Sei sulla pagina 1di 15

The Journal of Emergency Medicine, Vol. 31, No. 2, pp.

185199, 2006
Copyright 2006 Elsevier Inc.
Printed in the USA. All rights reserved
0736-4679/06 $see front matter

doi:10.1016/j.jemermed.2006.05.008

Selected Topics:
Critical Care Medicine

MANAGEMENT OF SEPSIS DURING THE EARLY GOLDEN HOURS


Murugan Raghavan,

MD, MRCP (UK),*

and Paul E. Marik,

MD, FCCP, FCCM

*Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania and Division of Pulmonary
and Critical Care Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
Reprint Address: Paul Marik, md, fccp, Chief, Pulmonary and Critical Care Medicine, Thomas Jefferson University, 1015 Chestnut Street,
Suite M 100, Philadelphia, PA 19107

e AbstractSevere sepsis and septic shock are common


causes of morbidity and mortality. Interventions directed at
specific endpoints, when initiated early in the golden
hours of patient arrival at the hospital, seem to be promising. Early hemodynamic optimization, administration of
appropriate antimicrobial therapy, and effective source
control of infection are the cornerstones of successful management. In patients with vasopressor-dependent septic
shock, provision of physiologic doses of replacement steroids may result in improved survival. Administration of
drotrecogin alfa (activated), (activated protein C) has been
shown to improve survival in patients with severe sepsis
and septic shock who have a high risk of mortality. In this
article we review the multi-modality approach to early
diagnosis and intervention in the therapy of patients with
severe sepsis and septic shock. 2006 Elsevier Inc.

of sepsis occur each year, at least 225,000 of which are


fatal (2). Sepsis is now the 10th leading cause of death in
the United States (2,5). Despite the use of antimicrobial
agents and advanced life-support care, the case fatality
rate for patients with sepsis has remained between 30%
and 50% over the past three decades (3,6). Survivors of
sepsis and septic shock are observed to have a higher 6and 12-month mortality rate and a significantly lower
health-related quality of life (7,8). Recent advances in
understanding the pathophysiology of sepsis have led to
the evolution of newer therapies that have demonstrated
surprising efficacy (9 15). The current systematic review closely focuses on evidence-based recommendations, made by the Society of Critical Care Medicines
Surviving Sepsis Campaign 2004 guidelines (16). In
addition, we also performed literature search using terms
such as severe sepsis, septic shock, sepsis syndrome,
critical illness, and early resuscitation using the Medline database from 1960 to the present, and have attempted to include major clinical trials and other systematic reviews published in this field.

e Keywordssevere sepsis; septic shock; sepsis syndrome; critical illness; early resuscitation; golden hour

INTRODUCTION
In recent decades the reported incidence of sepsis has
increased dramatically, largely due to the advancing age
of the population, an increased number of invasive procedures performed, and immunosuppressive therapy (1
3). Sepsis is among the most common reasons for admission to intensive care units throughout the world (4).
In the United States alone, approximately 750,000 cases

SEPSIS PATHOPHYSIOLOGY
When the body is challenged by foreign microbial
agents, homeostatic mechanisms come into play that
attempt to rid the body of the foreign agent without

Selected Topics: Critical Care Medicine is coordinated by Joseph Varon, MD, of Baylor College of Medicine, Houston,
Texas

RECEIVED: 17 November 2004;


