
the kidney”).
In human studies, cystatin C can predict the development of AKI (4) and the
requirement for renal replacement therapy (5), although its superiority over serum creatinine has not been a universal
finding (6). Serum and/or urine NGAL
levels have been shown to be accurate
predictors of AKI in settings as diverse as
*See also p. 553.
Key Words: acute renal failure; acute kidney injury;
neutrophil gelatinase–associated lipocalin; cystatin; interleukin 18; biomarkers; creatinine
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194dfd0
766
percutaneous coronary intervention (7),
pediatric (8) and adult (9) cardiac surgery, and septic (10) and nonseptic (11)
critically ill children. Rising urinary interleukin-18 levels have proved similarly predictive in a nonseptic critically
ill pediatric population (12), in critically ill patients with acute respiratory
distress syndrome (13), and after adult
and pediatric cardiac surgery (14). As
well as predicting AKI, there may be an
additional prognostic role for novel biomarkers (12, 13).
The article by Haase-Fielitz et al (15),
published in this issue of Critical Care
Medicine, adds to the existing literature.
They are the first to study the early use of
serum biomarkers in adult cardiac surgical patients (urine NGAL having already
been studied in this setting [9]). Compared to the intensive care unit admission creatinine, the contemporaneous serum NGAL and serum cystatin C were
found to have good predictive value for
the subsequent development of AKI. The
accuracy of cystatin C diminished after
patients with preexisting renal impairment were excluded from analysis, suggesting that it did not only indicate evolving AKI but was also an independent risk
factor for it, being a reflection of the
strong, predisposing effects of chronic renal dysfunction. Beyond AKI, both serum
NGAL and cystatin C carried excellent
prognostic value for the composite outcome of renal replacement therapy or
hospital mortality.
Taken in the context of the current
literature, the study has added further
evidence of potential value of the novel
biomarkers of AKI but its weaknesses
should be borne in mind: it was small
(only 23 of 100 subjects developed AKI),
confidence intervals of the area under the
receiver operating characteristic curves
were wide (in part, a reflection of the
former), and an absolute intensive care
unit admission serum creatinine was
used (a more valid comparison might
have been a ⌬ creatinine that would have
helped correct for the confounding effects of preexisting abnormalities).
What seems to be evident from the
literature as a whole is that no one biomarker fulfils all the desirable features
of early detection of active renal damage,
of rapid reflection of changes in renal
function, of risk stratification, or of differential diagnosis. It is entirely possible
that different panels of biomarkers will be
required to meet different needs (16). Exploration of this is currently the focus on
significant, multicentered effort.
If novel biomarkers of AKI are to be of
potential value, can we speculate about
how they might be utilized?
They have clear roles as research tools.
An early diagnosis of AKI may allow
timely experimental intervention—a major deficiency of previous human studies
(17) in which the initiating insult may
have been long over by the time AKI
was actually diagnosed. Close linkage
between the index insult and time of
diagnosis may be particularly important
if other, confounding insults are likely,
as is clearly the case in the critical care
setting.
Beyond research, the actual clinical
utility of these early biomarkers remains
untested with a key question being
whether they would add anything to
management beyond the information
provided using conventional, creatininebased diagnosis.
One area of possible use is to identify
groups of patients who have undergone a
discrete renal “hit” and who require subsequent, augmented monitoring. Highrisk outpatients undergoing contrastenhanced radiography might fall into this
category but would require a threshold
biomarker level of sufficient negative predictive value to definitively exclude evolving AKI.
Conversely, the utility of biomarkers
as a less targeted, sequential screening
tool, unlinked to a specific renal event, is
less clear although the results of largescale population studies are outstanding
and definitive therapeutic interventions
Crit Care Med 2009 Vol. 37, No. 2
are lacking. Although a case could be
made that early detection of AKI could
encourage the avoidance of harm (e.g.,
aminoglycoside antibiotics or premature
intensive care unit step down), this approach of risk stratification is also, as yet,
untested in the clinical environment.
In summary, it is increasingly evident
that a variety of novel biomarkers are
predictive of overt AKI in a range of different settings. Although holding promise for the future, the lure of their novelty
does not remove the need for a clear and
guarded appraisal of their potential.
Nigel S. Kanagasundaram, MD,
FRCP (UK)
Department of Renal Medicine
Newcastle upon Tyne
Hospitals
NHS Foundation Trust
Newcastle Upon Tyne, United
Kingdom
REFERENCES
1. Mehta RL, Kellum JA, Shah SV, et al: Acute
Kidney Injury Network: Report of an initiative to improve outcomes in acute kidney
injury. Crit Care 2007; 11:R31
2. Bagshaw SM, Gibney N: Conventional markers of kidney function. Crit Care Med 2008;
36(Suppl 4):S152–S158
3. Goligorsky MS: Whispers and shouts in the
pathogenesis of acute renal ischaemia. Nephrol Dial Transplant 2005; 20:261–266
4. Herget-Rosenthal S, Marggraf G, Husing J, et
al: Early detection of acute renal failure by
serum cystatin C. Kidney Int 2004; 66:
1115–1122
5. Herget-Rosenthal S, Poppen D, Husing J, et
al: Prognostic value of tubular proteinuria
and enzymuria in nonoliguric acute tubular
necrosis. Clin Chem 2004; 50:552–558
6. Ahlstrom A, Tallgren M, Peltonen S, et al:
Evolution and predictive power of serum cystatin C in acute renal failure. Clin Nephrol
2004; 62:344 –350
7. Bachorzewska-Gajewska H, Malyszko J, Sitniewska E, et al: Neutrophil-gelatinaseassociated lipocalin and renal function after
percutaneous coronary interventions. Am J
Nephrol 2006; 26:287–292
8. Mishra J, Dent C, Tarabishi R, et al: Neutrophil gelatinase-associated lipocalin (NGAL)
as a biomarker for acute renal injury after
cardiac surgery. Lancet 2005; 365:
1231–1238
9. Wagener G, Jan M, Kim M, et al: Association
between increases in urinary neutrophil gelatinase-associated lipocalin and acute renal
dysfunction after adult cardiac surgery. Anesthesiology 2006; 105:485– 491
10. Wheeler DS, Devarajan P, Ma Q, et al: Serum
neutrophil gelatinase-associated lipocalin
(NGAL) as a marker of acute kidney injury in
11.
12.
13.
14.
15.
16.
17.
critically ill children with septic shock. Crit
Care Med 2008; 36:1297–1303
Zappitelli M, Washburn KK, Arikan AA, et
al: Urine neutrophil gelatinase-associated
lipocalin is an early marker of acute kidney
injury in critically ill children: A prospective cohort study. Crit Care 2007; 11:R84
Washburn KK, Zappitelli M, Arikan AA, et
al: Urinary interleukin-18 is an acute kidney injury biomarker in critically ill children. Nephrol Dial Transplant 2008; 23:
566 –572
Parikh CR, Abraham E, Ancukiewicz M, et al:
Urine IL-18 is an early diagnostic marker for
acute kidney injury and predicts mortality in
the intensive care unit. J Am Soc Nephrol
2005; 16:3046 –3052
Parikh CR, Mishra J, Thiessen-Philbrook
H, et al: Urinary IL-18 is an early predictive
biomarker of acute kidney injury after
cardiac surgery. Kidney Int 2006; 70:
199 –203
Haase-Fielitz A, Bellomo R, Devarajan P, et
al: Novel and conventional serum biomarkers predicting acute kidney injury in adult
cardiac surgery—A prospective cohort study.
Crit Care Med 2009; 37:553–560
Parikh CR, Devarajan P: New biomarkers of
acute kidney injury. Crit Care Med 2008;
36(Suppl 4):S159 –S165
Chertow GM: On the design and analysis of multicentre trials in acute renal failure. Am J Kidney
Dis 1997; 30(Suppl 4):S96–S101
Improving function following cardiopulmonary bypass in children:
Digging deeper than steroids*
I
n the United States alone, approximately 400,000 cardiac surgical
operations are performed using
cardiopulmonary bypass (CPB)
each year, of which approximately 20,000
are performed on children. This volume
of pediatric cardiac surgical procedures
is, partly, because of the fact that
progress in corrective and palliative surgery for congenital heart disease means
that most lesions are now operable. Although overall perioperative mortality
*See also p. 577.
Key Words: cardiopulmonary bypass; inflammation; ischemia; reperfusion
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194b302
Crit Care Med 2009 Vol. 37, No. 2
has declined considerably in the past two
decades, short- and long-term morbidity
remains a significant healthcare burden.
Mortality and morbidity following cardiac surgery is multifactorial, including
changes in the inflammatory response,
disordered hemostasis, reduced cardiac
output, and multiorgan dysfunction.
Most corrective procedures require the
use of CPB, which invariably activates the
inflammatory system. Frequently, this
leads to the development of a systemic
inflammatory response syndrome, which
is characterized by alterations in cardiopulmonary function, coagulopathy, and
multiorgan system dysfunction/failure
(1). Although cardiac surgery may be
technically successful, children are at
risk of harm both in terms of increased
risk of death and of long-term disability,
as a consequence of these CPB-related
dysfunctions. This response is predictably
more severe in neonates and small infants because of the relatively high exposure of the child’s blood (small volume)
to the relatively large volume of the cardiac bypass circuit.
The etiology of the inflammatory response is thought to include activation of
blood components by the bypass apparatus, ischemia with subsequent reperfusion injury, and endothelial damage. This
leads to reversible contractile dysfunction
and impairment to flow at the microvascular level secondary to neutrophil plugging and vasoconstriction. Once inflammation is initiated in response to CPB, it
is maintained and amplified by cytokine
production. Clinical indicators of organ
injury and dysfunction, and worse clinical outcome, are associated with increased levels of certain cytokines (inter767
leukin [IL]-6, IL-8) following CPB. The
pattern and magnitude of elevations have
been routinely associated with post-CPB
morbidity in both animal models and human investigations (2).
Nuclear factor-(NF-B) is a ubiquitous
inducible transcription factor involved in
the regulation of transcription of many
proinflammatory genes. It is activated by
stimuli such as IL-1, tumor necrosis factor-␣, and oxygen-free radicals (3). Normally, NF-B is bound to the inhibitory
protein of NF-B (inhibitory kappa-B alpha [IkBa]) (4). Triggered by ischemia
and reperfusion, and proinflammatory
cytokines themselves, phosphorylation of
IkBa and p38 MAPK promotes gene expression of potential myocardial damaging mediators such as tumor necrosis factor-␣, IL-, and IL-6 (5). NF-B
translocates to the nucleus where, binding to DNA, it is able to induce the expression of several inflammatory mediators. This pathway plays an important
role in both the inflammatory response
triggered by CPB and the ischemia and
reperfusion associated with circulatory
arrest. In fact, the proposed mechanisms for the local anti-inflammatory
glucocorticoid’s actions inhibition of
IkBa degradation resulting in decreased
NF-B activation and inhibition of p38
MAPK through induction of the mitogen-activated protein kinase phosphatase-1 (6).
Understanding the triggers, timing,
and pattern of the complex inflammatory
cascade associated with CPB is essential
for modifying or arresting it. Unlike other
triggers associated with whole-body inflammatory reactions such as trauma or
sepsis, cardiac surgical teams have the
advantage of knowing when the trigger
will occur (i.e., during CPB), and hence
have an opportunity for preemptive intervention in an effort to attenuate or minimize the response. Current strategies involve modulation of the inflammatory
response and include the use of corticosteroids (7, 8), coated circuits, (9), and
aprotinin (10). These interventions have
thus far focused on large-scale interference with inflammation rather than a
specific, tailored therapy. Additionally,
specific data examining the efficacy of
these therapies in various age populations are not available, leaving open the
possibility of tailored therapies based on
age. Although steroids have gained widespread use in this setting in pediatric
cardiac surgery (11), they have potential
detrimental effects on neonates (12).
768
In this issue of Critical Care Medicine,
Duffy et al (13) examine NF-B and its
role in the inflammatory response associated with CPB. Their hypothesis is that
partial inhibition of NF-B can alleviate
cardiopulmonary dysfunction associated
with ischemia and reperfusion injury
CPB. The article describes the effects of
SN50, an inhibitor of NF-B translocation on hemodynamic and pulmonary
function. Specifically, SN50 was administered 1 hour before CPB with subsequent
measurement of NF-B activity, endothelin-1, troponin I degradation, and cardiopulmonary function. Their results indicate that the administration of SN50
decreased NF-B activity levels in nuclear
extracts from left venticular myocardium. Additionally, SN50 treatment
maintained IkBa protein at higher levels
and lower total troponin I degradation
than in untreated animals. This resulted
in preservation of myocardial contractile
function and prevented the increase in
pulmonary vascular resistance.
Their findings are consistent with previous reports that NF-B inhibition protects the myocardium from ischemic injury. By inhibition of NF-B with the
compound curcumin, Yeh et al (14) ameliorated the surge of proinflammatory cytokines during CPB and decreased the
occurrence of cardiomyocyte apoptosis
after global cardiac ischemia and reperfusion. This previous report, however, did
not measure a clinical effect. Additionally, work by Schwartz et al (15) examined the relationship among glucocorticoid administration, decreased NF-B
activity in the heart, and improved cardiopulmonary function. Finally, inhibition of NF-B has been demonstrated to
play a role in injury following circulatory
arrest (16).
The findings of Duffy et al are important in two fundamental aspects. First,
they give insight into a potential mechanism of action of corticosteroid administration in the modulation of CPBassociated inflammation. As previously
stated, the local anti-inflammatory action
of glucocorticosteroids maybe, partly,
due to the NF-B pathway (6). Additionally, Duffy et al have proposed the development of a clinically useful, more specific agent that avoids the potential
negative effects found with steroids. Because administration of corticosteroids
during CPB remains controversial, the
goal of therapies targeted at the beneficial
aspects of the mechanism of action of
steroids would be welcome.
Those of us who work in the field of
congenital heart disease and cardiac critical care medicine have a unique opportunity to modify or arrest an inflammatory process that contributes to the
morbidity and mortality of children undergoing CPB and cardiac surgery. With
clinical investigations, such as the study
carried out by Duffy et al, the therapeutic
strategies available to treat the postbypass inflammatory response will increase, potentially leading to improved
outcomes. As the authors themselves
state in their conclusion, targeting a specific activity level for NF-B would be
clinically challenging; yet, it may provide
a mechanism to move beyond steroid adminstration.
Paul A. Checchia, MD, FCCM
St. Louis Children’s Hospital
and Washington University
School of Medicine
St. Louis, MO
Ronald A. Bronicki, MD
Children’s Hospital of Orange
County, California
Orange, CA
REFERENCES
1. Kirklin JK, Westaby S, Blackstone EH, et al:
Complement and the damaging effects of
cardiopulmonary bypass. J Thorac Cardiovasc Surg 1983; 86:845– 857
2. Holmes JHT, Connolly NC, Paull DL, et al:
Magnitude of the inflammatory response to
cardiopulmonary bypass and its relation to
adverse clinical outcomes. Inflamm Res
2002; 51:579 –586
3. Christman JW, Lancaster LH, Blackwell TS:
Nuclear factor kappa B: A pivotal role in the
systemic inflammatory response syndrome
and new target for therapy. Intensive Care
Med 1998; 24:1131–1138
4. Baldwin AS Jr: The NF-kappa B and I kappa B
proteins: New discoveries and insights. Annu
Rev Immunol 1996; 14:649 – 683
5. Qing M, Schumacher K, Heise R, et al: Intramyocardial synthesis of pro- and antiinflammatory cytokines in infants with congenital cardiac defects. J Am Coll Cardiol
2003; 41:2266 –2274
6. Liakopoulos OJ, Schmitto JD, Kazmaier S, et
al: Cardiopulmonary and systemic effects of
methylprednisolone in patients undergoing
cardiac surgery. Ann Thorac Surg 2007; 84:
110 –118; discussion 118 –119
7. Bronicki RA, Backer CL, Baden HP, et al:
Dexamethasone reduces the inflammatory
response to cardiopulmonary bypass in children. Ann Thorac Surg 2000; 69:1490 –1495
8. Checchia PA, Backer CL, Bronicki RA, et al:
Dexamethasone reduces postoperative troponin levels in children undergoing cardiopul-
Crit Care Med 2009 Vol. 37, No. 2
monary bypass. Crit Care Med 2003; 31:
1742–1745
9. Moen O, Hogasen K, Fosse E, et al: Attenuation of changes in leukocyte surface markers and complement activation with heparincoated cardiopulmonary bypass. Ann Thorac
Surg 1997; 63:105–111
10. Seghaye MC, Duchateau J, Grabitz RG, et al:
Complement activation during cardiopulmonary bypass in infants and children. Relation to
postoperative multiple system organ failure.
J Thorac Cardiovasc Surg 1993; 106:978 –987
11. Checchia PA, Bronicki RA, Costello JM, et al:
Steroid use before pediatric cardiac opera-
tions using cardiopulmonary bypass: An international survey of 36 centers. Pediatr Crit
Care Med 2005; 6:441– 444
12. Yeh TF, Lin YJ, Lin HC, et al: Outcomes at
school age after postnatal dexamethasone
therapy for lung disease of prematurity.
N Engl J Med 2004; 350:1304 –1313
13. Duffy JY, McLean KM, Lyons JM, et al: Modulation of nuclear factor-kappaB improves cardiac dysfunction associated with cardiopulmonary bypass and deep hypothermic circulatory
arrest. Crit Care Med 2009; 37:577–583
14. Yeh CH, Chen TP, Wu YC, et al: Inhibition of
NFkappaB activation with curcumin attenu-
ates plasma inflammatory cytokines surge
and cardiomyocytic apoptosis following cardiac ischemia/reperfusion. J Surg Res 2005;
125:109 –116
15. Schwartz SM, Duffy JY, Pearl JM, et al: Glucocorticoids preserve calpastatin and troponin
I during cardiopulmonary bypass in immature
pigs. Pediatr Res 2003; 54:91–97
16. Yeh CH, Chen TP, Lee CH, et al: Cardioplegia-induced cardiac arrest under cardiopulmonary bypass decreased nitric oxide production which induced cardiomyocytic
apoptosis via nuclear factor kappa B activation. Shock 2007; 27:422– 428
“Pas de DEux” for phosphodiesterase-2 in acute lung injury*
T
he phosphodiesterase (PDE)-2
isozyme was first discovered as
a cyclic guanosine monophosphate (cGMP)-stimulated cyclic
nucleotide PDE2 capable of hydrolyzing
both cyclic adenosine monophosphate
(cAMP) and cGMP (1). Three variants of the
unique PDE2 gene have been described. All
of them are specifically inhibited by 9-(2hydroxy-3-nonyl) adenine monohydrocrochloride (EHNA) when PDE2 is activated by
cGMP in vitro. PDE2 is expressed in both
endothelial and pulmonary epithelial cells
(2) and, beyond membrane receptors, this
isozyme regulates cyclic nucleotide levels
(cAMP and cGMP) and several highly localized intracellular signaling pathways such
as cAMP dependent protein kinase and
cGMP dependent protein kinase-dependent
phosphorylation. Furthermore, when regulating local cAMP and cGMP, PDE2 controls the (adenosine triphosphate)/(cAMP)
energetic ratio.
PDEs are ubiquitous enzymes constituted by a multigenic super family
(PDE1–11) (3). They have been involved
in various feedback processes, and PDE
inhibitors have been developed and used
as bronchorelaxant (theophylline), cardiotonic (milrinone), anti-inflammatory
(rolipram), and vasodilating drugs (such
as sildenafil). The putative role of PDE2
*See also p. 584.
Key Words: phosphodiesterases; inhibitors; acute
lung injury; lung; permeability; pneumonia
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194c295
Crit Care Med 2009 Vol. 37, No. 2
in inflammatory processes or infectious
diseases has been evoked, although evidences have been somewhat scant.
Hence, the classic PDE2 inhibitor EHNA
has been mainly used in in vitro assays,
because its poor specificity has precluded
its use in in vivo studies.
PDE2s, which are mainly cytosolic as
well as particulate (membrane attached)
enzymes, are likely surrounded by a complex microenvironment in vivo, according to intracellular molecular crowding.
Consequently, it would not be surprising
that PDE2s are associated with specific
transmembrane receptors within specific
microdomains. Such intracellular location would facilitate the control of local
concentrations of cAMP (or cGMP) in the
vicinity of the molecular complexes. Conversely, since PDE2 hydrolyzing activity
is stimulated by cGMP (5 M) via an
allosterically regulated site, the presence
of a guanylate cyclase in contiguity with
the signaling complex could also represent a hypothesis to be ascertained.
Using combined classic methodologic
approaches and an array of pharmacologic conditions involving two new PDE2
inhibitors, namely PDP and hydroxylPDP, Witzenrath et al in this issue of
Critical Care Medicine (4) demonstrated
that prophylactic and systemic infusion
of these specific PDE2 inhibitors reduced
acute lung injury (ALI) in a mice model
of pneumococcal infection. This noteworthy observation was also supported by a
constellation of preliminary and original
results obtained in endothelial cell monolayers (5). Hence, both PDE inhibitors
were able to oppose the electrophysiologic effects of pneumococcal pneumo-
lysin in endothelial cell (human umbilical veinous endothelial cells [HUVEC])
monolayers. Thus, PDE2 appears as a key
target in this process, since the contribution of other PDE isozymes was elegantly
ruled out in control experiments.
Despite a relevant set of preliminary
data gained on endothelial cell monolayers in which PDE2 expression was stimulated by exogenous tumor necrosis factor-␣ (4), this study partially performed
in a murine model and in isolated lungs
did not allow to pinpoint the precise location of the inhibited PDE2. In addition,
other more specific types of endothelial
cell cultures, such as lung microvascularderived cells, should have been favorably
compared with HUVEC. Furthermore, although endothelial cells are likely candidates, the effect of PDE2 inhibitors on
epithelial cell layers at the infection site
(nasal mucosa) or within the respiratory
track may have been underestimated (6).
In this respect, although the selection of
electrical resistance of endothelial cell
(HUVEC) and human serum albumin
leakage certainly represented a sound
choice, other tools were not considered
such as small molecular weight molecule
(e.g., Evans blue, fluorescein) or microprotein (e.g., Clara cell protein 16) leakages (7), for assessing epithelial damage.
Histopathologic examination of lung tissues should have also served for analysis
of interstitial edema analysis to further
consolidate PDE2 inhibition-induced decreased permeability.
Sepsis-induced tissue and lung hyperpermeability is the “underdog” in terms of
research on mechanistic events leading to
ALI/acute respiratory distress syndrome.
769
Indeed, hyperpermeability is a neglected
but important event leading to sepsisinduced organ disability, due to a lack of
specific tools to assess this particular phenomenon. However, it is a potentially leading cause of organ dysfunction/failure, either by impairing exchange ability (e.g.,
lung, kidney) or by compromising organ
perfusion, especially for capsule- or membrane-bearing tissues or those exhibiting
bone-limited compliances (e.g., kidney, adrenal gland, liver, brain) (8). However, as
stated above, microvascular permeability is
likely only half of the story, especially with
regard to organ exchanges such as in lung
or kidney. Indeed, the epithelium should be
considered as the other side of the coin,
since it is clearly committed in lung barrier
permeability (9). Water and solutes (i.e.,
sodium), through aquaporins and eNac
pumps (in conjunction with Na⫹-K⫹adenosine triphosphatase basal pumps), are
able to clear excess permeability in lung
distal airspaces in ALI (10, 11), and therefore have to be taken into account together
with endothelium impairment. Finding inhibitors of tissue permeability is a challenging issue requiring extensive research,
which in return could yield invaluable applications at least for combination therapies for ALI at the bedside. This issue
should have been explored in the study by
Witzenrath et al (4), at least by specific in
vitro lung epithelial cultures or optimally
by epithelial-endothelial cocultures.
In summary, the data described by
Witzenrath et al (4) undoubtedly provide
an important addition to our current
knowledge on the role of PDE2 in host
infection by Streptocococcus pneumoniae. This isozyme may indeed regu-
770
late key cell signaling processes, whose
activation would be required to facilitate
the early steps directly involved in the
infectious cascade triggered by S. pneumoniae strains. In an explosion of interest
for this concept, a rapid succession of studies will likely identify the missing links, the
next step being the identification of the
membrane receptor upstream (such as tolllike receptor) and the cyclic nucleotidesensitive effectors, downstream of PDE2 in
either endothelial or epithelial cell layers.
An alternative issue would be that low levels of cAMP would facilitate interaction of
S. pneumoniae exotoxin proteins with specific membrane receptors, whereas highly
localized increases in cAMP levels (on
PDE2 inhibition) would abolish this
early step. Despite the fact that the role
of proinflammatory cytokines cannot be
ruled out, clinical teams will likely focus on the ability of new specific PDE2
inhibitors to decrease morbidity in ALIrelated conditions to sever S. pneumoniae in curative protocols.
Olivier Lesur, MD, PhD
MICU, Department of Medicine
CHU Sherbrooke-MICU
Sherbrooke, Quebec, Canada
Eric Rousseau, PhD
Department of Physiology and
Biophysics, FMSS
Université of Sherbrooke
Quebec, Canada
3.
4.
5.
6.
7.
8.
9.
10.
REFERENCES
1. Beavo JA, Hardman JG, Sutherland EW: Stimulation of adenosine 3⬘,5⬘- monophosphate hydrolysis by guanosine 3⬘,5⬘-monophosphate. J Biol
Chem 1971; 246:3841–3846
2. Rousseau E, Gagnon J, Lugnier C: Biochem-
11.
ical and pharmacological characterisation of
cyclic nucleotide phosphodiesterase in airway epithelium. Mol Cell Biochem 1994; 140:
171–175
Keravis T, Lugnier C: Cyclic nucleotide phosphodiesterase (PDE) superfamily and smooth
muscle signaling. In: New Frontier in
Smooth Muscle Biology and Physiology.
Chap.13. Savineau JP (Ed), Transworld Research Network, Kerala, India, 2007, pp
269 –289
Witzenrath M, Gutbier B, Schmeck B, et al:
Phosphodiesterase 2 inhibition diminished
acute lung injury in murine pneumococcal
pneumonia. Crit Care Med 2009; 37:584 –590
Seybold J, Thomas D, Witzenrath M, et al:
Tumor necrosis factor-␣-dependent expression of phosphodiesterase 2: Role in
endothelial hyperpermeability. Blood 2005;
105:3569 –3576
Boswell-Smith V, Spina D, Page CP: Phosphodiesterase inhibitors [review]. Br J Pharmacol 2006; 147(Suppl 1):S252–S257
Lesur O, Hermans C, Chalifour JF, et al:
Pneumoprotein (CC-16) vascular transfer
during mechanical ventilation in rats: Effect
of KGF pretreatment. Am J Physiol Lung
Cell Mol Physiol 2003; 284:L410 –L419
Farand P, Hamel M, Lauzier F, et al: Effects
of norepinephrine and vasopressin on sepsisinduced organ perfusion/permeability [review]. Can J Anaesth 2006; 53:934 –946
Kim K-J, Malik AB: Protein transport
across the lung epithelial barrier. Am J
Physiol Lung Cell Mol Physiol 2003; 284:
L247–L259
Zemans RL, Matthay MA: Bench-to-bedside
review: The role of the alveolar epithelium in
the resolution of pulmonary edema in ALI.
Crit Care 2004; 8:469 – 477
Verkman AS, Matthay MA, Song Y: Aquaporins water channels and lung physiology.
Am J Physiol Lung Cell Mol Physiol 2000;
278:L867–L879
Crit Care Med 2009 Vol. 37, No. 2
Hemorrhagic shock and reperfusion injury: The critical interplay
of fibrin fragments, leukocytes, and vascular endothelial-cadherin*
R
eperfusion, the restoration of
blood flow after a period of
ischemia, can place ischemic
organs at risk of further cellular necrosis and thereby limit the recovery of function. In particular, the microvasculature is vulnerable to the
deleterious consequences of ischemia and
reperfusion (I/R). The underlying pathophysiology of I/R injury is not fully understood, but several mechanisms are involved such as membrane damage
inflicted by oxygen radicals, intracellular
calcium overload, and tissue damage
caused by infiltration and activation of
white blood cells (1, 2). Reperfusion of
ischemic tissues is often associated with
microvascular dysfunction that is manifested as impaired endothelium-dependent dilation in arterioles, enhanced fluid
filtration, leukocyte plugging in capillaries, and the trafficking of leukocytes and
plasma protein extravasation in postcapillary venules. The resulting imbalance
between superoxide and nitric oxide in
endothelial cells leads to the production
and release of inflammatory mediators
(e.g., platelet-activating factor, tumor necrosis factor) and enhances the biosynthesis of adhesion molecules that mediate
leukocyte– endothelial cell adhesion. The
bioavailability of nitric oxide, an important mediator of vasodilatation, is profoundly decreased during the reperfusion
period, resulting in impaired vasodilatation
of arterioles. Activated endothelial cells in
all segments of the microcirculation produce more oxygen radicals, but less nitric
oxide, in the initial period following reperfusion (3). Induction of complement activation and subsequent release of inflammatory mediators as well as increased
*See also p. 598.
Key Words: B15-42; fibrin-related peptide; FX06;
hemorrhagic shock; ischemia; organ failure; reperfusion
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194bd9e
Crit Care Med 2009 Vol. 37, No. 2
expression of adhesion molecules initiate
inflammatory and coagulation cascades
that culminate in the occlusion of capillaries, known as the “no-reflow” phenomenon. In postcapillary venules, the recruitment and transmigration of leukocytes
further compromise the integrity of the
endothelial barrier and increase the oxidative burden, resulting in leakage and tissue
edema (4). The trafficking of leukocytes in
venules is the rate-limiting determinant of
I/R-induced endothelial barrier dysfunction. The magnitude of albumin leakage in
postischemic venules is highly correlated
with the number of adherent and emigrated leukocytes. Adhesion moleculedirected antibodies that effectively blunt
leukocyte adherence/emigration also exert
an attenuating action on I/R-induced albumin leakage (5). Leukocytes attach to the
blood vessel wall via interaction of selectins
and integrins with their respective receptors, and become progressively activated.
To accomplish the final step of transmigration through the endothelial layer, leukocytes must cross a multilayered molecular
zipper of cell– cell junctions. Leukocyte
transmigration is critically controlled by
fibrin fragments. These short-lived intermediates engage leukocytes to the endothelial cell junction, which breaks up the VEcadherin interaction between neighboring
endothelial cells, allowing the leukocytes to
pass through into the tissue (diapedesis).
In myocardial infarction, about 50% of
overall tissue damage is caused by reperfusion injury. For several decades, it has
been known that the inflammatory response during reperfusion is the major
cause of myocardial I/R injury. If uncontrolled, the inflammation results in irreversible damage of the heart muscle, limiting the success of reperfusion
procedures. The inflammatory reaction
can be mitigated by application of B1542, also called FX06, concomitant with
reperfusion (6 –9). FX06, is a polypeptide,
derived from the neo-N-terminus of fibrin, being the natural cleavage product
of fibrin after being exposed to plasmin.
FX06 targets an endothelial adherens
junction protein, VE-cadherin, and reduces leukocyte transmigration across
endothelial junctions and the release of
proinflammatory cytokines (6 –9). Therefore, FX06 prohibits the emigration of all
leukocyte subtypes into myocardial tissue
by preventing the critical interaction between fibrin fragments and VE-cadherin,
which is rate limiting and irreversible.
Previous data on acute and chronic
myocardial I/R models were promising
and appeared to reduce myocardial inflammation and infarct size (6 –9). In occlusion–reperfusion studies in rats, FX06
(2.4 mg/kg optimal dose) caused 40% reduction in infarct size (6). The positive
effects of FX06 on the inflammatory response were reflected by a decrease in
cytokines (interleukin [IL]-6, troponin).
The safety and tolerability of FX06 has
been demonstrated in a phase I trial in 30
healthy male volunteers, as no significant
toxicity and adverse events were observed
(7). A phase II trial on the treatment with
FX06 for reperfusion injury in myocardial
infarction (F.I.R.E. trial) has been completed recently and is designed to clinically evaluate infarct size at 5–7 days
post-percutaneous coronary intervention
(www.clinicaltrial.gov) (10).
In this issue of Critical Care Medicine,
Roesner et al (11) report on the beneficial
effects of this novel endogenous polypeptide FX06 in a hemorrhagic shock model.
Hemorrhagic shock followed by volume
resuscitation largely resembles the state
of myocardial I/R. The key treatment of
hemorrhagic shock involves early hemorrhage control, aggressive resuscitation,
and the prevention and correction of coagulopathy (12). The current management of hemorrhagic shock relies heavily
on transfusion of red blood cells that are
associated with the development of multiple organ failure, increased intensive
care unit admissions and length of stay,
increased hospital length of stay, and
mortality (13). Other therapeutic options
are limited because of lack of understanding the pathophysiological mechanisms
and lack of a specific target. Some novel
771
(experimental) interventions to limit
reperfusion damage have some promise
such as sodium/hydrogen exchange suppression (14), melatonin (15), IL-11 (16),
or selectin inhibition (17), but failed to
reach the clinical stage. In this context,
the salutary effects of the compound
FX06 on hemorrhagic shock-induced organ injury are potentially important and
clinically relevant (11). The controlledshock model was characterized by rather
severe tissue damage (I/R and laparotomy), a long (5 hours) reperfusion time,
and adequate volume resuscitation
guided by volumetric preload parameters.
FX06-treated animals showed a reduction
in myocardial damage and leukocyte infiltration in the heart, as well as less liver/
intestinal damage. The effects on lung
damage are less clear; extra vascular lung
water and PaO2/FIO2 ratios were only improved at the end of reperfusion. Apparently, IL-6 may be a key factor, as the
reduced organ failure and inflammation
strongly correlated with a reduction in
IL-6 in the FX06 group. These intriguing
results are very promising and will certainly lead to further preclinical research
in I/R injury. The translation of the findings in the hemorrhagic shock reperfusion model with limitations toward the
complex clinical traumatic shock setting
with far more tissue injury is too premature. Some questions remain unclear;
whether an endogenous HX06 deficit is
present during shock or resuscitation or
whether multiple (with respect to the
very short half-life of 15 minutes) or
higher doses of FX06 may be superior.
The exact mechanisms involved in the
observed improved organ function remain obscure, but similar to previous
772
findings on myocardial I/R damage, the
impact of FX06 on neutrophil organ infiltration seems to be most prominent.
Albertus Beishuizen, MD
Armand R. J. Girbes, MD
Department of Intensive Care
University Hospital VU
Medical Center
Amsterdam, The Netherlands
10.
REFERENCES
1. Liem DA, Honda HM, Zhang J, et al: Past and
present course of cardioprotection against
ischemia-reperfusion injury. J Appl Physiol
2007; 103:2129 –2136
2. Carden DL, Granger DN: Pathophysiology of
ischaemia-reperfusion injury. J Pathol 2000;
190:255–266
3. Grisham MB, Granger DN, Lefer DJ: Modulation of leukocyte-endothelial interactions
by reactive metabolites of oxygen and nitrogen: Relevance to ischemic heart disease.
Free Radic Biol Med 1998; 25:404 – 433
4. Granger DN: Ischemia-reperfusion: Mechanisms of microvascular dysfunction and the
influence of risk factors for cardiovascular
disease. Microcirculation 1999; 6:167–178
5. Kurose I, Anderson DC, Miyasaka M, et al:
Molecular determinants of reperfusioninduced leukocyte adhesion and vascular
protein leakage. Circ Res 1994; 74:336 –343
6. Petzelbauer P, Zacharowski PA, Miyazaki Y,
et al: The fibrin-derived peptide Bbeta15-42
protects the myocardium against ischemiareperfusion injury. Nat Med 2005; 11:
298 –304
7. Roesner JP, Petzelbauer P, Koch A, et al: The
fibrin-derived peptide Bbeta15-42 is cardioprotective in a pig model of myocardial ischemia-reperfusion injury. Crit Care Med 2007;
35:1730 –1735
8. Zacharowski K, Zacharowski P, Reingruber
S, et al: Fibrin(ogen) and its fragments in the
pathophysiology and treatment of myocardial infarction. J Mol Med 2006; 84:469 – 477
9. Zacharowski K, Zacharowski PA, Friedl P, et
11.
12.
13.
14.
15.
16.
17.
al: The effects of the fibrin-derived peptide
Bbeta(15-42) in acute and chronic rodent
models of myocardial ischemia-reperfusion.
Shock 2007; 27:631– 637
Atar D, Huber K, Rupprecht HJ, et al: Rationale
and design of the ‘F.I.R.E.’ study. A multicenter, double-blind, randomized, placebocontrolled study to measure the effect of FX06
(a fibrin-derived peptide Bbeta(15– 42)) on
ischemia-reperfusion injury in patients with
acute myocardial infarction undergoing primary percutaneous coronary intervention.
Cardiology 2007; 108:117–123
Roesner JP, Petzelbauer P, Koch A, et al:
B15-42 (FX06) reduces pulmonary, myocardial, liver, and small intestine damage in a
pig model of hemorrhagic shock and reperfusion. Crit Care Med 2009; 37:598 – 605
Kauvar DS, Lefering R, Wade CE: Impact of
hemorrhage on trauma outcome: An overview of epidemiology, clinical presentations,
and therapeutic considerations. J Trauma
2006; 60:S3–S11
Malone DL, Dunne J, Tracy JK, et al: Blood
transfusion, independent of shock severity, is
associated with worse outcome in trauma.
J Trauma 2003; 54:898 –905
Buerke U, Pruefer D, Carter JM, et al: Sodium/hydrogen exchange inhibition with cariporide reduces leukocyte adhesion via Pselectin suppression during inflammation.
Br J Pharmacol 2008; 153:1678 –1685
Mathes AM, Kubulus D, Pradarutti S, et al:
Melatonin pretreatment improves liver function and hepatic perfusion after hemorrhagic
shock. Shock 2008; 29:112–118
Honma K, Koles NL, Alam HB, et al: Administration of recombinant interleukin-11 improves the hemodynamic functions and decreases third space fluid loss in a porcine
model of hemorrhagic shock and resuscitation. Shock 2005; 23:539 –542
Calvey CR, Toledo-Pereyra LH: Selectin inhibitors and their proposed role in ischemia
and reperfusion. J Invest Surg 2007; 20:
71– 85
Crit Care Med 2009 Vol. 37, No. 2
Peroxisome proliferator-activated receptor-gamma agonists,
control of bacterial outgrowth, and inflammation*
I
n the 1980s, at a time when few
cytokines were described, a new
paradigm emerged that sepsis may
be a syndrome that consists of an
inappropriate and maladaptive systemic
inflammatory response induced in response to infection. Numerous attempts
were made to treat sepsis in the clinic
using immune modulators that block
proinflammatory cytokine mediators
such as tumor necrosis factor or interleukin-1, or other components of innate immunity. Ultimately, all of these downregulating immune modulators failed in
clinical trials to treat sepsis.
Despite their failure in the acute setting of infection, many of the same antiinflammatory treatments are now proven
to be effective treatments for chronic inflammatory conditions such as rheumatoid arthritis and Crohn’s disease. Ironically, many of the treatments that were
initially developed to treat sepsis have, in
fact, turned out to predispose to infection
and sepsis (1). This finding, coupled with
data in animal experiments, suggests that
blocking even single components of the
innate immune system runs the risk of
altering host defense sufficiently to compromise the elimination of microorganisms. For this reason, agents that suppress the inflammatory response in the
setting of infection have traditionally
been studied for use as adjuvant treatment in addition to primary antimicrobial treatment.