ACCEPTED: 30 March 2005
185

186

M. Raghavan and P. E. Marik

damaging the host. This involves the activation of proand anti-inflammatory pathways, which are tightly controlled and regulated (17). In most individuals, the body
is able to achieve a balance between pro-inflammatory
and anti-inflammatory mediators and homeostasis is restored. In some patients, however, this balance is upset
with an excessive pro-inflammatory response resulting in
systemic inflammatory response syndrome (SIRS),
multi-system organ dysfunction, septic shock and, ultimately, death (1720).
The constellation of abnormalities that characterize
severe sepsis and septic shock includes circulatory dysfunction with intravascular volume depletion, peripheral
vasodilatation, myocardial depression, and a diffuse microcapillary injury leading to hypoperfusion of vital organ vascular beds (2124). This occurs in the setting of
a hypermetabolic state resulting in an imbalance between
oxygen delivery and demand leading to tissue hypoxia.
This may be further complicated by cytopathic hypoxia
due to sepsis-induced mitochondrial dysfunction (25,26).
Tissue hypoxia is assumed to be the key factor leading to
multi-organ failure and death (17,27). Thus, early aggressive resuscitation of the septic patient may limit or
reverse tissue hypoxia and the progression to organ failure. This transition to organ failure occurs during the
critical golden hours, when time is survival, and the
definitive recognition and treatment of sepsis provides
maximal benefit in terms of outcome (9). These golden
hours may elapse in the Emergency Department (ED),
hospital ward, or the intensive care unit (ICU) (28 30).
Lundberg and coworkers demonstrated that for patients
with septic shock on the hospital wards, there were
clinically important delays in the transfer of patients to
the ICU, reception of intravenous fluid boluses, and
reception of inotropic agents, which had implications for
increased mortality (29).
Current management of sepsis includes early hemodynamic optimization with other physiologic organ supportive measures, timely and appropriate usage of antimicrobial agents, and treating or eliminating the source
of infection. Attempts at down-regulating the pro-inflammatory response with novel agents directed at specific
pro-inflammatory mediators has uniformly met with failure (17,3134). Recently several modalities including
activated protein C and stress doses of glucocorticoids
have been demonstrated to decrease mortality in critically ill septic patients (9 11,20,3537).

DIAGNOSIS OF SEVERE SEPSIS AND


SEPTIC SHOCK
Sepsis is a systemic process with a myriad of clinical
manifestations. The initial symptoms of sepsis are non-

specific and include malaise, tachycardia, tachypnea, fever, and sometimes hypothermia. Although most patients
with sepsis have an elevated white cell count, some
patients present with a low white cell count, which in
general is a poor prognostic sign. Other clinical manifestations include altered mental status, hypotension, respiratory alkalosis, metabolic acidosis, hypoxemia with
acute lung injury, thrombocytopenia, consumptive coagulopathy, proteinuria, acute tubular necrosis, intrahepatic cholestasis, elevated transaminases, hyperglycemia,
and hypoglycemia (see Table 1).
Patients may present with clinical features of a localized site of infection, such as cough, tachypnea, and
sputum production due to pneumonia; flank pain and
dysuria with complicated urinary tract infection; and
abdominal pain with intra-abdominal infection. The
manifestations of sepsis sometimes can be quite subtle,
particularly in the very young, the elderly, and those with
chronic debilitating or immunosuppressing conditions.
These patients may present with normothermia or hypothermia. The failure to generate a temperature greater
than 37.5C (99.6F) in the first 24 h of clinical illness
has been associated with an increased mortality rate
(38,39). An altered mental state or an otherwise unexplained respiratory alkalosis may be the presenting feature of sepsis.
The signs and symptoms of systemic inflammation are
not useful in distinguishing infectious from noninfectious causes of SIRS. Furthermore, a bacterial pathogen
is not isolated in all patients with sepsis. Consequently, a
number of markers have been evaluated as more specific
indicators of infection (40,41). Tools such as procalcitonin (PCT) may be useful and have been shown to be
superior to other inflammatory markers such as TNF-,
IL-6, IL-1, and C-reactive protein (CRP) in outcome

Table 1. Criteria for Organ Systems Dysfunction during


Severe Sepsis and Septic Shock
Cardiovascular
SBP 90 mm Hg or MAP 70 mm Hg for at
least 1 h despite adequate fluid resuscitation
Vasopressor use
Renal
Urine output 0.5 ml/kg /h for 1 h, despite
adequate fluid resuscitation
Respiratory O2
PaO2/FiO2 250 in the presence of other organs
or systems failure or
200 if the lung is the only dysfunctional organ
Hematologic
Platelet count 80,000/mm3 or 2 by 50% in the
preceding 3 days
Unexplained metabolic acidosis
pH 7.30 or base deficit 5.0 mmol/L
Lactate level 1.5 times normal
SBP systolic blood pressure; MAP mean arterial pressure.