Peroxisome proliferator-activated receptor-gamma (PPAR␥) is a member of
the nuclear receptor family of transcription factors that mediates transcriptional
*See also p. 614.
Key Words: peroxisome proliferator-activated
receptor-␥; inflammation; bacteria; mouse model;
pneumonia
The author has received grant support from the
National Institute of Health AI059010, GM 59694, and
the Shriners Hospital for Crippled Children 8720.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194c4f9
Crit Care Med 2009 Vol. 37, No. 2
activation or repression of genes related
to lipid metabolism, cell proliferation, angiogenesis, and inflammation. PPAR␥ agonist ligands diminish insulin resistance
and are used to help treat type 2 diabetes.
PPAR␥ ligands also repress the gene for
nitric oxide synthase, as well as tumor
necrosis factor, interleukin-6, and interleukin-1 (2, 3). Because of these and
other anti-inflammatory activities, it
seems reasonable to study their potential
use as an anti-inflammatory agent in the
setting of infection.
In this issue of Critical Care Medicine,
Stegenga et al (4) report a study in which
a synthetic PPAR␥ ligand, ciglitazone,
was evaluated in an established mouse
model of pneumonia with Streptococcus
pneumoniae. The drug was administered
to groups of mice either once at the start
of the infectious challenge or twice with
the first dose at challenge and a second
dose at 24 hrs. The mice were evaluated
at 24 and 48 hrs after infectious challenge. Mice that received ciglitazone had
decreased bacterial counts in the lungs
compared with controls; in addition,
there were decreased inflammatory cytokines in lung tissues at 24 hrs and decreased inflammation on histopathology
at 48 hrs. There was also a trend toward a
decreased mortality in the drug group.
It seems remarkable that a drug that
possesses anti-inflammatory effects would
decrease bacterial outgrowth. Because the
innate immune system presumably
evolved to control infection, one might
expect the opposite effect. Indeed specific
inhibition of the interleukin-1 or tumor
necrosis factor in a similar model has
been reported to lead to increased bacterial loads and decreased survival (5, 6).
Therefore, the results reported in the
current study in this model with the
PPAR␥ ligand ciglitazone seem the best
of all situations for an anti-inflammatory
drug— decreased inflammation and at
the same time, decreased bacteria.
The mechanism of the drug’s effect is
not clear. There are several possibilities
that the authors have addressed. First,
there could be some sort of direct antibacterial action if the drug was acting as
an antibiotic. There was apparently no
antibacterial effect when the authors
tested the drug in vitro at 5 g/mL. However, ciglitazone was administered to the
mice at 5 mg/kg; depending on the pharmacokinetics, it is possible that drug levels could have exceeded 5 g/mL for
some period of time. Second, a decrease
in bacterial proliferation could be explained if ciglitazone had stimulated bacterial clearance in some way. PPAR␥ ligands have been reported to increase
phagocytosis through increasing expression of CD36 (7), as well as through Fc␥
(8). The authors did not detect an effect
of the drug using a murine alveolar cell
line to assess phagocytosis of S. pneumoniae labeled with fluorescein isothiocyanate, and did not detect increased killing in these cells using a capsuledeficient strain of S. pneumoniae. It is
unclear, however, how well this in vitro
system with a cell line reflects the in vivo
situation in the mouse model using an
encapsulated strain of S. pneumoniae.
The results of Stegenga et al with S.
pneumoniae are consistent with two
other publications that reported that
ciglitazone decreased inflammation without an increase in bacterial counts. One
of these used the cecal ligation and puncture model (9), and the other a model of
brain abscess with Staphylococcus aureus (10). Thus, the results may extend to
organisms other than S. pneumoniae.
Interpretation of decreased inflammation in the setting of decreased bacterial
load is complex. It is not clear from the
present study if the antibacterial effect
and the decreased inflammation stem
from two different activities of the drug,
one leading to decreased inflammation
and the other to decreased bacterial
counts. An alternative and simpler explanation is that the decreased inflammation
may follow simply from the presence of
fewer bacteria in tissues leading to less of
a stimulus to the host.
773
As always, there are many cautions in
extrapolating results of animal models to
human disease. Mice are much more resistant than humans to most forms of
induced inflammation. It will be essential
to determine the mechanism of the antibacterial effect and to determine whether
there is any additional advantage when
ciglitazone is used as adjuvant treatment
in addition to simultaneous traditional
antibiotics. Regardless, this study is provocative and raises some hope that
PPAR␥ ligands could be a new approach
to an area that could use a breakthrough
in treatment.
H. Shaw Warren, MD
Infectious Disease Unit
Massachusetts General Hospital
Boston, MA
REFERENCES
1. Bongartz T, Sutton AJ, Sweeting MJ, et al:
Anti-TNF antibody therapy in rheumatoid ar-
2.
3.
4.
5.
6.
thritis and the risk of serious infections and
malignancies: Systematic review and metaanalysis of rare harmful effects in randomized controlled trials. J Am Med Assoc 2006;
295:2275–2285
Ricote M, Li AC, Willson TM, et al: The peroxisome proliferator-activated receptorgamma is a negative regulator of macrophage activation. Nature 1998; 391:79 – 82
Jiang C, Ting AT, Seed B: PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature 1998;391:82– 86
Stegenga ME, Florquin S, de Vos M, et al: The
thiazolidinedione ciglitazone reduces bacterial
outgrowth and early inflammation during
Streptococcus pneumoniae pneumonia in
mice. Crit Care Med 2009; 37:614 – 618
Rijneveld AW, Florquin S, Hartung T, et al:
Anti-tumor necrosis factor antibody impairs
the therapeutic effect of ceftriaxone in murine pneumococcal pneumonia. J Infect Dis
2003; 188:282–285
Rijneveld AW, Florquin S, Branger J, et al:
TNF-alpha compensates for the impaired
host defense of IL-1 type I receptor-deficient
7.
8.
9.
10.
mice during pneumococcal pneumonia.
J Immunol 2001; 167:5240 –5246
Asada K, Sasaki S, Suda T, et al: Antiinflammatory roles of peroxisome proliferatoractivated receptor gamma in human alveolar
macrophages. Am J Respir Crit Care Med
2004; 169:195–200
Aronoff DM, Serezani CH, Carstens JK, et al:
Stimulatory effects of peroxisome proliferator-activated receptor-gamma on Fcgamma
receptor-mediated phagocytosis by alveolar
macrophages. PPAR Res 2007; 2007:52546
Zingarelli B, Sheehan M, Hake PW, et al:
Peroxisome proliferator activator receptorgamma ligands, 15-deoxy-Delta(12,14)prostaglandin J2 and ciglitazone, reduce
systemic inflammation in polymicrobial
sepsis by modulation of signal transduction pathways. J Immunol 2003; 171:
6827– 6837
Kielian T, Syed MM, Liu S, et al: The synthetic peroxisome proliferator-activated receptor-gamma agonist ciglitazone attenuates
neuroinflammation and accelerates encapsulation in bacterial brain abscesses. J Immunol 2008; 180:5004 –5016
Look on the “air side” in pneumonia*
P
neumonia is the leading cause
of acute lung injury (ALI) (1).
The pathophysiology of ALI involves an uncontrolled host defense response, with a malignant alveolar
cross talk between inflammation and hemostasis activation (2), which further
propagates as the natural anticoagulant
axis is disrupted (3).
Despite the use of early broad-spectrum antibiotics, the mortality of pneumonia is still high with an “attributable
mortality” from hospital-acquired Grampositive and Gram-negative pneumonia
of 33% to 50% (4). One explanation is
that the mortality is not only attributed
to the infection per se but also to the
antigens of the bacteria.
In this issue of Critical Care Medicine,
Hoogerwerf et al (5) shows for the first time
*See also p. 619.
Key Words: pneumonia; inhaled activated protein
C; Gram-positive antigen; Gram-negative antigen; diffuse alveolar damage; pulmonary deposition; alveolar
hemostasis activation
The author is a CSO x1 in a biotech company that
owns the patent on inhaled APC.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194cf47
774
that antigens from cell walls of both Grampositive and Gram-negative bacteria have
the same effect in humans. Furthermore,
they also showed that alveolar levels of
antithrombin and activated protein C
(APC) concentrations were reduced.
This was meticulously documented by
this randomized study of instillation of
saline, lipoteichoic acid, or lipopolysaccharide, i.e., applying the antigens from
the “air side.” Their findings are not
self-explanatory in as much as Grampositive and Gram-negative pathogens
actuate immune and procoagulant responses in the lung via different recognition receptors, i.e., via toll-like receptors 2 and 4, respectively.
Severe pneumonia in hospitals has
still a high mortality despite applying
evidence-based early use of broadspectrum antibiotics. As documented
by Hoogerwerf et al, the problem is that
the pulmonary dysfunction in pneumonia is further aggravated by the antigens of the bacterial cell walls.
How do we then offset the changes
induced from the air side, and, further
does pharmacotherapy with natural anticoagulants and fibrinolytics have a role in
the treatment of ALI? It is logical to in-
troduce such a therapy using natural anticoagulants because the intra-alveolar
hemostasis activation is not sufficiently
counterbalanced by natural inhibitors,
such as tissue factor pathway inhibitor
(6) and APC (7, 8).
Tissue factor pathway inhibitor was
shown to reduce lung injury and systemic
levels of inflammatory cytokines in an
animal study (9), and, further, in a subsequent phase II trial, the results were
promising. However, this benefit was lost
in a subsequent phase III trial (10). At
present, we are awaiting the results from
a placebo-controlled trial in communityacquired pneumonia using intravenous
tissue factor pathway inhibitor (11).
Since the introduction of the recombinant APC for the treatment of severe
sepsis (Drotrecogin alpha activated) (12),
several studies have used APC intravenously to counteract the hemostatic
changes in ALI (8). Recently, a controlled
trial using intravenous APC in patients
with acute lung injury was published
(13). The study revealed that APC did not
improve outcomes of acute lung injury
and concluded that the results “do not
support a large clinical trial of APC in
acute lung injury.” Furthermore, intraveCrit Care Med 2009 Vol. 37, No. 2
nous APC has been shown to result in
noteworthy bleeding as alluded to in two
recent articles (14, 15). The adverse effects of systemic bleeding may be avoided
if APC is inhaled and provided that there
is no spillover from the alveoli to the systemic circulation (16).
From a theoretical viewpoint, inhaled
APC has all the essential properties to
counteract the pathophysiologic changes
seen in ALI. In an animal study by
Slofstra et al (17), the lipopolysaccharideinduced inflammation was reduced after
inhalation of APC. A human experience
with inhaled APC has been reported in a
patient with ALI (16), with an important
improvement in pulmonary gas exchange
concomitant with a noteworthy clearing
of the pulmonary opacities.
There is thus a good reason to “look at
the pneumonia from the air side” not only
with respect to explaining the acute pathophysiologic changes ensuing pneumonia,
but also treating the condition from the
same side by inhaling a natural anticoagulant, such as APC. APC may be an important adjunctive intervention in severe
pneumonia caused by both Gram-positive
and Gram-negative bacteria. There is, however, only sparse information to substantiate such a new treatment paradigm. The
next step, therefore, is to conduct controlled trials with inhaled APC because the
intravenous intervention has already
proved ineffective.
Lars Heslet, MD, MSc, DMedSci
Department of Intensive Care
Rigshospitalet
Copenhagen, Denmark
Crit Care Med 2009 Vol. 37, No. 2
REFERENCES
1. Gunther A, Mosavi P, Heinemann S, et al:
Alveolar fibrin formation caused by enhanced
procoagulant and depressed fibrinolytic capacities in severe pneumonia. Comparison
with the acute respiratory distress syndrome.
Am J Respir Crit Care Med 2000; 161:
454 – 446
2. Ware LB: Pathophysiology of acute lung injury and the acute respiratory distress syndrome. Semin Respir Crit Care Med 2006;
27:337–349
3. Schultz MJ, Haitsma JJ, Zhang H, et al: Pulmonary coagulopathy as a new target in therapeutic studies of acute lung injury or pneumonia: A review. Crit Care Med 2006; 34:
871– 877
4. Isakow W, Kollef MH: Preventing ventilatorassociated pneumonia: An evidence-based
approach of modifiable risk factors. Semin
Respir Crit Care Med 2006; 27:5–17
5. Hoogerwerf JJ, de Vos AF, Levi M, et al:
Activation of coagulation and inhibition of
fibrinolysis in the human lung on bronchial instillation of lipoteichoic acid and
lipopolysaccharide. Crit Care Med 2009;
37:619 – 625
6. Gando S, Kameue T, Matsuda N, et al:
Imbalance between the levels of tissue factor and tissue factor pathway inhibitor in
ARDS patients. Thromb Res 2003; 109:
119 –124
7. Ware LB, Fang X, Matthay MA: Protein C and
thrombomodulin in human acute lung injury. Am J Physiol Lung Cell Mol Physiol
2003; 285:L514 –L521
8. van der Poll T, Levi M, Nick JA, et al: Activated protein C inhibits local coagulation
after intrapulmonary delivery of endotoxin in
humans. Am J Respir Crit Care Med 2005;
171:1125–1128
9. Welty-Wolf KE, Carraway MS, Miller DL, et
10.
11.
12.
13.
14.
15.
16.
17.
al: Coagulation blockade prevents sepsisinduced respiratory and renal failure in baboons. Am J Respir Crit Care Med 2001;
164:1988 –1996
Abraham E, Reinhart K, Opal S, et al: Efficacy and safety of tifacogin (recombinant tissue factor pathway inhibitor) in severe sepsis: A randomized controlled trial. JAMA
2003; 290:238 –247
LifeSciencesWorld: Chiron begins phase III
trial for tifacogin. May 18, 2004. Available at:
www.lifesciencesworld.com/rss/tifacogin/
news. Accessed October 12, 2006
Bernard GR, Vincent JL, Laterre PF, et al;
Recombinant human protein C Worldwide
Evaluation in Severe Sepsis (PROWESS)
study group: Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med 2001; 344:
699 –709
Liu KD, Levitt J, Zhuo H, et al: Randomized
clinical trial of activated protein C for the
treatment of acute lung injury. Am J Respir
Crit Care Med 2008; 178:618 – 623
Martí-Carvajal A, Salanti G, Cardona AF: Human recombinant activated protein C for severe sepsis. Cochrane Database Syst Rev
2008; (1):CD004388
Levi M: Activated protein C in sepsis: A critical review. Curr Opin Hematol 2008; 15:
481– 486
Heslet L, Andersen JS, Sengeløv H, et al:
Inhalation of activated protein C: A possible
new adjunctive intervention in acute respiratory distress syndrome. Targets Ther 2007;
1:1– 8
Slofstra SH, Groot AP, Maris NA, et al:
Inhalation of activated protein C inhibits
endotoxin-induced pulmonary inflammation in mice independent of neutrophil recruitment. Br J Pharmacol 2006; 149:
740 –746
775
The role of angiotensin-converting enzyme inhibition in
endotoxin-induced lung injury in rats*
T
he renin-angiotensin system
(RAS) plays an important role
in the homeostasis of systemic
blood pressure, but it can also
regulate inflammation. RAS consists of
the renin protease that cleaves angiotensinogen into angiotensin I (Ang I).
Angiotensin-converting enzyme (ACE) is
primarily expressed on the surface of pulmonary microvascular endothelial cells
and cleaves Ang I into angiotensin II (Ang
II) (1). ACE also inactivates the vasodilator bradykinins. The biological effects of
the RAS system are mediated by Ang II on
its specific receptors, Ang II receptor type
1 and Ang II receptor type 2. Production
of Ang II and stimulation of Ang II receptor type 1 are responsible for vasoconstriction and endothelial dysfunction in
models of systemic inflammation with
endotoxin. Together with the observation
that pulmonary Ang II was up-regulated
in patients with acute respiratory distress
syndrome (ARDS), inhibition of RAS became a potential treatment for these
acute inflammatory disorders (2, 3). The
discovery of a new homolog termed angiotensin-converting enzyme 2 (ACE2)
revived interest and supported a potential
role for RAS in the pathogenesis of lung
inflammation. By counterbalancing ACE
and reducing Ang II levels, ACE2 can play
a protective role in sepsis-induced ARDS
(4). These pathways are shown in Figure
1. Polymorphisms of the ACE gene were
also the first to be associated with susceptibility and outcome of ARDS (5).
In this issue of Critical Care Medicine,
Haqiwara et al (6) report a beneficial role
for ACE inhibition with enalapril on systemic inflammation and pulmonary in-
*See also p. 626.
Key Words: angiotensin; angiotensin-converting
enzyme; angiotensin-converting enzyme inhibitor;
acute lung injury; acute respiratory distress syndrome;
sepsis; inflammation
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194cfa6
776
Angiotensinogen
Renin
Ang-(1-9)
Ang I
ACE2
ACE-I
Ang-(1-7)
Ang II
ACE
AT1R
AT2R
vasodilation
•Inflammatory mediators
•cell adhesion
•cell growth
cytoskeletal
rearrangement
Vascular permeability vasoconstriction
inflammation
remoddeling
INJURY
Figure 1. The pathways by which angiotensinogen system can participate in the regulation of vascular
permeability, vasoconstriction, inflammation, remodeling, and vasodilation. As described in the text of
the editorial, Ang II receptor type 1 (AT1R) and Ang II receptor type 2 (AT2R) identify the known
receptors for angiotensin II (Ang II). ACE, angiotensin-converting enzyme; Ang I, angiotensin I.
jury using a rat model of intraperitoneal
endotoxin-induced inflammation. Enalapril was administered before endotoxin
exposure. The findings in this study agree
with the protective role of ACE inhibition
reported in models of ventilator-associated and oleic acid-induced lung injury.
In these models, ACE inhibition attenuated pulmonary Ang II production with a
reduction of 1) inflammatory cytokines;
2) procoagulant effects; and 3) endothelial and epithelial apoptosis (7, 8).
After endotoxin administration in the
study by Haqiwara et al, the investigators
measured increased systemic levels of
Ang II that were attenuated by ACE inhibition with enalapril. However, there was
no measure of systemic or local pulmonary ACE activity. In the study by Idell et
al (9), ACE levels were increased in bronchoalveolar lavage fluid from patients
with sepsis-associated ARDS, and plasma
ACE activity was decreased in these patients. Animal models (10) of ventilator
induced lung injury also demonstrated
increased ACE activity in bronchoalveolar
lavage fluid. However, in other models of
lung injury, pulmonary endothelial ACE
activity (assessed by in vivo dilution
methods) was reduced as a result of enzyme downregulation caused by reactive
oxygen species, as reported by Orfanos et
al (11) in patients with ARDS. These findings indicate that it might be important
to consider compartmentalization of RAS
to understand its pathophysiology.
To identify the cellular and molecular
mechanisms of ACE inhibition in their
model of systemic inflammation, the authors focused on the role of macrophages. Besides endothelial cells, inflammatory cells are equipped with all the
components of RAS and can produce Ang
II. The key finding that enalapril supCrit Care Med 2009 Vol. 37, No. 2
presses nuclear factor kappa B activation
is important, given the central role of
nuclear factor kappa B in the regulation
of proinflammatory and proapoptic
genes. This finding is supported by the
systemic reduction of tumor necrosis factor-␣ and interleukin-6 with enalapril
treatment. The reduction in high mobility group box protein 1 in serum, lung
tissue, and macrophage supernatant was
also associated with enalapril treatment.
The mechanistic insights in this study
are not complete because the investigators did not study the effect of ACE inhibition on other cell types in this model.
Others have provided in vitro evidence
that Ang II induces apoptosis in alveolar
epithelial cells (12). Altered myeloperoxidase activity suggests effects on endothelial cell adhesion molecules and/or direct
neutrophil effects. Also, transcription factors such as activation protein-1 and the
production of proinflammatory mediators might provide additional insights
into the role of RAS during acute inflammatory changes. The primary site of action of Ang II in this model is still unclear. Exploration of specific interactions
with Ang II receptor type 1, as has been
done in other injury models (13), or receptors of the bradykinin system, might
be necessary.
A role for Ang II in acute lung injury
(ALI) may not be limited to the acute
phase. Locally produced Ang II also plays
a potential role in the fibroproliferative
phase of the disease. Inhibition of the
RAS system reduces procollagen production and transforming growth factor-
expression in human lung fibroblasts
(14). It would be interesting to study the
effect of early or late ACE inhibition on
the fibroproliferative response in ALI.
Additional preclinical animal studies
will be necessary to evaluate the potential
clinical value of ACE inhibition in sepsis
and ALI/ARDS. The potential systemic
side effects of vasodilation and systemic
hypotension need to be investigated, and
dose–response effects and drug admission
Crit Care Med 2009 Vol. 37, No. 2
after exposure to the insult will need to
be tested. One clinical study that reported
outpatient use of ACE inhibitors reduced
30-day mortality for patients with community-acquired pneumonia (15).
In summary, the findings of this experimental study should stimulate further preclinical studies to test the potential value of ACE inhibition as a treatment
in infectious and noninfectious models of
ALI (16, 17). If the results of these experimental studies continue to be promising,
then a phase II clinical trial may be warranted with ACE inhibition for early ALI/
ARDS.
Arne P. Neyrinck, MD
Cardiovascular Research Institute
University of California
San Francisco, CA
Michael A. Matthay, MD
Cardiovascular Research Institute
University of California
San Francisco, CA
Departments of Anesthesia and
Medicine
University of California
San Francisco, CA
REFERENCES
1. Imai Y, Kuba K, Penninger JM: The discovery
of angiotensin-converting enzyme 2 and its
role in acute lung injury in mice. Exp
Physiol 2008; 93:543–548
2. Lund DD, Brooks RM, Faraci FM, et al: Role
of angiotensin II in endothelial dysfunction
induced by lipopolysaccharide in mice. Am J
Physiol Heart Circ Physiol 2007; 293:
H3726 –H3731
3. Wiel E, Pu Q, Leclerc J, et al: Effects of the
angiotensin-converting enzyme inhibitor
perindopril on endothelial injury and hemostasis in rabbit endotoxic shock. Intensive
Care Med 2007; 30:1653–1659
4. Imai Y, Kuba K, Rao S, et al: Angiotensinconverting enzyme 2 protects from severe
acute lung failure. Nature 2005; 436:
112–116
5. Marshall RP, Webb S, Bellingan GJ, et al:
Angiotensin converting enzyme insertion/
deletion polymorphism is associated with
susceptibility and outcome in acute respira-
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
tory distress syndrome. Am J Respir Crit
Care Med 2002; 166:646 – 650
Hagiwara S, Iwasaka H, Matumoto S, et al:
Effects of an angiotensin-converting enzyme
inhibitor on the inflammatory response in in
vivo and in vitro models. Crit Care Med 2009;
37:626 – 633
Chen CM, Chou HC, Wang LF, et al: Captopril decreases plasminogen activator inhibitor-1 in rats with ventilator-induced lung
injury. Crit Care Med 2008; 36:1880 –1885
He X, Han B, Mura M, et al: Angiotensinconverting enzyme inhibitor captopril prevents oleic acid-induced severe acute lung
injury in rats. Shock 2007; 28:106 –111
Idell S, Kueppers F, Lippmann M, et al: Angiotensin converting enzyme in bronchoalveolar lavage in ARDS. Chest 1987; 91:52–56
Wösten-van Asperen RM, Lutter R, Haitsma
JJ, et al: ACE mediates ventilator-induced
lung injury in rats via angiotensin II but not
bradykinin. Eur Respir J 2008; 31:363–371
Orfanos SE, Armaganidis A, Glynos C, et al:
Pulmonary capillary endothelium-bound angiotensin-converting enzyme activity in
acute lung injury. Circulation 2000; 102:
2011–2018
Wang R, Zagariya A, Ibarra-Sunga O, et al:
Angiotensin II induces apoptosis in human
and rat alveolar epithelial cells. Am J Physiol
Lung Cell Mol Physiol 1999; 276:L885–L889
Chan YC, Leung PS: AT1 receptor antagonism ameliorates acute pancreatitis-associated pulmonary injury. Regul Pept 2006;
134:46 –53
Marshall RP, Gohlke P, Chambers RC, et al:
Angiotensin II and the fibroproliferative response to acute lung injury. Am J Physiol
Lung Cell Mol Physiol 2004; 286:L156 –L164
Mortensen EM, Restrepo MI, Anzueto A, et al:
The impact of prior outpatient ACE inhibitor
use on 30-day mortality for patients hospitalized with community-acquired pneumonia. BMC Pulm Med 2005; 13:5–12
Su X, Robriquet L, Folkesson HG, et al: Protective effect of endogenous beta-adrenergic
tone on lung fluid balance in acute bacterial
pneumonia in mice. Am J Physiol Lung Cell
Mol Physiol 2006; 290:L769 –L776
Looney MR, Su X, Van Ziffle JA: Neutrophils
and their Fc gamma receptors are essential
in a mouse model of transfusion-related
acute lung injury. J Clin Invest 2006; 116:
1615–1623
777
A new approach to step on the vagal anti-inflammatory gas pedal*
I
n response to infection, the central nervous system initiates several anti-inflammatory pathways,
designed to prevent the detrimental effects of the uncontrolled release of
inflammatory mediators. One of these
pathways involves an increased activity in
the efferent vagus nerve, called the “nicotinic anti-inflammatory pathway,”
which reflexively modifies the inflammatory response (1, 2). The most compelling
evidence for role of the cholinergic nervous system in the regulation of inflammation is derived from studies on rodents
challenged with endotoxin, the proinflammatory component of the outer
membrane of Gram-negative bacteria (3,
4). In these studies, vagotomy led to enhanced systemic tumor necrosis factor-␣
production and accelerated the development of shock; in turn, electrical stimulation of the efferent vagus nerve downregulated tumor necrosis factor-␣
production and protected animals from
hypotension (3). These findings were
later confirmed and expanded in animal
models of sepsis, pneumonia, and pancreatitis (5–9). Further studies showed
that the anti-inflammatory properties of
the efferent vagus nerve are mediated
through its major neurotransmitter acetylcholine (Ach), which interacts with
nicotinic Ach receptors on macrophages
resulting in inhibition of endotoxininduced responses (3, 4), showing that
the nicotinic Ach receptor (and not the
muscarinic Ach receptor) and, specifically, the alpha7 subunit are required for
this effect (4). The molecular target for
this pathway has been identified as it
has been shown that the vagal antiinflammatory pathway acts by alpha7
subunit-mediated Jak2-STAT3 activation in macrophages (10). On the basis
of these studies, it has been suggested
*See also p. 634.
Key Words: vagus nerve; sepsis; endotoxin shock;
anti-inflammatory; anisodamine
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194d004
778
that stimulation of nicotinic acetylcholine receptors might be beneficial in
clinical syndromes involving overshoot
inflammation such as septic shock, inflammatory bowel disease, or rheumatoid arthritis. Indeed in several studies,
administration of nicotine or specific
nicotinic Ach agonists, such as GTS-21,
have been shown to reduce inflammation in various animal models (4, 5, 7).
In this issue of Critical Care Medicine,
Liu et al (11) evaluated the effect of
pretreatment with anisodamine, a muscarinic receptor antagonist, in rodent
models of endotoxic shock. This is a
surprising approach because, as stated
above, all reported studies on antiinflammatory compounds derived from
the theory of the vagal anti-inflammatory
reflex have involved ligands for nicotinic
Ach receptors. Anisodamine is a compound that is derived from a Chinese
medicinal herb and is chemically related
to atropine. The authors state that anisodamine is widely used in the treatment of
septic shock in China, although evidence
from clinical studies is lacking. However,
on the basis of their results there might
be some truth and rationale behind the
myth.
In their report, Liu et al (11) show that
blockade of muscarinic receptors using
anisodamine results in an anti-inflammatory phenotype in endotoxin shock. Furthermore, they show that this antiinflammatory phenotype can be reversed
when nicotinic receptors are blocked
concurrently with muscarinic blockade.
The authors hypothesize that muscarinic
receptor blockade results in rerouting of
Ach to nicotinic receptors resulting in
increased Ach-mediated activation of nicotinic receptors and activation of the nicotinic anti-inflammatory pathway. These
results are strengthened by studies involving vagotomy as well as alpha7
knockout mice because the beneficial effects of anisodamine are absent in vagotomized mice as well as alpha7 knockouts.
Using in vitro studies, the authors show
that indeed the binding of the selective
alpha7 selective agonist bungarotoxin is
increased in anisodamine-treated macrophages.
Taken together, the authors demonstrate that pretreatment with anidosamine results in an anti-inflammatory
phenotype, decreases shock, and increases survival. The observed effects of
anisodamine are probably unrelated to
muscarinic receptors per se but intimately linked to an alpha7 nicotine receptor-dependent pathway suggesting
that a secondary enhanced activation of
this pathway is a result of blockade of
muscarinic receptors. The exact mechanism, however, can only be suggested
based on the presented data and, therefore, remains to be elucidated. The study
confirms the potential beneficial effects
of anisodamine in the treatment of shock.
However, an endotoxin model was used,
and in the intensive care setting, septic
shock is a different ballgame because it
involves systemic circulation of live bacteria. An anti-inflammatory phenotype in
genuine sepsis might result in enhanced
bacterial load that may be clinically relevant. Although this study suggests that
anisodamine might be of use in clinical
sepsis, one should be very cautious and
remember that no anti-inflammatory intervention has been shown to be beneficial in human studies. Therefore, I
strongly feel that the use of this compound in septic shock, which as the authors state, is quite a common practice in
China, cannot be recommended. To evaluate the clinical potential in sepsis, I
would be interested in the phenotype of
anisodamine treatment observed in animal models of sepsis such as cecal ligation and puncture. The most important
question in studies involving the vagal
anti-inflammatory reflex and pertaining
to the location of the “immune synapse”
remains. The exact location where the
physiologic interaction between vagus
nerve, derived acetylcholine and immune
cells takes place has not been elucidated
so far, although the spleen has been suggested (12, 13). Finally, this study suggests that activation of the vagal immune
reflex can not only be pursued by stimulation of the vagus nerve or nicotinic
receptors but also by inhibition of muscarinic receptors, which results in secondary activation of the pathway. The auCrit Care Med 2009 Vol. 37, No. 2
thors suggest a new approach to activate
the vagal anti-inflammatory pathway,
which is very interesting and needs further study on the molecular and preclinical level. Their findings are new and expand our possibilities of using the vagal
anti-inflammatory pathway for clinical
benefit because we now know that stepping on the muscarinic brake results in
stepping on the vagal anti-inflammatory
gas pedal.
David J. van Westerloo, MD, PhD
Department of Intensive
Care Medicine
Academic Medical Centre
University of Amsterdam
Amsterdam, The Netherlands
REFERENCES
1. Tracey KJ: The inflammatory reflex. Nature
2002; 420:853– 859
2. Blalock JE: Harnessing a neural-immune
circuit to control inflammation and shock.
J Exp Med 2002; 195:F25–F28
3. Borovikova LV, Ivanova S, Zhang M, et al:
Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin.
Nature 2000; 405:458 – 462
4. Wang H, Yu M, Ochani M, et al: Nicotinic
acetylcholine receptor alpha7 subunit is an
essential regulator of inflammation. Nature
2003; 421:384 –388
5. Giebelen IA, van Westerloo DJ, LaRosa GJ, et
al: Local stimulation of alpha7 cholinergic
receptors inhibits LPS-induced TNF-alpha
release in the mouse lung. Shock 2007; 28:
700 –703
6. Giebelen IA, van Westerloo DJ, LaRosa GJ, et
al: Stimulation of alpha 7 cholinergic receptors inhibits lipopolysaccharide-induced
neutrophil recruitment by a tumor necrosis
factor alpha-independent mechanism. Shock
2007; 27:443– 447
7. van Westerloo DJ, Giebelen IA, Florquin S, et
al: The vagus nerve and nicotinic receptors
modulate experimental pancreatitis severity
in mice. Gastroenterology 2006; 130:
1822–1830
8. van Westerloo DJ, Giebelen IA, Florquin S, et
al: The cholinergic anti-inflammatory pathway regulates the host response during sep-
9.
10.
11.
12.
13.
tic peritonitis. J Infect Dis 2005; 191:
2138 –2148
van Westerloo DJ, Giebelen IA, Meijers JC, et
al: Vagus nerve stimulation inhibits activation of coagulation and fibrinolysis during
endotoxemia in rats. J Thromb Haemost
2006; 4:1997–2002
de Jonge WJ, van der Zanden EP, The FO, et
al: Stimulation of the vagus nerve attenuates
macrophage activation by activating the
Jak2-STAT3 signaling pathway. Nat Immunol 2005; 6:844 – 851
Liu C, Shen F-M, Le YY, et al: Antishock
effect of anisodamine involves a novel pathway for activating ␣7 nicotinic acetylcholine
receptor. Crit Care Med 2009; 37:634 – 641
Rosas-Ballina M, Ochani M, Parrish WR, et
al: Splenic nerve is required for cholinergic
antiinflammatory pathway control of TNF in
endotoxemia. Proc Natl Acad Sci USA 2008;
105:11008 –11013
Huston JM, Ochani M, Rosas-Ballina M, et al:
Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis. J Exp Med
2006; 203:1623–1628
Role for bacteriophages in the preparedness for fighting against
pneumonia in the postantibiotic era*
T
he discovery of penicillin and
the rise of antibiotic drugs
changed the nature of medicine. Antibiotics, which actually killed bacteria, were hailed as “miracle drugs” (1), able to strike at the “root”
cause of diseases like pneumonia, tuberculosis, malaria, syphilis, and gonorrhea.
Unfortunately, bacteria soon demonstrated that they were able to become
resistant to different antibiotics and, in
turn, to pass their resistance genes onto
their descendants, eventually producing
populations of bacteria could survive even
the strongest drugs. As a consequence,
pneumonia that was once thought could be
eradicated remained a persistent problem.
*See also p. 642.
Key Words: pneumococcal pneumonia; bacteriophage; endolysin; Cpl-1; mouse; postantibiotic era
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194fe0d
Crit Care Med 2009 Vol. 37, No. 2
Pneumonia is a leading cause of death,
particularly in children and elderly. The
use of antibiotics and advances in health
care lessened the risk; however, pneumonia remained the first cause of death
among respiratory and infectious diseases
globally. Streptococcus pneumoniae is
the commonest etiology of mild, moderate, and severe community-acquired
pneumonia. There is an increase in antibiotic resistance of pneumococci limiting
the number of antimicrobial agents that
produce reliable treatment results for
these infections. Other microorganisms
producing community-acquired or nosocomial pneumonia show the same resistance acquisition problem. Newer antimicrobials and combination therapy by
antimicrobials with different mechanisms of action have been used to treat
infections for decades with the goal of
producing a wider spectrum of action,
preventing the emergence of drugresistant subpopulations, reducing the
dose of a single agent, or achieving a
synergistic effect; however, this use of
antibiotics promoted the development of
increasing resistance. This phenomenon
has been named the antibiotic paradox—
miracle drugs are destroying the miracle
(2). The ways to face the antimicrobials
resistance challenge include the increase
of our efforts to preserve the activity of
available antibiotics, or at least expand, as
much as possible, the period of their use,
while intense research efforts should be
focused on the development and introduction of new antimicrobial agents into
clinical practice. However, the problem
generated with the evolving antimicrobial
resistance in Pseudomonas aeruginosa,
Acinetobacter baumannii, Staphylococcus
aureus, and Klebsiella pneumoniae has led
to the emergence of clinical isolates susceptible to only one class of antimicrobial
agents and eventually to pandrug-resistant
(i.e., resistant to all available antibiotics)
isolates.
Bacteriophages or phages, so called
bacteria-killing viruses, described at the
beginning of the 20th century and characterized by Delbrück, Luria, and Hershey who were awarded the novel prize in
1969 for their discoveries (3), have been
779
used successfully in treatments against
antibiotic-resistant bacteria in various infections, including pneumonia. These
data have come from clinical trials in
Eastern Europe, mostly uncontrolled.
However, recent findings in well-controlled animal models demonstrating
that phages can rescue animals from a
variety of fatal infections produced by S.
pneumoniae, S. aureus, and P. aeruginosa are encouraging (4 – 6). Therapeutic
phages appear to kill their target bacteria
by replicating inside and lysing the host
cell via a lytic cycle; endolysins are lytic
enzymes that ensure phages’ progeny
survival, by selecting essential cell wall
components as target for destruction to
avoid phages’ breed from being locked up
inside the bacteria (7, 8). However, lysis
of host bacteria by a lytic phage is a
complex process consisting of a cascade
of events involving several structural and
regulatory genes, and it is possible that
some therapeutic phages have some
unique yet unidentified genes or mechanisms responsible for their ability to effectively lyse their target bacteria (9). The
therapeutic and prophylactic application
of phages is now experiencing a renaissance of interest.
In this issue of Critical Care Medicine,
Witzenrath et al (10) explored, in an experimental study on a mouse model of
severe pneumococcal pneumonia, the
therapeutic potential of Cpl-1, a purified
bacteriophage endolysin, which specifically kills pneumococci on contact. They
observed that when treatment was commenced 24 hours after experimental infection, 100% Cpl-1-treated mice survived the otherwise fatal pneumonia and
showed rapid recovery. When treatment
was started 48 hours after infection, mice
had developed bacteremia, and three of
seven Cpl-1-treated animals (42%) survived. Cpl-1 dramatically reduced pulmonary bacterial counts, and prevented bacteremia, systemic hypotension, and
lactate increase when treatment commenced at 24 hours. In vivo, treatment
780
with Cpl-1 effectively reduced counts of
penicillin-susceptible pneumococci. The
inflammatory response in Cpl-1-treated
mice, as determined by multiplex cytokine assay of lung and blood samples, was
lower than in untreated mice. They concluded that Cpl-1 may provide a new
therapeutic option in the treatment of
pneumococcal pneumonia.
This study adds new information
about the effectiveness of purified recombinant bacteriophage lytic enzymes, and
specifically on the role of Cpl-1 for the
control of diseases caused by S. pneumoniae. These findings warrant research
into further therapeutic uses of this novel
technology to advance its application in
the clinical setting.
Unfortunately, from the microbiological standpoint, pneumonia is a complex
problem that cannot be immediately derived from an animal model to the patients’ presentation in the real world.
Cpl-1, as most of the lytic enzymes, are
targeted to specific pathogenic bacteria.
It is difficult to ascertain, based on clinical or rapid microbiological techniques,
which is the etiology of pneumonia; additionally, if more than one pathogen is involved, as happen, in about 10% to 50% of
pneumonias, it is not clear if in these cases
a “cocktail” of different lytic enzymes or
bacteriophages could be used (11).
The postantibiotic era is more than a
hypothesis; actually, pandrug-resistant
bacteria are among us, producing severe
nosocomial pneumonia and other kinds
of infections in the healthcare setting.
The strategies to face this problem include the use of novel therapies to fight
against bacterial infections including
phage therapy, immunomodulation, nonbacterial killing effects of antibiotics, and
improved preventive measures. There is
still a great lack of formal large, scale
clinical-studies on bacteriophages safety
and effectiveness; the lack of newer antibiotics requires reevaluating and reinvesting in bacteriophages. Preparedness
for the eventual lack of miracle drugs is
essential; studies exploring newer ways to
struggle against bacterial infections, like
the original investigation presented by
Witzenrath et al, should be encouraged.