Management of Sepsis

prediction among critically ill patients (42,43). Nevertheless, various noninfectious insults such as major surgery, tissue trauma, and prolonged circulatory failure
may induce these markers, thereby decreasing the specificity in differentiating sepsis from other inflammatory
states (17). Consequently, an aggressive search and early
consideration of sepsis is required in distinguishing severe sepsis from other causes of hypotension and organ
dysfunction on the patients arrival at the hospital.
Numerous studies have evaluated the use of lactate as
a diagnostic, therapeutic, and prognostic marker of
global tissue hypoxia in patients with circulatory shock
(44 46). An elevated serum lactate concentration, although lacking in precision, may identify tissue hypoperfusion in non-hypotensive patients. Lactate concentrations 4 mmol/L in the presence of the systemic
inflammatory response syndrome (SIRS) have been
shown to significantly increase intensive care unit (ICU)
admission rates and mortality rate in normotensive patients (47). Although the sensitivity and specificity of
single lactate concentrations as markers of tissue hypoperfusion have been debated, serial measurements or
lactate clearance over time have been shown to be better
prognosticators of organ failure and mortality (48,49).
Persistent elevations in lactate 24 h are associated with
a mortality rate as high as 89% (50).
Nguyen et al. evaluated the clinical utility of the
lactate clearance as an indicator of outcome in severe
sepsis and septic shock in a prospective observational
study (44). Septic shock therapy was initiated in the ED
and continued in the ICU. Lactate clearance was defined
as the percent decrease in lactate from ED presentation to
hour 6. A total of 111 patients were enrolled, with an ED
length of stay 6.3 3.2 h, and an overall in-hospital
mortality rate of 42.3%. Survivors compared with nonsurvivors had a lactate clearance of 38.1 34.6 vs. 12.0
51.6%, respectively (p 0.005). Multivariate logistic
regression analysis showed that lactate clearance had a
significant inverse relationship with mortality (p 0.04).
There was an approximately 11% decrease in likelihood
of mortality for each 10% increase in lactate clearance.
Therefore, patients with higher lactate clearance after 6 h
of ED intervention have improved outcomes compared
with those with lower lactate clearance.

EARLY GOAL-DIRECTED THERAPY OF


SEPTIC SHOCK
Rivers et al. showed that early goal-directed hemodynamic optimization starting in the ED for severe sepsis
and septic shock patients significantly decreases morbidity and mortality (9). A key to this success is the early
recognition of sepsis and the mobilization of frontline

187

health care providers analogous to patients with acute


myocardial infarction, stroke, and trauma (51). The study
showed that early aggressive therapy that optimized cardiac preload, afterload, and contractility in patients with
severe sepsis and septic shock improved survival. The
authors employed infusions of colloid or crystalloid,
vasoactive agents, and transfusions of red cells to increase oxygen delivery. Resuscitation endpoints chosen
for assessment of the adequacy of oxygen delivery were
the normalization of values for mixed venous oxygen
saturation (ScvO2), lactate concentration, base deficit,
and pH. Patients in the group that received early goaldirected therapy received more fluid, inotropic support,
and blood transfusions during the first 6 h than did
control patients, who received standard resuscitation
therapy. During the interval from 7 to 72 h, patients in
the group receiving early goal-directed treatment had a
higher mean central venous oxygen concentration, a
lower mean lactate concentration, a lower mean base
deficit, and a higher mean pH than the control group.
Mortality was 30.5% in the group receiving early goaldirected treatment, as compared with 46.5% in the control group (p 0.009). The patients in the early-therapy
group received, on average, approximately 1500 mL
more in total fluids in the first 6 h of treatment than did
the standard-therapy group and had a significantly higher
mean arterial pressure (mean [ SD], 95 19 vs. 81
18 mm Hg, respectively; p 0.001). Although the
benefit of red cell transfusions and optimization of mixed
venous saturation are of questionable benefit in patients
with severe sepsis, early aggressive resuscitation with
fluids and vasopressor agents to restore the balance between oxygen delivery and oxygen demand may largely
explain the improved survival among those patients who
received early goal-directed therapy (52).
Similarly, Levy et al. analyzed the Sequential Organ
Failure Assessment (SOFA) scores during the first 48 h
in 1036 severely septic patients (53). They hypothesized
that if early resuscitation is associated with improved
outcome in severe sepsis, then a dynamic risk prediction
model based on changes in clinical parameters from
baseline to Day 1 should be predictive of outcome. In
their study, mortality rates correlated with baseline cardiovascular (p 0.0022), renal (p 0.0001), hematologic (p 0.0044), and respiratory (p 0.0013) SOFA
scores. From baseline to day 1, the direction of change in
cardiovascular, renal, respiratory, hematologic, and hepatic functions independently predicted 28-day mortality. The implications of this study are that if organ
dysfunction is not improving during the first day of
severe sepsis, the mortality risk is significantly increased,
underscoring the importance of early recognition and
therapeutic intervention to prevent sequential organ dysfunction.