Carlos M. Luna, MD, PhD
Didier Bruno, MD
Department of Medicine
Pulmonary Medicine Division
Hospital de Clinicas
Universidad de Buenos Aires
Argentina
REFERENCES
1. Parascandola J: From germs to genes: Trends
in drug therapy 1852–2002. Pharm Hist
2002, 44:3–11
2. Levy SB: The Antibiotic Paradox: How Miracle Drugs Are Destroying the Miracle. Vol. 1.
Plenum, New York, 1992
3. Pennazio S: The origin of phage virology.
Rivista di Biologia 2006; 99:103–129
4. Loeffler JM, Djurkovic S, Fischetti VA: Phage
lytic enzyme Cpl-1 as a novel antimicrobial
for pneumococcal bacteremia. Infect Immun
2003; 71:6199 – 6204
5. Watanabe R, Matsumoto T, Sano G, et al:
Efficacy of bacteriophage therapy against
gut-derived sepsis caused by Pseudomonas
aeruginosa in mice. Antimicrob Agents Chemother 2007; 51:446 – 452
6. Wills QF, Kerrigan C, Soothill JS: Experimental bacteriophage protection against
Staphylococcus aureus abscesses in a rabbit
model. Antimicrob Agents Chemother 2005;
49:1220 –1221
7. Fischetti VA: Bacteriophage lytic enzymes:
Novel anti-infectives. Trends Microbiol 2005;
13:491– 496
8. Young R: Bacteriophage lysis: Mechanism and
regulation. Microbiol Rev 1992; 56:430 – 481
9. Sulakvelidze A, Alavidze Z, Morris JG Jr: Bacteriophage therapy. Antimicrob Agents Chemother 2001; 45:649 – 659
10. Witzenrath M, Schmeck B, Doehn JM, et al:
Systemic use of the endolysin Cpl-1 rescues
mice with fatal pneumococcal pneumonia.
Crit Care Med 2009; 37:642– 649
11. Mc Vay CS, Velasquez M, Fralick JA: Phage
therapy of Pseudomonas aeruginosa infection in a mouse burn wound model. Antimicrob Agents Chemother 2007; 51:1934 –1938
Crit Care Med 2009 Vol. 37, No. 2
Functional hemodynamics and increased intra-abdominal pressure:
Same thresholds for different conditions . . .?*
W
hat this study tells us . . .
In this issue of Critical
Care Medicine, Renner et
al (1) report the results of
a study entitled “Influence of increased
intra-abdominal pressure on fluid responsiveness predicted by pulse pressure
variation and stroke volume variation
(SVV) in a porcine model.” On first sight,
it looks as if this study just repeats previously performed studies on SVV and
pulse pressure variation (PPV) during intra-abdominal hypertension (IAH) (2–5).
Is this really the case? The authors collected prospective data on 14 domestic
pigs during loading conditions and increased intra-abdominal pressure (IAP)
up to 25 mm Hg via CO2 pneumoperitoneum (PP). The application of PP increased the baseline values for SVV (from
9.6% ⫾ 3.2% to 16.3% ⫾ 5.9%) and PPV
(from 12.8% ⫾ 3.5% to 22% ⫾ 8.2%),
whereas global end-diastolic volume
(GEDV) decreased (from 836 ⫾ 210 mL
to 788 ⫾ 198 mL). After fluid loading, PP
still increased SVV (from 6.4% ⫾ 2.7% to
13.2% ⫾ 4.7%) and PPV (from 7.4% ⫾
1.6% to 15.7% ⫾ 4.5%). These changes
were correlated with changes in esophageal or intrathoracic pressure (ITP). They
also found that changes in PPV, SVV, and
GEDV showed significant correlations
with changes in SV after volume loading,
independent of IAP. The ability of PPV
and GEDV to predict fluid responsiveness
remained unchanged during PP, whereas
SVV lost this ability when IAP was increased up to 25 mm Hg. The bottom line
is that in conditions of increased ITP, as
*See also p. 650.
Key Words: abdominal pressure; abdominal compartment; preload; fluid responsiveness; functional
haemodynamics; stroke volume variation; pulse pressure variation
Dr. Malbrain has consulted for, received honoraria
from, and holds stock in Pulsion Medical Systems. Dr.
De laet has not disclosed any potential conflicts of
interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194c397
Crit Care Med 2009 Vol. 37, No. 2
is the case with IAH, different SVV and
PPV thresholds should be used to predict
fluid responsiveness.
What previous studies have shown . . .
Duperret et al (3) found similar results in
a pig model of abdominal banding with
IAP up to 30 mm Hg. A dose-related IAP
effect was suspected. In their model, the
systolic pressure variations (SPV), PPV,
and inferior vena cava flow fluctuations
were dependent on IAP values, which
caused changes in pleural pressure
swings, and this dependency was more
marked during hypovolemia. Although
the application of PP increased both PPV
and SPV, a statistical significant effect
was only observed for SPV. The increase
in SPV was mainly related to the ⌬up
component. Duperret et al (3) also looked
at left ventricular end-diastolic pressure
and left ventricular end-diastolic area and
found that the increase in the IAP induced a progressive increase in ITP and,
therefore, a relative hypovolemia owing
to a redistribution of blood volume. Although this factor is likely to play a role
at the highest values of IAP, a moderate
value of IAP was associated with an increase in thoracic blood volume, left ventricular end-diastolic area, and transmural left ventricular end-diastolic pressure,
suggesting an auto-transfusion effect.
Tournadre et al (5) found similar results for SPV in seven pigs undergoing
CO2 PP, but this time limited at 12 mm
Hg. The SPV increased whereas central venous pressure remained unchanged and
left ventricular end-diastolic area decreased. The 30% increase in SPV during
elevated IAP was mainly because of an increase in ⌬up component from 3.8% ⫾
5.2% to 7.3% ⫾ 5.5%. Volume loading with
hydroxylethyl starch (10 mL/kg) during PP
resulted in a decrease in SPV owing to a
decrease in ⌬up to 6% ⫾ 3.1%.
In a similar study by Bliacheriene et al
(2) in 11 rabbits undergoing CO2 PP with
IAP increase up to 10 mm Hg, SPV increased more than PPV. The PP in combination with hypovolemia induced by
hemorrhage further increased SPV and
PPV, confirming the superiority of PPV
over SPV to predict “fluid responsiveness.” The increase in SPV by PP was also
caused by an increase in ⌬up from 2% ⫾
1% to 6.7% ⫾ 2%. This study had some
limitations as discussed previously (4).
In a last study, Valenza et al (6) looked
at the effects of Helium-PP up to 25 mm
Hg in 15 rats. The PP increased SVV from
8.7% ⫾ 3% to 17.9% ⫾ 8% together with
the central venous pressure whereas
GEDV decreased. Table 1 gives an overview of the different studies.
What this study adds . . . The changes
observed in SVV and PPV for a given IAP
are the most important findings of this
study. Together with the previously documented increases in SPV that were
mainly related to the ⌬up component,
possible mechanisms are suggested.
First, a change in aortic compliance and
an increase in aortic transmural pressure
induced by increased IAP (either via direct compression or increased vasomotor
tone). Second, errors in the measurement of dynamic indices in conditions of
increased IAP, or, if we assume that no
measurement errors are induced by IAP
then this implies that these indices do
not perform well during IAH (since SVV
did no longer predict fluid responsiveness). Third, changes in extramural pressure, ITP, or chest wall compliance.
This study was one of the first looking
at the abdominothoracic index of transmission (7). The authors found a 47%
transmission of the ⌬IAP to the lung and a
45% transmission to the thoracic compartment (Table 1). This is in accordance with
previous studies showing on average an abdominothoracic index of transmission of
50% and confirms that traditional filling
pressures are erroneously increased during
increased ITP/IAP (7, 8). Interestingly, the
abdominothoracic index of transmission
was higher in nonresponders.
The authors also looked at receiver
operating characteristic curves to identify
the best thresholds for predicting fluid
responsiveness at baseline and during PP.
The results confirmed that GEDV is not
781
Table 1. Overview of studies on functional hemodynamics during intra-abdominal hypertension
Stage
Animal
N
Intra-abdominal
hypertension
IAP-BL (mmHg)
IAP-PP
⌬IAP
CVP-BL (mmHg)
CVP-PP
⌬CVP
Preload-BL
Preload-PP
⌬preload
Paw-BL (cmH2O)
Paw-PP
⌬Paw
⌬ITP
ATI-thor
ATI-car
ATI-lung
SPV-BL (%)
SPV-PP
⌬SPV
SVV-BL (%)
SVV-PP
⌬SVV
PPV-BL (%)
PPV-PP
⌬PPV
Tournadre
et al (5)
Bliacherine
et al (2)
Bliacherine
et al (2)
Duperret
et al (3)
Duperret
et al (3)
Valenza
et al (6)
Renner
et al (1)
Renner
et al (1)
NormoV
Pig
7
CO2
NormoV
Rabbit
11
CO2
HypoV
NormoV
Pig
7
Banding
HypoV
NormoV
Rat
15
Helium
NormoV
Pig
14
CO2
Fluid
0
25
25
0
25
25
2
25
23
7.7
12.7
5
290.4
267.2
–23.2
16.9
28.1
11.2
7
30.4%
21.7%
35.8%
6.5
25.5
19
6.2
11.1
4.9
836
788
–48
21.4
33.6
12.2
8.6
45.3%
25.8%
47.2%
7.2
26
18.8
10.2
14.4
4.2
1069
1034
–35
21.1
34.6
13.5
8.1
43.1%
22.3%
52.8%
8.7
17.9
9.2 (106%)
9.6
16.3
6.7 (70%)
12.8
22
9.2 (72%)
6.4
13.2
6.8 (106%)
7.4
15.7
8.3 (112%)
0
12
12
6b
7
1
9.9
9.5
–0.4
26
37
11
8.3%
67.4%
12.9
16.9
4 (31%)
0
10
10
0
10
10
15
12.5
–2.5
13.4
23.5
10.1
74.3%
8.5
13.3
4.8 (56%)
9.5
11.1
1.6 (17%)
13.4
23.4
10
73.5%
13.3
19.9
6.6 (50%)
11.1
24.9
13.8 (124%)
15
60.0%
14
56.0%
6
17.1
11.1 (185%)
4.1
12.2
8.1 (198%)
3.1
8.1
5 (161%)
12.7
19.7
7 (55%)
7.7
11.8
4.1 (53%)
6.6
9.8
3.2 (48%)
Meana
10.8
2.0
19.8
17.9
7.5
11.3
3.8
731.8
696.4
–35.4
18.7
30.0
11.3
10.5
40.5%
19.6%
58.5%
10.7
17.4
6.7 (75.4%)
7.3
14.3
7.0 (106.5%)
8.4
15.3
6.9 (89.2%)
ATI-car, abdomino-thoracic index of transmission to the cardiovascular compartment (calculated as ⌬CVP divided by ⌬IAP); ATI-res, abdomino-thoracic
index of transmission to the lungs (calculated as ⌬Paw divided by ⌬IAP); ATI-thor, abdomino-thoracic index of transmission to the thoracic compartment
(calculated as ⌬ITP divided by ⌬IAP); BL, baseline values; CVP, central venous pressure; IAP, intra-abdominal pressure; ITP, intra thoracic pressure; Paw:
peak airway pressure; PP, pneumoperitoneum; PPV, pulse pressure variation; SPV, systolic pressure variation; SVV, stroke volume variation.
Preload, left ventricular end diastolic area (in cm2) was used for the studies by Tournadre et al and Duperret et al, while global end diastolic volume
(in mL) was in the other studies.
a
Looking at the pooled data one can see that an increase in IAP (⌬IAP) of 17.9 mm Hg results in a ⌬CVP of 3.8 mm Hg, a ⌬ITP of 10.5 mm Hg, and
a ⌬Paw of 11.3 cm H2O. The average abdomino-thoracic index of transmission was 47% to the thorax, 20% to the cardiovascular space, and 58% to the
lungs (airway pressure). The bottom line is that dynamic indices like SPV, SVV, or PPV are not exclusively related to volaemia in the presence of increased
ITP or IAP; bin the study by Tournadre data was given for pulmonary artery occlusion pressure instead of CVP.
only a good static “volumetric” preload
parameter (far more superior than the
“barometric” indices of preload like central venous pressure or pulmonary artery
occlusion pressure) but also a good indicator of fluid responsiveness (equal to PPV),
regardless of IAP. In contrast to other studies, transmural filling pressures did not
perform better than the central venous
pressure and pulmonary artery occlusion
pressure values taken at end-expiration
(central venous pressureee) (9).
Interesting to see was that PPV (but
not SVV) kept its ability to predict fluid
responsiveness even at IAP levels of 25
mm Hg; however, receiver operating
characteristic curve analysis identified
20.5% as the best threshold for fluid
responsiveness (instead of the classic
12% and the 9.5% identified in this
study at baseline). This is an important
782
clinical message, meaning that we
probably cannot use the same thresholds for different conditions. The
threshold value will depend on the
amount of tidal volume, positive endexpiratory pressure application, or increased ITP and consequent changes in
pleural pressure and chest wall elastance, the presence of obesity, heart
failure with changes in right and left
ventricular preload and afterload, pulmonary hypertension, the use of a PP or
increased IAP . . . and may also differ in
children or neonates. However, it must
be said that because of the limited sample size the cut-off values only give a
rough estimation and need to be validated in a larger patient population.
This study showed that PPV performed
better than SVV derived from pulse contour analysis based on a complex algo-
rithm. This is a bit surprising because
PPV is a surrogate of SVV and the latter
should be less influenced by changes in
vasomotor tone. The present study suggests that changes in pulse contour due
to increased ITP may be more complex
than previously thought.
The study also showed that ⌬IAP correlates well with ⌬ITP and thus confirms
the rule of thumb to calculate the transmural filling pressure: central venous
pressuretm ⫽ central venous pressureee ⫺
IAP/2 (10).
What this study does not tell us . . . To
play the devil’s advocate one could argue
that the questions that the authors tried
to answer have been addressed previously. While the study was simple in its
concept, it turned out to be complex in
its execution and it may leave the average
Crit Care Med 2009 Vol. 37, No. 2
reader with more questions than it provides answers . . .
First, one could argue that the small
number of animals does not allow proper
receiver operating characteristic curve
analysis to define thresholds. The prediction of fluid responsiveness implies even
smaller subgroups of responders vs. nonresponders, proving that dynamic indices
work in animals with normal IAP is nothing new.
Second, the model for creating IAH,
namely a CO2 PP may have affected the
results. Gases are likely to have systemic
effects. Although helium seems quite inert,
argon has been observed to influence hemodynamic parameters, and CO2 can cause
a change in vasomotor tone (aortic compliance), partly explaining the observations
(11).
Third, this study was performed on
healthy pigs with normal cardiovascular
and respiratory function. Therefore, it remains questionable whether these results
can be extrapolated to a pathologic condition with a primary insult, capillary
leak, resuscitation, and tissue edema.
Finally, the study period and the timecourse between the different stages was
quite short (only 15 minutes). The current model therefore is one that mimics
“hyperacute” IAH rather than the clinically encountered “acute” or “subacte”
IAH (12).
What future animal studies should
look at . . . Despite the fact that the data
raises a lot of questions, this study is
important and the authors have to be
congratulated. Future studies should
look at the long-term effects (24 – 48
hours) of IAH at clinically relevant IAP
levels (15–20 mm Hg) on dynamic indices of fluid responsiveness during PP,
with and without hemorrhage and before
and after fluid loading. Future studies
should try to integrate these results with
Crit Care Med 2009 Vol. 37, No. 2
global indices of perfusion (lactate, base
deficit) and the presence of clinical overt
shock in animals and even better in patients who are critically ill. Furthermore,
they should also look at a good gold standard for SV measurement by means of an
ultrasonic flow probe in the aorta to exclude the possibility for possible mathematical coupling of data.
The results of this study confirm the
importance of IAH/abdominal compartment syndrome (13). The world society of
the abdominal compartment syndrome
(www.wsacs.org) invites interested researchers to join the society, to adhere to
the consensus definitions posted at the
Web site and to submit some prospective
data for the next world congress (www.
wcacs.org), to be held in Dublin, Ireland,
June 24 –27, 2009.
Manu L. N. G. Malbrain, MD,
PhD
Inneke de laet, MD
Department of Intensive
Care
ZiekenhuisNetwerk
Antwerpen
ZNA Stuivenberg, Belgium
REFERENCES
1. Renner J, Gruenewald M, Quaden R, et al:
Influence of increased intra-abdominal pressure on fluid responsiveness predicted by
pulse pressure variation and stroke volume
variation in a porcine model. Crit Care Med
2008; 37:650 – 658
2. Bliacheriene F, Machado SB, Fonseca EB,
et al: Pulse pressure variation as a tool to
detect hypovolaemia during pneumoperitoneum. Acta Anaesthesiol Scand 2007; 51:
1268 –1272
3. Duperret S, Lhuillier F, Piriou V, et al: Increased intra-abdominal pressure affects respiratory variations in arterial pressure in
normovolaemic and hypovolaemic mechanically ventilated healthy pigs. Intensive Care
Med 2007; 33:163–171
4. Malbrain ML, De Laet I: Functional haemodynamics during intra-abdominal hypertension: What to use and what not. Acta Anaesthesiol Scand 2008; 52:576 –577
5. Tournadre JP, Allaouchiche B, Cayrel V, et al:
Estimation of cardiac preload changes by systolic pressure variation in pigs undergoing
pneumoperitoneum. Acta Anaesthesiol Scand
2000; 44:231–235
6. Valenza F, Chevallard G, Porro GA, et al:
Static and dynamic components of esophageal and central venous pressure during intra-abdominal hypertension. Crit Care Med
2007; 35:1575–1581
7. Wauters J, Wilmer A, Valenza F: Abdominothoracic transmission during ACS: Facts and
figures. Acta Clin Belg Suppl 2007; 62:
200 –205
8. Malbrain ML, Wilmer A: The polycompartment syndrome: Towards an understanding
of the interactions between different compartments! Intensive Care Med 2007; 33:
1869 –1872
9. Ridings PC, Bloomfield GL, Blocher CR, et al:
Cardiopulmonary effects of raised intraabdominal pressure before and after intravascular volume expansion. J Trauma 1995; 39:
1071–1075
10. Cheatham ML, Malbrain ML: Cardiovascular
implications of abdominal compartment syndrome. Acta Clin Belg Suppl 2007; 62:
98 –112
11. Hazebroek EJ, Haitsma JJ, Lachmann B, et
al: Impact of carbon dioxide and helium insufflation on cardiorespiratory function during prolonged pneumoperitoneum in an experimental rat model. Surg Endosc 2002;
16:1073–1078
12. Malbrain ML, Cheatham ML, Kirkpatrick A,
et al: Results from the International Conference of Experts on intra-abdominal hypertension and abdominal compartment syndrome. I. Definitions. Intensive Care Med
2006; 32:1722–1732
13. Malbrain ML, De Laet I: AIDS is coming to
your ICU: Be prepared for acute bowel injury and acute intestinal distress syndrome. Intensive Care Med 2008; 34:
1565–1569
783
Designing clinical trials to improve neurobehavioral outcome after
traumatic brain injury: From bench to bedside*
T
raumatic brain injury (TBI), a
physiologic disruption of brain
structure and function resulting from the application of external physical force, is a problem of increasing concern within the medical
community, among healthcare policymakers, and to the general public. Between one and two million persons in the
United States suffer from TBI each year,
of which ⬃230,000 are hospitalized. Improvements in prehospital care (1) and
refinements in acute care management
strategies (2) make survival after TBI
more likely today than ever before. Despite improvements in survival after TBI,
however, the Centers for Disease Control
and Prevention estimate that ⬎80,000
Americans develop long-term TBI-related
disability each year and that about 3.2
million persons in the United States are
living with such disabilities (3, 4).
Mitigating the neurologic, neurobehavioral, and functional sequelae of
TBI, therefore, is an essential treatment
goal at all stages of postinjury care.
Unfortunately, the development of therapies that accomplish this goal has not
kept pace with those that improve TBI
survival. At least 21 multicentered randomized controlled trials of agents with
potential neuroprotective properties,
including N-methyl-D aspartate receptor antagonists, calcium channel blockers, magnesium sulfate, cannabinoids,
corticosteroids, aminosteroids, progesterone, cyclosporine, and inflammatory
modulators, among others, have been
conducted in the TBI population (5, 6).
None of these studies convincingly demonstrated benefit.
*See also p. 659.
Key Words: traumatic brain injury; neurobehavioral
function; memory; outcome; clinical trials
Supported, in part, by HealthONE Spalding Rehabilitation Hospital.
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e3181959e46
784
Interpreting the failures of these randomized controlled trials is a complicated
endeavor (6). The proportion of the variance in clinical outcome accounted for by
any general neuroprotective intervention
is likely to be relatively modest. When
investigated in groups that are heterogeneous with respect to TBI characteristics—particularly the presence, type, severity of intracranial abnormalities,
clinical severity, and baseline prognostic
risk—the likelihood of effecting change
through a single intervention is made
more modest still. Additionally, when the
outcome by which that intervention is
assessed is a global measure (such as the
Glasgow Outcome Scale [7]) dichotomized into “favorable” or “unfavorable”
categories, statistical power to detect
clinical neuroprotective effects is attenuated further.
The design of such randomized controlled trials and the selection of an outcome measure for use therein is more
than a matter of statistics. Several decades of improvements in survival after
TBI necessitate a shift, or at least an expansion, of focus from general measures
of TBI outcome to the most enduring and
difficult problem experienced by the majority of TBI survivors and their families:
neurobehavioral disturbances (8, 9).
These problems contribute substantially
to postinjury disability (10, 11) and
present major challenges for TBI survivors and their families throughout the
postinjury period (12, 13). Despite the
prevalence and functional relevance of
posttraumatic neurobehavioral disturbances, their incorporation into the
design of randomized controlled trials
in TBI and stroke (14) is limited, if and
when such problems are considered
at all.
In this issue of Critical Care Medicine,
Longhi et al (15) present findings from an
experimental injury study in which neurobehavioral and neuropathological outcomes are afforded equal consideration.
Recognizing the complement system as a
contributor to the pathobiology of TBI,
they investigated the effects of C1inhibitor (C1-INH)—an endogenous
serine-protease inhibitor of complement
activation within the classic, alternative,
and lectin pathways— given 10 minutes
or 1 hour postinjury on neurobehavioral sequelae and histologic damage in
a controlled cortical impact mouse
model of TBI.
The selection of both this agent and
this particular model of TBI are important initial considerations: the activation
of complement and associated inflammatory responses are substantial contributors to brain injury in the setting of cerebral contusion, but are less relevant in
the absence of such (i.e., in the setting of
diffuse axonal injury alone). In addition
to defining a subcategory of TBI in which
the agent selected might be useful, the
agent itself affects several elements of the
complement cascade. Its multiplicity of
effects increases the likelihood of attenuating injury with this single agent. Additionally, the authors anchor their investigation to outcomes that more readily
model important clinical parameters
than do histologic measures alone: they
evaluate the effect of C1-INH on neurobehavioral function, including motor performance (as assessed by the composite
neuroscore) and cognitive ability (as assessed by the Morris Water Maze).
In this study, a single administration
of C1-INH at 10 minutes following controlled cortical impact attenuated posttraumatic motor and cognitive impairments, and reduced histologic damage
when assessed 4 weeks after injury. When
this agent was administered 1 hour
postinjury, motor deficits, but not cognitive function or histologic injury, were
attenuated significantly. The efficacy of
this agent and the timing of administration required to optimize its effects, if
such are replicated in additional studies,
remain uncertain. Nonetheless, the ability of a single administration of C1INH—when given at any point postinjury—to attenuate motor, cognitive, and
histologic abnormalities is noteworthy.
Crit Care Med 2009 Vol. 37, No. 2
Also intriguing is the dissociation between motor, cognitive, and histologic
outcomes following delayed C1-INH administration. The mechanism by which
this dissociation developed is not clear.
Nonetheless, its occurrence echoes the
importance of expanding the concept of
outcome beyond those used conventionally in experimental injury research: if
only one of these outcomes was the sole
subject of this investigation, then a very
different set of the conclusions regarding
the possible promise of this agent would
be drawn.
Given the history of failed clinical investigations of neuroprotection in TBI,
skepticism regarding the future of the
line of investigation described by Longhi
et al would not be unexpected. The most
of which we can be certain is that replicating at the bedside the same types and
magnitudes of neuroprotective effects observed in the laboratory is an uncertain
endeavor, at best.
Independent of the future of the neuroprotective strategy described in this report, however, these investigators have
provided the field with a model for future
translational research in TBI. At the earliest stage of research, a subtype of injury
is identified in the service of reducing
heterogeneity and of developing a population-targeted therapy. An attempt is
made to identify an intervention that
might reasonably be expected to intervene on one or more aspects of the pathophysiology of that TBI subtype. If a single
agent is used, then one with multiple
mechanisms of action is selected to maximize the proportion of variance in out-
come for which it might be expected account. Finally, neurobehavioral function,
including both motor and cognitive performance, is incorporated into the research program at its earliest stages and
is afforded the same level of attention and
scrutiny as histopathology—an essential
first step in the improvement of neurobehavioral and functional outcomes of persons with TBI and their families.
David B. Arciniegas, MD
Brain Injury Rehabilitation Unit
HealthONE Spalding
Rehabilitation Hospital
Aurora, Colorado
Department of Psychiatry
Neurobehavioral Disorders
Program
University of Colorado School of
Medicine
Aurora, Colorado
6.
7.
8.
9.
10.
11.
REFERENCES
1. Gabriel EJ, Ghajar J, Jagoda A, et al: Brain
Trauma Foundation: Guidelines for prehospital managment of traumatic brain injury.
J Neurotrauma 2002; 19:111–174
2. Marion DW: Evidenced-based guidelines for
traumatic brain injuries. Prog Neurol Surg
2006; 19:171–196
3. Thurman D, Guerrero J: Trends in hospitalization associated with traumatic brain injury. JAMA 1999; 282:954 –957
4. Zaloshnja E, Miller E, Langlois JA, Selassie
AW: Prevalence of long-term disability from
traumatic brain injury in the civilian population of the United States, 2005. J Head
Trauma Rehabil 2008; 23:394 – 400
5. Beauchamp K, Mutlak H, Smith WR, et al:
Pharmacology of traumatic brain injury:
12.
13.
14.
15.
Where is the “golden bullet”? Mol Med 2008;
14:731–740
Maas AI, Marmarou A, Murray GD, et al:
Prognosis and clinical trial design in traumatic brain injury: The IMPACT study.
J Neurotrauma 2007; 24:232–238
Jennett B, Bond M: Assessment of outcome
after severe brain damage. Lancet 1975;
1:480 – 484
Levin HS: Neurobehavioral outcome of
closed head injury: Implications for clinical
trials. J Neurotrauma 1995; 12:601– 610
Levin HS, Gary HE Jr, Eisenberg HM, et al:
Neurobehavioral outcome 1 year after severe
head injury. Experience of the Traumatic
Coma Data Bank. J Neurosurg 1990; 73:
699 –709
Dikmen S, Machamer J, Miller B, et al: Functional status examination: A new instrument
for assessing outcome in traumatic brain injury. J Neurotrauma 2001; 18:127–140
Hall KM, Bushnik T, Lakisic-Kazazic B, et al:
Assessing traumatic brain injury outcome
measures for long-term follow-up of community-based individuals. Arch Phys Med Rehabil 2001; 82:367–374
Dikmen SS, Machamer JE, Powell JM, et al:
Outcome 3 to 5 years after moderate to severe traumatic brain injury. Arch Phys Med
Rehabil 2003; 84:1449 –1457
Groom KN, Shaw TG, O’Connor ME, et al:
Neurobehavioral symptoms and family functioning in traumatically brain-injured adults.
Arch Clin Neuropsychol 1998; 13:695–711
Anderson CA, Arciniegas DB, Filley CM:
Treatment of acute ischemic stroke: Does it
impact neuropsychiatric outcome? J Neuropsychiatry Clin Neurosci 2005; 17:486 – 488
Longhi L, Perego C, Ortolano F, et al: C1inhibitor attenuates neurobehavioral deficits
and reduces contusion volume after controlled cortical impact brain injury in mice.
Crit Care Med 2009; 37:659 – 665
Pediatric sepsis: Time is of the essence*
C
linical practice guidelines
have been widely accepted as a
tool to standardize care and
improve outcomes in critically ill patients. Periodic revision of
*See also p. 666.
Key Words: sepsis; pediatrics; guidelines; septic
shock; hemodynamic support
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e3181931210
Crit Care Med 2009 Vol. 37, No. 2
guidelines, especially those addressing
clinical conditions for which a large
number of new studies are published
each year, is critical to keeping such
guidelines up to date and relevant. Septic
shock is one of the most common disorders
managed in both adult and pediatric intensive care units, and one in which there is a
rapidly growing body of literature. In 2002,
the American College of Critical Care Medicine developed clinical practice parameters
for hemodynamic support of children and
neonates with septic shock (1). In this issue
of Critical Care Medicine, the American
College of Critical Care Medicine presents
the 2007 revision of these clinical practice
parameters, chaired by Dr. Joseph Carcillo
(2). This revision of the 2002 guidelines
incorporates data from new publications
over the 5-yr interval. Thirty interested parties, many but not all of whom are recognized experts in the field, participated in
the revision process. Recommendations included in the guidelines required that 90%
of the committee members agreed with the
recommendation. As with the 2002 guidelines, many of the recommendations have
limited support in the literature, but the
785
90% agreement requirement demonstrates
that there is broad clinical support for these
recommendations.
A growing body of literature has reported improved outcomes with the implementation of the 2002 guidelines (3,
4). This finding is consistent with the
current emphasis on standardization of
care to improve outcomes in the critically
ill. The 2007 revision includes new trials
relevant to both the initial resuscitation
and the ongoing hemodynamic management of children with septic shock. It
also includes information on new and developing technologies for hemodynamic
monitoring of these children. The rapid
development of new therapeutics and
technologies makes periodic revision of
these guidelines essential.
The Task Force revising the 2002
guidelines completed a thorough literature review. Overall, there are relatively
minor changes in the recommendations,
but there is increasing emphasis on the
importance of early rapid management.
There is continued emphasis on the first
hour fluid resuscitation and treatment
with vasopressors/inotropes. Antibiotic
administration within the first hour has
been added to the revised guidelines. It is
increasingly recognized that rapid early
fluid resuscitation and antibiotic administration are critical elements in the management of the patient with septic shock. The
Surviving Sepsis Campaign guidelines emphasize these points as well (5). Initial resuscitation goals for children and infants
are clinical goals, aiming for normalization
of the heart rate, blood pressure, capillary
refill, and mental status. The age-old question of colloids vs. crystalloids for initial
resuscitation remains unresolved; the clinician is free to select the fluid of his/her
preference. The 2007 revision of the pediatric sepsis guidelines does depart from the
conventional wisdom that catecholamines
786
should only be infused through central venous catheters. As a practical matter, it can
sometimes take a prolonged period of time
to establish central venous access in a small
child with shock. The 2007 guidelines include the recommendation from the American Heart Association Pediatric Advanced
Life Support guidelines to administer vasoactive drugs through a peripheral intravenous catheter until central access is attained, monitoring the peripheral access
site closely for infiltration (6).
The revised guidelines continue to recommend monitoring of cardiac index in
children with catecholamine-resistant
shock, and titrating therapy to a goal of
3.3– 6 L/min/m2. A new addition is that
there are several new techniques that can
be used instead of the pulmonary artery
catheter, including Doppler echocardiography, the PICCO catheter, or femoral artery
thermodilution catheter. Several new techniques are under investigation. None of
these techniques, however, is possible in
neonates and small infants, so the managing physician must continue to rely on
clinical goals.
The role of corticosteroid therapy remains uncertain. Hydrocortisone is recommended for children with absolute adrenal
insufficiency and catecholamine-resistant
shock, but the doses used in the literature
have varied enormously from 2 to 50 mg/
kg/day; no specific dose recommendations
can be made at this time. There continues
to be clinical equipoise regarding the use of
adjunctive steroid therapy for sepsis, outside the use in children with classic adrenal
or known hypothalamic-pituitary-adrenal
axis insufficiency.
The 2007 guidelines include stepwise
algorithms for children and for neonates
that will help the clinician at the bedside.
Like many guidelines, the guidelines may
not be as specific as some clinicians
might like, but that is a reflection of the
state of our knowledge, and not the quality of the guidelines. As we strive to decrease the mortality in children and infants with septic shock, we need to
ensure that they receive early aggressive
management with fluids, vasoactive
agents, and antibiotics. This study provides a framework that each institution
can use to develop its own specific guidelines to ensure this level of care.
Margaret M. Parker, MD, FCCM
Department of Pediatrics
Stony Brook University
Stony Brook, NY
REFERENCES
1. Carcillo JA, Fields AI, American College of
Critical Care Medicine Task Force Committee
members: Clinical practice parameters for hemodynamic support of pediatric and neonatal
patients in septic shock. Crit Care Med 2002;
30:1365–1378
2. Brierley J, Carcillo JA, Choong K, et al: Clinical
practice parameters for hemodynamic support
of pediatric and neonatal septic shock: 2007 update from the American College of Critical Care
Medicine. Crit Care Med 2009; 37:666 – 688
3. Han YY, Carcillo JA, Dragotta MA, et al: Early
reversal of pediatric-neonatal septic shock by
community physicians is associated with improved outcome. Pediatrics 2003; 112:793–799
4. de Oliveira CF, de Oliveira DS, Dottschald AF,
et al: ACCM/PALS haemodynamic guidelines
for paediatric septic shock: An outcomes comparison with and without monitoring central
venous oxygen saturation. Intensive Care Med
2008; 34:1065–1075
5. Dellinger RP, Levy MM, Carlet JM, et al: Surviving Sepsis Campaign: International guidelines for management of severe sepsis and
septic shock: 2008. Crit Care Med 2008; 36:
296 –327
6. International Liaison Committee on Resuscitation: The International Liaison Committee
on Resuscitation (ILCOR) consensus on science with treatment recommendations for pediatric and neonatal patients: Pediatric basic
and advanced life support. Pediatrics 2006;
117:e955– e977
Crit Care Med 2009 Vol. 37, No. 2
Brain oxidative stress after traumatic brain injury . . . cool it?*
H
ypothermia has been demonstrated to reduce intracranial pressure, decrease
excitotoxicity, attenuate oxidative stress, and limit the consumption
of protective antioxidant agents. Moderate hypothermia has demonstrated benefit and has been specifically recommended in adults after cardiac arrest and
in neonates who have sustained hypoxicischemic encephalopathy (1, 2). In children, moderate hypothermia has demonstrated efficacy in the control of intracranial
pressure after traumatic brain injury
(TBI), yet evidence of a survival or morbidity benefit has remained elusive (3, 4).
The failure of a recent, large, multinational, randomized controlled trial to
demonstrate the clinical benefit of hypothermia after TBI in children (5) has further diluted confidence that hypothermia
may improve survival and neurologic
outcome.
The brain is particularly sensitive to
oxidative injury due to metabolic demands and high rate of oxygen consumption. Evidence of oxidative stress after
TBI has been demonstrated via cerebrospinal fluid (CSF) analysis in animals (6),
children (7, 8), and adults (9). Evidence
of increased oxidative stress and disordered energy metabolism after TBI has
preceded refractory increases in intracranial pressure (10). This latter finding suggests a role for the analysis of CSF biomarkers as potential predictors of
clinical evolution and outcome and, in
particular, elevated CSF markers of oxidative damage (11).
The article by Bayir et al (12) in this
issue of Critical Care Medicine explored
the effect of moderate hypothermia on
markers of oxidative stress after TBI. A
cohort of pediatric patients were re-
*See also p. 689.
Key Words: traumatic brain injury; hypothermia;
therapy; oxidative stress; children
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194be10
Crit Care Med 2009 Vol. 37, No. 2
cruited in a single center for inclusion in
two larger multicenter studies investigating the role of therapeutic hypothermia
after TBI. CSF was obtained via external
ventricular drains and assayed for oxidative stress with measurements of antioxidant status, protein oxidation, and lipid
peroxidation. Children who had been randomized to moderate hypothermia were
shown to significantly preserve CSF antioxidant reserve, compared with children
in the normothermic group. Analysis of
the markers of free radical attack and
lipid peroxidation did not demonstrate a
similar beneficial profile in the hypothermic patients, although the authors do
suggest a trend to decreased protein oxidation in the hypothermia group. The
authors conclude that hypothermia attenuates brain oxidative stress after severe TBI in children. This reported investigation is intriguing because it
demonstrates a potential mechanism for
neuroprotection by therapeutic hypothermia. This study also demonstrates
the utility of CSF analysis for TBI prognosis and provides measurable factors
that could aid in identifying beneficial
therapeutic interventions. Importantly,
their work indicates that moderate hypothermia in the context of pediatric TBI is
not yet a therapeutic dead end.
The study by Bayir et al (12) is, however, limited by a small sample size, a
heterogeneous patient population, and
limited study protocol information. The
data presented also lack fidelity in a time
course critical for understanding the
pathophysiologic changes in oxidative
state after TBI and for determining a potential optimal therapeutic window. It is
also unclear if the preservation in oxidative state by therapeutic hypothermia reflects changes in compromised tissue,
compared with areas of relatively uninjured brain. Finally, although antioxidant
reserve is preserved by hypothermia, the
significance of the improved oxidant profile
is unclear. A causal relationship between
oxidative stress and outcome has not been
established. Despite these caveats, Bayir et
al (12) should be commended for their reported investigation. Enrolling children
with TBI in therapeutic trials is challenging, particularly when combined with external ventricular drain placement and regular CSF sampling for laboratory analysis
(13). This report indicates that regular laboratory analysis of CSF obtained via external ventricular drains may offer valuable
prognostic information and potentially
guide future therapeutic interventions.
Furthermore, this study is the first to demonstrate preserved antioxidant reserve in
children with therapeutic hypothermia.
Hypothermia-induced preservation
of the antioxidant profile may be beneficial in the prevention of delayed brain
injury after TBI. Excessive oxidative
stress damages lipids, proteins, and nucleic acids. Cytoskeleton and mitochondria disruption are consequences of oxidative stress, and, eventually, synaptic
plasticity and cognitive function are
impaired.
Similar to the action of therapeutic
hypothermia reported by Bayir et al (12),
anesthetic agents commonly administered after TBI may also provide protection against oxidative stress. Barbiturates
and propofol limit oxidative stress by
reducing the cerebral metabolic rate
and decreasing glucose and oxygen consumption (14). Anesthetics also induce
direct scavenging of reactive oxygen
species and inhibit lipid peroxidation
and excitotoxicity.
In conclusion, Bayir et al (12) who
have previously identified the increased
oxidative stress in the context of TBI now
report a beneficial effect of therapeutic
hypothermia on brain oxidative stress.