188

M. Raghavan and P. E. Marik

Recognize severe sepsis and septic shock on arrival to ED


Maintain airway and establish i.v. access

Pre-ICU Management

Aggressive volume resuscitation with


crystalloids and colloids (see Table 2)

If MAP < 70 mm Hg, after at least 2000


mL of fluids

Start Norepinephrine (0.01 g/kg/min)


Continue i..v. fluids
Monitor MAP
Monitor urine output

If MAP < 70 mm Hg despite


norepinephrine of 0.5 g/kg/min with poor
tissue perfusion
(Catecholamine- resistant shock)

Early empiric broad spectrum


antimicrobial therapy and source control

Fluid responsive
Draw random Cortisol
and administer Hydrocortisone
100 mg i.v. q 8 h*

Consider Recombinant Activated


Protein C (24 g/kg/h) i.v. for 96
h if new 2 or organ systems
failure @ (see Table 1)

Echocardiogram or PAC assessment of


LV function

ICU Management

Low CI

Start Dobutamine

High CI with low SVR

Start Vasopressin 0.01U/min

Persistent shock

Shock resolution

Observe

Monitor
MAP
pH
Lactic acid
Oxygenation
Urine output
Sublingual Co2

Intensive Glycemic control with i.v. Insulin (Target glucose 80-110 mg/dL)
Low Tidal volume ventilation for lung injury (VT 6 mL/kg; Ppl < 30 cms H20)
Narrow antimicrobials based on culture results
Early CVVH for sepsis-induced acute renal failure
Semi-recumbent head positioning to 45
Early enteral nutrition
Sedation goal with intermittent awakening
Deep vein thrombosis prophylaxis
Stress ulcer prophylaxis

Figure 1. Suggested algorithm for management of patients


presenting with severe sepsis and septic shock during the
initial golden hours. MAP mean arterial pressure; CVVH
continuous veno-venous hemofiltration; VT tidal volume;
CI cardiac index; PAC pulmonary artery catheter. *See
Figure 2 for further evaluation of adrenocortical function and
treatment. @If no contraindications present such as active
internal bleeding, recent (< 3 months) hemorrhagic stroke,
recent (< 2 months) intracranial or intraspinal surgery, or
severe head trauma, trauma with an increased risk of lifethreatening bleeding, presence of an epidural catheter, intracranial neoplasm or mass lesion or evidence of cerebral
herniation.

Therefore, it is imperative that a clinical decision to


stratify at-risk patients with sepsis and to identify those
with an increased risk of dying begins as soon as the
patient arrives in the hospital. Use of scoring systems
such as the Mortality in Emergency Department Sepsis
Score (MEDS) may prove helpful with triage decisions,
ICU placement, selection and activation of various therapeutic interventions (54).

VOLUME RESUSCITATION
The resuscitation of a patient with severe sepsis or sepsis-induced tissue hypoperfusion (hypotension or lactic

acidosis) should begin as soon as the syndrome is recognized, and should not be delayed pending hospital or
ICU admission (Figure 1). The hallmarks of severe sepsis and septic shock include intravascular volume depletion, peripheral vasodilatation with hypotension, myocardial depression, and abnormal blood flow distribution
(17,55). Several studies have demonstrated that indices
of inadequate tissue perfusion and oxygenation are
strong predictors of multi-organ dysfunction and death in
patients with sepsis (56 59).
Aggressive volume resuscitation is considered the
best initial approach to therapy for the cardiovascular
instability of sepsis (16,60,61). Hypotension often can be
reversed with fluid administration alone. Volume repletion alone in patients with septic shock has been shown
to produce significant improvement in cardiac function
and systemic oxygen delivery, thereby enhancing tissue
perfusion (60,62 64). Fluid requirements for the initial
resuscitation of patients with septic shock are commonly
large, with up to 10 L of crystalloids or 4 L of colloids
required in the first 24 h (9,60,62,63,65). Rapid infusion
of fluids is best initiated with boluses of 500 to 1000 mL
titrated to endpoints, such as a mean arterial pressure
70 mm Hg, heart rate 100 beats/min, and urine output
0.5 mL/kg/h during the initial 6 h (Figure 1, Table 1)
(9,16,66). After the initial volume resuscitation with
approximately 5 L of fluids in the first 6 h, maintenance
fluids should be administered at a rate of 150 200 mL/h,
with 500-cc boluses as required (17,66). In approximately 50% of patients with sepsis who initially present
with hypotension, fluids alone will reverse hypotension
and restore hemodynamic stability.
The choice of crystalloid or colloid (or both) for
resuscitation of patients with sepsis remains a controversial issue (6770). Crystalloids have generally been recommended as the volume expander of first choice (60).
However, in sepsis, the interstitial volume is increased
due to the marked increase in capillary permeability.