Their work indicates a likely mode of
secondary brain injury, and suggests that
hypothermia may offer therapeutic benefit. Further, they offer CSF biomarker
analyses as worthy of further evaluation
not only as an aid in prognostication, but
as an indicator of therapeutic efficacy. This
work by Bayir et al (12) and others will fuel
the emerging interest in drugs that have
potent antioxidative effects, such as agonists of the nuclear peroxisome proliferator-activated receptors (15), and suggest
that if therapeutic hypothermia has a role
to play in the treatment of TBI, it is not as
the magic bullet, but as one of a combina787
tion of therapies that act beneficially on
brain oxidation status.
Douglas D. Fraser, MD, PhD, FRCPC
Critical Care Medicine and
Paediatrics, University of
Western Ontario
London, Ontario, Canada
Physiology and Pharmacology,
University of Western Ontario
London, Ontario, Canada
Children’s Health Research
Institute
London, Ontario, Canada
Gavin Morrison, MRCP, DCH
Critical Care Medicine and
Paediatrics, University of
Western Ontario
London, Ontario, Canada
4.
5.
6.
7.
REFERENCES
1. Abate MG, Cadore B, Citerio G: Hypothermia
in adult neurocritical patients: A very “hot”
strategy not to be hibernated yet! Minerva
Anestesiol 2008; 74:425– 430
2. Polderman KH: Induced hypothermia and
fever control for prevention and treatment of
neurological injuries. Lancet 2008; 371:
1955–1969
3. Adelson PD, Ragheb J, Kanev P, et al: Phase
8.
9.
II clinical trial of moderate hypothermia after severe traumatic brain injury in children.
Neurosurgery 2005; 56:740 –754; discussion
740 –754
Biswas AK, Bruce DA, Sklar FH, et al: Treatment of acute traumatic brain injury in children with moderate hypothermia improves
intracranial hypertension. Crit Care Med
2002; 30:2742–2751
Hutchison JS, Ward RE, Lacroix J, et al:
Hypothermia therapy after traumatic brain
injury in children. N Engl J Med 2008; 358:
2447–2456
Ansari MA, Roberts KN, Scheff SW: Oxidative stress and modification of synaptic
proteins in hippocampus after traumatic
brain injury. Free Radic Biol Med 2008;
45:443– 452
Bayir H, Kagan VE, Tyurina YY, et al: Assessment of antioxidant reserves and oxidative
stress in cerebrospinal fluid after severe traumatic brain injury in infants and children.
Pediatr Res 2002; 51:571–578
Wagner AK, Bayir H, Ren D, et al: Relationships between cerebrospinal fluid markers of
excitotoxicity, ischemia, and oxidative damage after severe TBI: The impact of gender,
age, and hypothermia. J Neurotrauma 2004;
21:125–136
Bayir H, Marion DW, Puccio AM, et al:
Marked gender effect on lipid peroxidation
10.
11.
12.
13.
14.
15.
after severe traumatic brain injury in adult
patients. J Neurotrauma 2004; 21:1– 8
Cristofori L, Tavazzi B, Gambin R, et al:
Biochemical analysis of the cerebrospinal
fluid: Evidence for catastrophic energy failure and oxidative damage preceding brain
death in severe head injury: A case report.
Clin Biochem 2005; 38:97–100
Darwish RS, Amiridze N, Aarabi B: Nitrotyrosine as an oxidative stress marker: Evidence for involvement in neurologic outcome in human traumatic brain injury.
J Trauma 2007; 63:439 – 442
Bayir H, Adelson PD, Wisniewski SR, et al: Therapeutic hypothermia preserves antioxidant defenses after severe traumatic brain injury in infants and children. Crit Care Med 2009; 37:
689–695
Kochanek PM, Berger RP, Bayir H, et al:
Biomarkers of primary and evolving damage
in traumatic and ischemic brain injury: Diagnosis, prognosis, probing mechanisms,
and therapeutic decision making. Curr Opin
Crit Care 2008; 14:135–141
Wilson JX, Gelb AW: Free radicals, antioxidants, and neurologic injury: Possible relationship to cerebral protection by anesthetics.
J Neurosurg Anesthesiol 2002; 14:66 –79
Jennings JS, Gerber AM, Vallano ML: Pharmacological strategies for neuroprotection in
traumatic brain injury. Mini Rev Med Chem
2008; 8:689 –701
How possibly are we to choose albumin or hydroxyethyl starch?*
A
lbumin treatment of hypovolemia has been shown to be superior to crystalloid solutions
in virtually every pathologic
condition, including its use as part of the
pump prime for pediatric cardiopulmonary support. No known cases of infectious transmission have been reported
and the incidence of either urticaria or
anaphylactic shock is estimated at ⬍1
per10,000 (1). Unit cost is high and shortages could occur. It is with these facts in
mind that we evaluate the prospective,
randomized, and quasi-blinded trial of albumin vs. hydroxyethyl starch (HES)
130/0.4 in the perioperative volume ex-
*See also p. 696.
Key Words: albumin; hydroxyethyl starch; pediatric; volume expansion; cardiac surgery; editorial
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194de57
788
pansion during pediatric cardiac surgery
requiring cardiopulmonary bypass (2).
Hanart et al from Brussels, Belgium
painstakingly evaluated perioperative
fluid balance and blood loss in children
who received 4% albumin (n ⫽ 59) or 6%
HES 130/0.4 (n ⫽ 60). The volume of
colloid used and recorded blood loss was
similar between groups. Intraoperative
fluid balance was more positive in the
albumin group and the difference
reached significance. In addition, more
children in the albumin group received
blood transfusions but less fresh frozen
plasma. Taking the results at face value,
equivalence of albumin and HES 130/0.4
was the conclusion. Factoring in the differential treatment cost, a recommendation to
replace albumin with HES 130/0.4 for pediatric cardiac surgery seemed warranted.
However, I believe this was premature
without further study.
The authors are commended for a
well-controlled study. There are minor
flaws and bias that deserve discussion. In
the randomization scheme, the population was blocked for presence or absence
of cyanotic cardiac lesions and the type of
bypass circuit as determined by subject
weight. This led to a reasonable representation of age, diagnose, and similar intraoperative times in the groups. However,
the blinding did not include the intraoperative anesthesia team. Since the intraoperative team directed fluid treatment,
an element of unintentional bias was
possible. The authors were precise in
the blinded quantification of fluid balance and the types of blood products
used. However, there was less precision
in the postoperative care protocols, in
example, determining the need for ventilatory and inotropic support. I do not
think this affects evaluation of the primary end point, but evaluation of safety
is difficult.
Hanart et al, do discuss the possible
complications of starch-based colloid
solutions and discuss an evidence that
the newer formulations, of which HES
Crit Care Med 2009 Vol. 37, No. 2
130/0.4 is thought to be the safest, with
less severe clinical bleeding and renal
dysfunction. However, in their study, the
range in subject ages and diagnoses coupled with only minimal short-term safety
data make it impossible to determine even
an estimate of the risks in comparison with
albumin.
With respect to the altered coagulation caused by HES 130/0.4, this study
reports equivalence in blood loss; however, I do note that the HES 130/0.4
group received more fresh-frozen
plasma. Fresh-frozen plasma exposure
was protocol driven based on clinical
bleeding and prothrombin times so it is
probable that HES 130/0.4 did indeed
make a clinically significant, but controlled deterioration in coagulation.
There are reports of clinical bleeding
after use of HES 130/0.4 over a wide
range of uses (3).
Clear signals exist in the literature
that long lasting, osmotic renal damage
occurs in the face of prior renal disease
and exposure to colloid substitutes. There
is accumulation of the colloid within renal cells with effects that are both short
and long term (4). Renal failure does occur with HES 130/0.4 and it does not
seem to be 100% predicable based on
pretreatment renal function (5, 6).
In addition, long lasting and severe
pruritus can occur with HES 130/0.4
(7), and there is 41⁄2 times the incidence
of potentially life-threatening anaphylactic reactions compared with albumin
(3). HES colloids, in general, are relatively contraindicated in the face of sepsis where renal dysfunction and mortality is higher than with other colloids
(8).
Hanart et al report solid, but preliminary, data that HES 130/0.4 seems
equivalent to albumin within the first
24 hours after uncomplicated pediatric
cardiac surgery. Now we need an evaluation of long-term efficacy and safety
in a multicentered, randomized block
design with true blinding at all stages.
We are now challenged to take on the
question, “Is HES 130/0.4 equivalent in
efficacy and safety to albumin for pediatric congenital cardiac surgery?” before a fiscal decision voids a decision
based on quality of care.
Brian D. Hanna, MD, PhD
Division of Cardiology, Children’s
Hospital of Philadelphia
Pennsylvania, PA
Department of Pediatrics, School
of Medicine, University of
Pennsylvania
Pennsylvania, PA
REFERENCES
1. Albumin Product Insert, Octapharma, October
2006
2. Hanart C, Khalife M, De Villé A, et al: Perioperative
volume replacement in children undergoing cardiac surgery: Albumin versus hydroxyethyl starch
130/0.4. Crit Care Med 2009; 37:696–701
3. Wiedermann CJ: Hydroxyethyl starch—Can
the safety problems be ignored? Wien Klin
Wochenschr 2004; 116:583–594
4. Dickenmann M, Oettl T, Mihatsch MJ: Osmotic nephrosis: Acute kidney injury with accumulation of proximal tubular lysosomes
due to administration of exogenous solutes.
Am J Kidney Dis 2008; 51:491–503
5. Boldt J: New light on intravascular volume replacement regimens: What did we learn from the past
three years? Anesth Analg 2003; 97:1595–1604
6. Brunkorst FM, Oppert M: Nephrotoxicity on
hydroxyethyl starch solution. Br J Anaesth
2008; 100:856 – 857
7. Bork K: Pruritus precipitated by hydroxyethyl
starch: A review. Br J Dermatol 2005; 152:3–12
8. Weidermann CJ: Systematic review of randomized clinical trials on the use of hydroxyethyl starch for fluid management in sepsis.
BMC Emerg Med 2008; 8:1
Making catheter-related bloodstream infections history: From the
slogan to the serious strategy*
C
entral venous catheters (CVC)
have become an essential and
necessary component of the
modern management of critically ill patients. However, the benefits of
the CVC are often offset by the fact that
such devices have now been recognized
as the leading source of bloodstream infections in this critically ill patient population, which is associated with high
morbidity and mortality (1).
*See also p. 702.
Key Words: catheter-related bloodstream infection;
critically ill patients; anti-infective central venous catheters
The author has received honoraria from Cook and
royalties from Cook, AMX, Tyrx, and Horizon.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194c3dd
Crit Care Med 2009 Vol. 37, No. 2
For years, it has been recognized that
catheter-related bloodstream infection
(CRBSI) in critically ill patients is a preventable serious complication. In 1988,
Maki et al (2) predicted that “binding an
antimicrobial to the entire catheter surface may ultimately prove to be the most
effective technological innovation for reducing the risk of device-related infections.” Subsequently, Maki et al have
demonstrated that CVC impregnated with
antimicrobial agents are the “most intensively studied technology for the prevention of CRBSI over the past 30 years” and
have also shown that such anti-infective
CVC (AI-CVC) are highly cost-effective,
safe, and do not appear to select for resistance (3).
In the systematic review and metaanalysis by Hockenhull et al (4) published
in this issue of Critical Care Medicine, 38
prospective randomized controlled trials
of AI-CVC were evaluated. Meta-analysis
data from 27 trials have demonstrated a
strong treatment effect in favor of AI-CVC
(odds ratio: 0.49; 95% confidence interval: 0.3– 0.64). However, further subgroup analysis of different types of AICVC showed that the direction of the
treatment effect favored the antimicrobial-coated catheters in all subgroups, except for the benzalkonium chloride–
treated CVC. Further subanalysis
demonstrated that the minocycline/
rifampin coating was associated with the
most significant treatment effect (odds
ratio: 0.26; 95% confidence interval:
0.15– 0.47), whereas other antimicrobial
coatings varied as far as their efficacy
with the second-generation chlorhexidine/silver sulfadiazine (CHSS⫹) “just
failing to achieve statistical significance.”
789
The review and meta-analysis by
Hockenhull et al also demonstrated that
the AI-CVC is associated with a decrease
in medical costs. They have estimated a
cost savings in the United Kingdom for
every patient who receives an AI-CVC to
be equal to £138.2. This cost benefit is
consistent with various analyses that have
been demonstrated in multiple reviews
conducted in the United States (5, 6).
During the last two decades, various
infection control interventions have also
been shown to be effective in reducing
CRBSI, including the use of maximal
sterile barrier precautions during insertion of the CVC and applying chlorhexidine at the insertion site (7, 8). More
recently, Pronovost et al (9) have shown
that when these effective infection control interventions are used concurrently
as part of a “bundle” (that includes maximal sterile barrier precautions, hand
washing, cleaning the insertion site with
chlorhexidine, and avoidance of femoral
vein insertion and unnecessary prolonged
use of the catheter), a significant decrease in CRBSI is observed in critically
ill patients. Despite the fact that the
mean rate of CRBSI in the Pronovost et al
study was reduced from 7.7 per 1000
catheter days to 1.4 per 1000 catheter
days over a 6 –18-month study with a
reported median of zero, CRBSI continued to occur, despite the implementation
of such infection control interventions.
Although the Pronovost et al study
included a large number of intensive care
unit and critically ill patients, it demonstrated a significant and prolonged reduction in CRBSI that persisted for an 18month period. However, there were
several limitations to the study. Among
the limitations are the lack of assessment
of compliance as far as the implementation of the infection control bundle, the
crossover design of the study, the poor
definitions of CRBSI included in the
study, and the lack of assessment of confounding variables (such as the introduction of AI-CVC into the units being studied during the study period). Like
Pronovost et al, other investigators have
demonstrated that infection control interventions (such as the use of maximal
sterile barriers) are associated with a decrease in CRBSI (7, 10). However, such
measures on their own do not eliminate
CRBSI completely. Although it is now
well established that the infection control
bundle is the mainstay of preventing
CRBSI, it is also well recognized that
790
these measures are often associated with
high cost and poor compliance, are not
very durable, and do not completely eliminate or prevent infections (10).
Hockenhull et al highlight the fact
that “it is important to establish whether
the strong treatment effect of AI-CVC remains after effective infection control
bundles are established.” Two recent prospective randomized trials that are cited
by Hockenhull et al could shed light on
this issue (11, 12). In a multicenter prospective randomized study by Rupp et al
(11), the second-generation CHSS⫹ was
compared with uncoated CVC. During
the trial, the infection control bundle was
implemented into both arms of the study.
In the uncoated CVC control arm, where
such infection control bundle measures
were implemented, the rate of CRBSI was
1.24 per 1000 catheter days. However, in
the test arm, whereby the infection control bundles were implemented in addition to the use of AI-CVC (in this case
CHSS⫹), the risk of CRBSI was decreased by more than three-fold to a level
as low as 0.4 per 1000 catheter days (11).
Another prospective randomized trial by
Hanna et al (12) compared the use of an
AI-CVC coated with minocycline and rifampin with uncoated CVC. Again, in this
study, the elements of the infection control bundle were implemented, including
the maximal sterile barrier precautions.
In the uncoated CVC arm, the rate of
CRBSI was 1.28 per 1000 catheter days,
which was further reduced significantly
with the use of the AI-CVC (coated with
minocycline/rifampin) to a low level of
0.25 per 1000 catheter days (12). Hence,
the AI-CVC could complement the infection control bundle measures in further
bringing the rate of CRBSI to a very low
level approaching zero and, therefore,
making CRBSI a very rare entity in highrisk patients.
Hockenhull et al also referred to as
AI-CVC as a “safety net to prevent contaminating microorganisms from developing into CRBSI.” The referral of AICVC as a “safety net” is appropriate from
several view points. First, it is well recognized that the absolute and complete
compliance with all elements of the infection control bundle is never at a 100%
level. The fact that the CVC is transformed into an anti-infective device that
will prevent the microbial adherence of
resistant pathogens represents another
major barrier against biofilm colonization and, ultimately, CRBSI independent
of human behavior and lack of compliance with infection control measures.
Furthermore, the AI-CVC indeed serves
as a safety net in that the infection control bundle, including maximal sterile
barrier precautions and sterilization of
the skin insertion site with chlorhexidine, do prevent contamination of the
CVC during insertion. However, the AICVC do prevent biofilm colonization of
the external and the internal surface not
only during insertion but also subsequently during the dwell time of the catheter, where organisms could migrate
from the external skin insertion site surface or from the hub into the lumen of
the catheter.
In addition, impregnating the CVC
with antimicrobial agents is very much
similar to the concept of inoculating the
patient with a modified attenuated microbial organism through vaccination.
Whereas good infection control measures, including good hygiene, are important in preventing the transmission of
various infections, such as polio, measles,
and mumps, the use of effective vaccines
does, indeed, represent an important
safety net to further eliminate such infections (13). Appropriate infection control
precautions do not substitute, but rather
complement, vaccination as an intervention. The same is true for the AI-CVC.
In conclusion, the review and metaanalysis of Hockenhull et al does demonstrate the strong treatment effect that
favors AI-CVC in the prevention of
CRBSI, particularly in critically ill patients. Furthermore, the review demonstrates the cost effectiveness of such
intervention, showing that such antimicrobial technology represents a safety net
in the prevention of CRBSI. Given the
new directives by the Central Medical
Service System in the United States, including Medicare and Medicaid in not reimbursing hospitals for CRBSI, it is important to further press forward toward
the elimination of such infections. To
achieve such a zero end point and to
realistically eliminate CRBSI, the use of
AI-CVC in addition to effective infection
control bundle should become the standard of care. Slogans such as “zero tolerance” will not achieve a zero end point of
CRBSI and could not eliminate such preventable, serious infections unless they
are associated with the implementation
of a serious strategy that relies on evidence-based medicine. Using the combination of effective infection control
measures with highly efficacious techCrit Care Med 2009 Vol. 37, No. 2
nologies, such as AI-CVC, is a solid
foundation for this serious strategy.
Issam Raad, MD, FACP, FIDSA
Department of Infectious Diseases,
Infection Control and
Employee Health
The University of Texas M. D.
Anderson Cancer Center
Houston, TX
REFERENCES
1. Richards MJ, Edwards JR, Culver DH, et al:
Nosocomial infections in medical intensive
care units in the United States. National Nosocomial Infections Surveillance System. Crit
Care Med 1999; 27:887– 892
2. Maki DG, Cobb L, Garman JK, et al: An
attachable silver-impregnated cuff for prevention of infection with central venous
catheters: A prospective randomized multicenter trial. Am J Med 1988; 85:307–314
3. Crnich CJ, Maki DG: Are antimicrobialimpregnated catheters effective? Don’t throw
out the baby with the bathwater. Clin Infect
Dis 2004; 38:1287–1292
4. Hockenhull JC, Dwan KM, Smith GW, et al:
5.
6.
7.
8.
9.
The clinical effectiveness of central venous
catheters treated with anti-infective agents
in preventing catheter-related bloodstream
infections: A systematic review. Crit Care
Med 2009; 37:702–712
Marciante KD, Veenstra DL, Lipsky BA, et al:
Which antimicrobial impregnated central venous catheter should we use? Modeling the
costs and outcomes of antimicrobial catheter
use. Am J Infect Control 2003; 31:1– 8
Shorr AF, Humphreys CW, Helman DL: New
choices for central venous catheters: Potential financial implications Chest 2003;
4:275–284
Raad II, Hohn DC, Gilbreath BJ, et al: Prevention of central venous catheter-related infections by using maximal sterile barrier precautions during insertions. Infect Control
Hosp Epidemiol 1994; 15(4 Pt 1):231–238
Maki DG, Ringer M, Alvarado CJ: Prospective
randomized trial of povidone-iodine, alcohol,
and chlorhexidine for prevention of infection
associated with central venous and arterial
catheters. Lancet 1991; 338:339 –343
Pronovost P, Needham D, Berenholtz S, et al:
An intervention to decrease catheter-related
bloodstream infections in the ICU (published
10.
11.
12.
13.
corrections appears in N Engl J Med 2007;
356:2660). N Engl J Med 2006; 355:
2725–2732
Sherertz RJ, Ely EW, Westbrook DM, et al:
Education of physicians-in-training can decrease the risk for vascular catheter infection. Ann Intern Med 2000; 132:641– 648
Rupp ME, Lisco SJ, Lipsett PA, et al: Effect of
second generation venous catheter impregnated with chlorhexidine and silver sulfadiazine on central catheter-related infections: A
randomized controlled trial. Ann Intern Med
2005; 143:570 –580
Hanna HA, Benjamin R, Chatzinikolaou I, et
al: Long-term silicone central venous catheters impregnated with minocycline and rifampin decrease rates of catheter-related
bloodstream infection in cancer patients: A
prospective randomized clinical trial (published correction appears in J Clin Oncol
2005; 23:3652). J Clin Oncol 2004; 22:
3163–3171
Centers for Disease Control and Prevention
(CDC): Progress toward interruption of wild
poliovirus transmission—Worldwide, January 2007-April 2008. MMWR Morb Mortal
Wkly Rep 2008; 57:489 – 494
The ethics of quality improvement research*
I
n December 2006, an article in the
New England Journal of Medicine
showed how a simple intervention
virtually eliminated central venous
catheter-related blood stream infections at
hospitals participating in the Michigan
Keystone ICU Project (1). Lay and medical
authors hailed the findings that suggested
opportunities to save thousands of lives and
billions of dollars (1–3).
Soon afterward, the Office for Human
Research Protections (OHRP), responding
to an anonymous complaint, launched an
investigation culminating in Keystone’s
suspension, citing the failure to obtain institutional review board (IRB) approval at
participating hospitals and informed consent from patients (4). At first blush, the
action seems absurd. The intervention
*See also p. 725.
Key Words: quality improvement; human subjects
research; research ethics; institutional review board;
informed consent; expedited review
The authors have not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194c4d6
Crit Care Med 2009 Vol. 37, No. 2
studied was harmless—simply formal implementation of standard practice (1, 5).
Outrage followed, fueled by concern that
the OHRP’s action would discourage similar projects (2, 6 – 8). Leaders of professional organizations protested (6), whereas
authors in the lay press labeled the action
“bizarre and dangerous” (2). As one ethicist
suggested, the government office responsible for protecting human subjects seemed
to have taken action that would only increase harm (7).
In this issue of Critical Care Medicine,
Savel et al (9) illuminate the factors precipitating the OHRP’s actions, particularly the lack of consensus regarding the
relationship between quality improvement (QI) and human subjects research
(HuSR). In advance of the project, the
IRB at the lead investigator’s institution,
Johns Hopkins, had exempted Keystone
from review and waived the informed
consent requirement (1). The OHRP disagreed with this exemption, believing the
study constituted HuSR, thus mandating
oversight from each participating hospital and informed consent (4). Months
later, the OHRP seemed to modify its
findings, indicating that research like
Keystone that entailed negligible risk
would qualify for expedited review and
waiver of consent (10). Projects that were
QI only would not need IRB oversight.
Although the OHRP’s final opinion
may have closed this particular case,
Savel et al (9) argue persuasively for more
clarity regarding the oversight needed for
work combining QI and HuSR. To prevent future uncertainty, they offer three
recommendations. First, they suggest
streamlining approval for QI/HuSR and
clarifying the rules guiding the use of
central IRBs and waiver of informed consent. Second, they suggest developing
ways to make IRB approval less onerous.
Third, they suggest that hospitals too
small to have IRBs could use IRBs from
“nearby regional centers of excellence.”
Several others have contributed to
this debate (7, 8, 11, 12). Miller and
Emanuel (12) believe three questions are
key to evaluating projects like Keystone.
First, does the project involve HuSR?
Second, if yes, is expedited review appropriate? Third, is informed consent
needed? At present, there is little controversy regarding questions two and three:
expedited review and waived consent
791
would be appropriate if the intervention
entails minimal risk, obtaining formal
consent is unfeasible, and the waiver will
not adversely affect the rights and welfare
of subjects (8 –10, 12).
However, the first question remains unanswered. Was Keystone HuSR or not? The
question is important because HuSR, by
definition, requires IRB review, and institutions engaged in federally funded research must file a Federalwide Assurance
with the U.S. government (13). However,
requiring IRB oversight for studies not involving human subjects could impose unnecessary barriers (7). Many small hospitals
do not have IRBs (9). Central IRBs, which
could streamline approval (8, 9), are not
always available. Furthermore, institutions
committed to using a central IRB still have
to file a Federalwide Assurance, assuring
compliance with HuSR regulations (13).
Such documentation could overwhelm institutions lacking research infrastructures
and discourage worthy projects.
In contrast, if QI projects like Keystone
are not HuSR, then IRB oversight is unnecessary. The Code of Federal Regulations
Common Rule, which governs human research activity in the United States, defines
research as a “systematic investigation. . . designed to develop or contribute to
generalizable knowledge (13).” By this definition, Keystone, and work like it, seems to
constitute research. The Code of Federal
Regulations goes on to define a human
subject as “a living individual about whom
an investigator . . . conducting research
obtains 1) data through intervention or
interaction with the individual or 2) identifiable private information.”
So was Keystone HuSR or something
else? The technique used to reduce central venous catheter infections had already been proven effective; its value in
individual patients was not being questioned (5). Rather, Keystone asked if the
technique could reduce institutional infection rates (1). Thus, we agree with
Baily (7) that the research did not meet
the regulatory definition of HuSR. Instead, it seems to have been an example
of QI coupled with research on organizations and did not require IRB oversight.
In this view, the full implications of what
is often perceived as an inflexible and
burdensome system (IRB application, review, and approval; negotiation of a Federalwide Assurance for those without
792
one; and continuing review and review of
all changes before their implementation)
need not be invoked.
Another ethical issue surrounds the
notion of informed consent as it relates to
this project. Savel et al state that the
purpose of informed consent is to protect
the subjects from whatever risks may be
inherent in the research project. But, we
would argue that informed consent flows
more from the ethical principle of Respect for Persons, as described in the Belmont report (14). This principle relates to
a respect for autonomy, allowing individuals to make their own decisions and
accept whatever risks are inherent in the
project. It reflects the concept of “voluntariness” in deciding to participate in research. This is in contrast to QI implementation, which explicitly is not done
on a voluntary basis, but rather is an
operational implementation on the part
of healthcare organizations. Thus, it
could be argued that the Keystone project
did not invoke the need for informed consent for participation in research.
Research like the Keystone project offers useful guidance to healthcare institutions required to find ways to successfully implement recommended practices
(1). Although unfortunate in many ways,
the Keystone/OHRP saga has offered an
opportunity to consider the ethical framework on which the nascent field of QI research is being built. The resulting discussion, including the contribution by Savel et
al, should ensure that future work is performed both effectively and ethically.
Mark D. Siegel, MD, FCCP
Pulmonary and Critical Care
Section
Department of Internal Medicine
Yale University School of
Medicine
New Haven, CT
Sandra L. Alfano, PharmD, FASHP,
CIP
Chair, Human Investigation
Committees
Department of Internal Medicine
Yale University School of Medicine
New Haven, CT
REFERENCES
1. Pronovost P, Needham D, Berenholtz S, et al:
An intervention to decrease catheter-related
bloodstream infections in the ICU. N Engl
J Med 2006; 355:2725–2732
2. Gawande A: A lifesaving checklist. New York
Times. December 30, 2007. Available at:
http://www.nytimes.com/2007/12/30/opinion/
30gawande.html. Accessed October 22, 2008
3. Wenzel RP, Edmond MB: Team-based prevention of catheter-related infections. N Engl
J Med 2006; 355:2781–2783
4. Borror KC: Human research protections under
Federalwide Assurances FWA-5752, FWA-287,
and FWA-3834. July 19, 2007. Available at:
http://www.dhhs.gov/ohrp/detrm_letrs/YR07/
jul07d.pdf. Accessed October 5, 2008
5. Berenholtz SM, Pronovost PJ, Lipsett PA, et
al: Eliminating catheter-related bloodstream
infections in the intensive care unit. Crit
Care Med 2004; 32:2014 –2020
6. Hanson MD, Thomas A, Ingbar DH, et al:
Letter to the Honorable Michael Leavitt.
February 5, 2008. Available at: http://
www.hospitalmedicine.org/AM/Template.
cfm?Section⫽Advocacy_Policy&Template⫽/
CM/ContentDisplay.cfm&ContentID⫽16830.
Accessed September 28, 2008
7. Baily MA: Harming through protection?
N Engl J Med 2008; 358:768 –769
8. Kass N, Pronovost PJ, Sugarman J, et al:
Controversy and quality improvement: Lingering questions about ethics, oversight, and
patient safety research. Joint Comm J Qual
Patient Saf 2008; 34:349 –353
9. Savel RH, Goldstein EB, Gropper MA: Critical care checklists, the Keystone Project, and
the Office for Human Research Protections:
A case for streamlining the approval process
in quality-improvement research. Crit Care
Med 2009; 37:725–734
10. Borror KC: Human subject research protections under Federalwide Assurances
FWA5752, FWA-287, and FWA-3834. February
14, 2008. Available at: http://www.hhs.gov/
ohrp/detrm_letrs/YR08/feb08b.pdf. Accessed
October 22, 2008
11. Lynn J, Baily MA, Bottrell M, et al: The ethics of
using quality improvement methods in health
care. Ann Intern Med 2007; 146:666 – 673
12. Miller FG, Emanuel EJ: Quality-improvement research and informed consent. N Engl
J Med 2008; 358:765–767
13. Protection of human subjects: Basic HHS
policy for protection of human research subjects. June 23, 2005. Available at: http://
www.hhs.gov/ohrp/humansubjects/guidance/
45cfr46.htm#46.102. Accessed October 19,
2008
14. The National Commission for the Protection
of Human Subjects of Biomedical and Behavioral Research: The Belmont report, ethical
principles and guidelines for the protection
of human subjects of research. April 18,
1979. Available at: http://www.hhs.gov/ohrp/
humansubjects/guidance/belmont.htm. Accessed August 15, 2008
Crit Care Med 2009 Vol. 37, No. 2
Refractory shock in the intensive care unit—Don’t fail to spot
obstruction of the left ventricular outflow tract!*
L
eft ventricular outflow tract
obstruction (LVOTO) is a wellrecognized feature of hypertrophic cardiomyopathy (1). The
condition of LVOTO, in the absence of
hypertrophic cardiomyopathy, has also
been described in stress echocardiography with dobutamine (2– 4) or exercise
(5, 6) and after cardiac valve surgery (7–
10). The main mechanisms behind the
development of LVOTO, also referred to
as dynamic LVOTO, are inotropic stimulation of left ventricles (LV) with higher
than normal LV wall to lumen ratios such
as in hypertensive heart disease. Thus,
forceful contractions, particularly of the
basal part of an LV with small dimensions
and increased wall thickness in combination with peripheral vasodilation, may
precipitate subaortic flow obstruction and a
decline in cardiac output and hypotension.
If the LVOTO is severe enough, blood will
be ejected at a high velocity, which may
cause the anterior mitral valve leaflet to be
drawn to the septum by a Venturi effect, a
so-called systolic anterior motion (SAM).
Thus, parts of the anteriorly displaced mitral valve leaflet will extend beyond their
coaptation point and protrude into the
rapid flow velocity of the left ventricular
outflow tract. This may result in a further
LVOTO and a more or less pronounced
mitral valve regurgitation and eventually
cardiogenic shock.
In cardiac surgery, dynamic LVOTO is
a well-recognized phenomenon, which
may develop early after aortic valve replacement (7, 8) or mitral valve repair (9,
10) and may be totally reversible after
correction of the precipitating factors
such as high inotropic state, tachycardia,
hypovolemia, and systemic vasodilation.
*See also p. 729.
Key Words: left ventricle outflow tract obstruction;
cardiogenic shock; left ventricle hypertrophy; hypovolemia; cardiac inotropism
The author has not disclosed any potential conflicts of interest.
Copyright © 2009 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e318194fe38
Crit Care Med 2009 Vol. 37, No. 2
LVOTO with SAM may also occur in the
perioperative setting of noncardiac surgery. Luckner et al (11) described two
patients undergoing orthopedic surgery
who developed cardiovascular collapse
early after induction of anesthesia unresponsive to catecholamines and a third
patient with severe intraoperative bleeding. LVOTO and SAM were diagnosed by
transesophageal echocardiography, and
hypotension was resolved by colloid infusion, phenylephrine, and -blocker therapy. In these patients, LVOTO and SAM
were triggered by hypovolemia and anesthetic drug–mediated vaso- and venodilation in conjunction with increased catecholamine concentrations.
In this issue of Critical Care Medicine,
Chockalingam et al (12) describe the clinical recognition and management options
of five patients (all women) presenting
with unexplained hypotension and systolic
murmur in the emergency/critical care setting. The underlying cause of their critical
condition was dynamic LVOTO, with peak
left ventricular outflow tract pressure gradients ranging from 60 to 150 mm Hg,
diagnosed by echocardiography. The common clinical feature of all patients was hypertensive heart disease with more or less
pronounced LV hypertrophy. Three patients presented in the emergency room
with chest discomfort, anterior T-wave abnormalities, suggestive of myocardial ischemia, and mild to moderate troponin-I elevations. Two of these patients had a
normal coronary angiogram, and one had a
significant stenosis of the posterior (not
anterior) descending artery. Dobutamine
treatment was initiated in one patient because of shock. Severe LV apical dysfunction (apical ballooning syndrome) was confirmed in these three patients in
association with LVOTO and SAM of the
anterior mitral valve leaflet. Echocardiogram also revealed hypercontractility of the basal portions of the LV as a
compensatory mechanism for the apical
ballooning syndrome–related severe
apical dysfunction, which resulted in
LVOTO and SAM. The apical ballooning
syndrome, also known as Takatsubo
cardiomyopathy, is a reversible cardiomyopathy, which is triggered by severe
psychological stress occurring in older
women with normal coronary arteries,
and may mimic evolving acute myocardial infarction and coronary syndrome
(13). This condition is associated with a
severe elevation of plasma catecholamines (14), which further
strengthens the importance of cardiac
sympathetic stimulation for the development of LVOTO and SAM in patients
with aortic ballooning syndrome. Mild
to moderate troponin elevations were
observed in these patients with virtually
normal coronary arteries. According to
the authors, this may be explained by
high levels of peak systolic subendocardial wall stress, which may considerably
impair the oxygen demand–supply relationship of the subendocardial layers.
These patients were successfully treated
with -blockers, and there was no evidence of LVOTO or SAM on repeat echocardiograms.
The authors also review two mechanically ventilated patients in the intensive
care unit presenting with hypotension,
tachycardia, systolic murmur, and mild
to moderate troponin elevations. In one
of the patients, the clinical condition was
not improved by dopamine treatment.
Bedside echocardiograms revealed hyperdynamic LVs with hypertrophy and
LVOTO in both cases, with or without
SAM, which was fully resolved by vigorous fluid treatment in combination with
a -blocker. These two cases highlight
the clinical problem one may encounter
when inotropic treatment is instituted in
volume-depleted, iatrogenically or not,
patients in the intensive care unit with
hypertrophied LVs, particularly when
inotropic agents with vaso- and venodilating properties are used, e.g., dobutamine, low-dose dopamine, phosphodiesterase inhibitors, or levosimendan.
Furthermore, this report emphasizes
the importance of the use of echocardiography for early diagnosis of LVOTO in
793
patients in the intensive care unit with
refractory shock not responsive to
“fluid challenge” and inotropic therapy.
An echocardiogram in this situation
may reveal that the fluid challenge was
not vigorous enough and that inotropic
support of the hypertrophied, hypovolemic, and hyperdynamic LV generated
an LVOTO and caused a functional aortic stenosis in an LV not used to such
high levels of afterload. The echocardiogram will also serve as a guide in the
treatment of this critical condition consisting of blood volume expansion,
-blockers, and normalization of a low
systemic vascular resistance with a vasoconstrictor, if necessary. This treatment strategy may completely resolve
this clinical problem.
The authors of this interesting report
conclude that dynamic LVOTO may occur
in the emergency room and the critical
care units more often than is recognized.
To date, the true incidence of dynamic
LVOTO as a cause of hypotension in the
intensive care unit is not known, and
most likely, this condition is underdiagnosed. The good news is that early recognition and appropriate medical management may dramatically improve the
condition of these patients.
Sven-Erik Ricksten, MD, PhD
Department of Cardiothoracic
Anesthesia and Intensive
Care
794
Sahlgrens University
Hospital
Gothenburg, Sweden
8.
REFERENCES
1. Wigle ED, Rakowski H, Kimball BP, et al:
Hypertrophic cardiomyopathy: Clinical
spectrum and treatment. Circulation 1995;
92:1680 –1692
2. Pellika PA, Oh JK, Bailey KR, et al: Dynamic intraventricular obstruction during
dobutamine stress echocardiography. A
new observation. Circulation 1996; 86:
1429 –1432
3. Sorrentino MJ, Marcus RH, Lang RM, et al:
Left ventricular outflow tract obstruction as
a cause for hypotension and symptoms during dobutamine stress echocardiography.
Clin Cardiol 1996; 19:225–230
4. Scandura S, Arcidiacono S, Felis S, et al:
Dynamic obstruction to left ventricular
outflow during dobutamine stress echocardiography: The probable mechanisms and
clinical implications. Cardiologia 1998; 43:
1201–1208
5. Peteiro J, Montserrat L, Castro-Beiras A:
Labile subaortic obstruction during exercise stress echocardiography. Am J Cardiol
1999; 84:1119 –1123
6. Cabrera-Bueno F, Garcia-Pinilla JM, GomezDoblas JJ, et al: Beta-blocker therapy for dynamic left ventricular outflow induced by
exercise. Int J Cardiol 2007; 117:222–226
7. Aurigemma G, Battista S, Orsinelli, et al:
Abnormal left intracavitary flow acceleration
in patients undergoing aortic valve replacement for aortic stenosis. A marker for high
9.
10.
11.
12.
13.
14.
postoperative morbidity and mortality. Circulation 1992; 86:926 –936
Bartunek J, Sys SU, Rodrigues AC, et al:
Abnormal left ventricular intracavitary
flow acceleration after valve replacement
for aortic stenosis. Circulation 1996; 93:
712–719
Freeman WK, Schaff HV, Khanderia BK, et
al: Intraoperative evaluation of mitral valve
regurgitation and repair by transesophageal
echocardiography: Incidence and significance of systolic anterior motion. J Am Coll
Cardiol 1992; 20:599 – 609
Kreindel MS, Schiavone WA, Lever HM, et
al: Systolic anterior motion of the mitral
valve after Carpentier ring valvuloplasty for
mitral valve prolapse. Am J Cardiol 1986;
57:408 – 412
Luckner G, Margreiter J, Jochberger S, et al:
Systolic anterior motion of the mitral valve
with left ventricular outflow tract obstruction: Three cases of acute perioperative hypotension in noncardiac surgery. Anest
Analg 2005; 100:1594 –1598
Chockalingam A, Dorairajan S, Bhalla M, et al:
Unexplained hypotension: The spectrum of dynamic left ventricular outflow obstruction in
critical care settings. Crit Care Med 2009; 37:
729–734
Sharkey SW, Lesser JR, Zenovich AG,
et al: Acute and reversible cardiomyopathy
provoked by stress in women from the
United States. Circulation 2005; 111:
472– 479
Wittstein IS, Thiemann DR, Lima JA: Neurohormonal features of myocardial stunning
due to sudden emotional stress. N Engl
J Med 2005; 352:539 –548
Crit Care Med 2009 Vol. 37, No. 2