Table 2. Suggested Fluid Resuscitation Algorithm for


Hemodynamic Instability of Severe Sepsis and
Septic Shock
1 L N/S 1520 min
1 L Ringers 30 min
Start Norepinephrine if MAP 70 mm Hg
1 L Hextend 3040 min
1 L Ringers 3040 min
1 L Hextend 3040 min
Ringers lactate 200 cc/h
Bolus 500 cc Hextend as required
Monitor
MAP, Urine Output
Oxygenation
LV functionbedside ECHO
Sublingual CO2

Management of Sepsis

Therefore, crystalloids will further increase the interstitial fluid volume with less than a quarter of the volume
remaining intravascularly (71). It has been suggested,
therefore, that in patients with sepsis, colloids such as
albumin may limit the degree of third space loss and
prevent tissue and pulmonary edema by enhancing
plasma oncotic pressure (56,62,70,72). However, in the
critically ill patient, there is a very poor relationship
between the serum albumin and colloid osmotic pressure
(73,74). Furthermore, sepsis is characterized by a generalized capillary leak, reducing the importance of the
colloid osmotic pressure in influencing fluid flux.
Currently, 0.9% NaCl (normal saline) is the most
commonly used resuscitation fluid, in patients with sepsis (75). However, large volumes of saline infusion have
been shown to cause hyperchloremic metabolic acidosis
(76 78). Although the clinical consequences of this metabolic acidosis are not well understood, it has been
shown to have lower mortality when compared to metabolic acidosis of other etiologies in humans (79). The
hyperchloremia associated with saline-based solutions
has been shown to decrease splanchnic mucosal perfusion, glomerular filtration, and cause coagulopathy (80
83). Balanced salt solutions such as Lactated Ringers and
colloids have therefore been proposed as alternatives.
Although resuscitation with Lactated Ringers solution
results in improved arterial pH, it fails to improve survival time in animal models of septic shock (75). As the
volume of distribution is much larger for crystalloids
than for colloids, resuscitation with crystalloids requires
more fluid to achieve the same endpoints and results in
more edema (16).
Finfer et al., in a recent multicenter, randomized,
double-blind trial, compared the effect of fluid resuscitation with 4% albumin or saline on mortality in
6997 heterogeneous ICU patients (65). There were no
significant differences between the groups in the mean
number of days spent in the ICU, days spent in the
hospital, days of mechanical ventilation, days of renalreplacement therapy, or mortality between the two
groups. However, in the subgroup analysis of patients
with severe sepsis, the relative risk of death during the
28-day study period among those randomly assigned
to receive albumin as opposed to saline was 0.87, as
compared with a corresponding relative risk of 1.05
among patients without severe sepsis (p 0.06 by the
test for a common relative risk). In contradistinction to
an earlier meta-analysis that suggested that albumin
administration increases mortality in critically ill patients, this study provides evidence that volume resuscitation that includes a colloid (in this case albumin)
may be preferable to saline alone for intravascular
volume resuscitation of patients with severe sepsis
(69). However, given the concerns regarding the cost

189

and risks associated with human blood products, routine administration of human albumin to septic patients cannot be recommended (65).
Similarly, the use of packed cells as first-line therapy to increase the ScvO2 and thereby oxygen delivery
cannot be recommended. Currently, there is good evidence that packed red cells do not increase oxygen
consumption (at least in the first 24 h) in patients with
sepsis (84,85). Paradoxically, old units of packed
red cells may cause tissue dysoxia in patients with
sepsis (84,85). Furthermore, transfusions of packed
red cells have been shown to increase mortality in
critically ill patients with sepsis (84,86,87).

VASOPRESSOR THERAPY
When an appropriate fluid challenge (at least 2 L) fails to
restore adequate blood pressure (mean arterial pressure
[MAP] 70 mm Hg) and organ perfusion, therapy with
a vasopressor agent should be started (Figure 1). Vasopressor therapy also may be required to maintain perfusion in the face of life-threatening hypotension, even
when fluid resuscitation is in progress and hypovolemia
has not yet been corrected (16). Patients with sepsis have
a markedly abnormal ventricular response to volume
infusion, with a significantly smaller increase in left
ventricular stroke work index than controls in response
to fluid challenges (61). Moreover, due to the massively
reduced systemic vascular resistance, patients may remain hypotensive despite adequate fluid resuscitation
(38).
Currently, norepinephrine remains the first-choice vasopressor agent in patients with septic shock (16). Although there is no high quality evidence, human and
animal studies suggest many advantages of norepinephrine over other vasopressor agents (88 92). Norepinephrine increases mean arterial pressure due to its vasoconstrictive effects, with little change in heart rate and less
increase in myocardial oxygen consumption compared
with dopamine (16,93). Norepinephrine has been shown
to improve hemodynamic parameters including splanchnic perfusion and tissue oxygen utilization in most patients with sepsis (9395). Despite many practitioners
concerns, there are no data to indicate that this agent has
a deleterious renal effect (93,96,97).
Dopamine has traditionally been the vasoactive drug
of choice in patients with sepsis (16,60,98,99). However,
dopamine may not be the ideal vasopressor for a number
of reasons (9395,99). During sepsis, dopamine has been
shown to cause more tachycardia and potentiate dysrhythmias as chronotropic sensitivity to -adrenergic
stimulation by dopamine is increased (95,100 102). In
addition, the positive chronotropic and inotropic effects

190

of dopamine elevate myocardial oxygen requirements,


which may not be adequately met by increased coronary
flow (102). In critically ill septic patients, low-dose dopamine has been shown to cause precapillary vasoconstriction with diversion of blood flow away from the gut,
hastening gut ischemia (103105). Furthermore, dopamine potentiates immune suppression during sepsis by
inhibiting stimulated lymphocyte proliferation, immunoglobulin synthesis, cytokine production; and promotes
lymphocyte apoptosis (106 110). Dopamine also decreases growth hormone secretion and thyrotropin release (111). Growth hormone deficiency may also contribute to a negative nitrogen balance during critical
illness (17).
The widespread use of low-dose dopamine to improve
renal function in critically ill patients has been refuted by
a large randomized trial and two meta-analyses comparing low-dose dopamine to placebo (112114). In the
Australian and New Zealand Trials Group randomized
controlled study, there was no difference in either primary outcomes (peak serum creatinine, need for renal
replacement therapy, urine output, time to recovery of
normal renal function) or secondary outcomes (survival
to either ICU or hospital discharge, ICU stay, hospital
stay, dysrhythmias) between low-dose dopamine and
placebo (112). Thus, the available data do not support
administration of low doses of dopamine to maintain or
improve renal function (112,113).
Hypotensive patients with severe sepsis may have a
low, normal, or increased cardiac output. In hemodynamically unstable patients who have signs of inadequate
tissue perfusion after adequate volume resuscitation, require assessment of left ventricular (LV) function
(115,116). This may be best achieved by bedside transthoracic echocardiography. Portable hand-carried ultrasound machines are now available that allow for repeated
assessment of global left ventricular function by the
emergency physician or intensivist (116 121). In addition to an accurate assessment of left and right ventricular function during septic shock, either trans-thoracic or
trans-esophageal echocardiography may be used to assess vena cava collapsibility as a gauge of preload responsiveness in mechanically ventilated patients
(119,121123). Furthermore, various other methods of
measuring cardiac output such as esophageal Doppler
probes and electrical bio-impedance techniques have
been successfully used by emergency physicians
(124,125).
Patients with poor left ventricular contractility (or low
to normal cardiac output) despite adequate fluid resuscitation should be started on dobutamine, in conjunction
with vasopressor therapy, to augment cardiac output.
Dobutamine has been demonstrated to increase oxygen
delivery and reverse both systemic and local indices of

M. Raghavan and P. E. Marik

tissue hypoxia in patients with sepsis (104,126 128).


Nevertheless, the use of dobutamine in isolation may
cause or potentiate hypotension due to 2-mediated vasodilatation. The coexistence of a decreased ejection
fraction and widespread vasodilatation implies that it
may be advantageous to use two drugs with different
receptor profiles to attain the desired effect (55,126). The
combination of dobutamine and norepinephrine has been
demonstrated to increase both the cardiac output and
peripheral vascular resistance and to improve indices of
tissue oxygenation in patients with severe sepsis (126).
There are concerns about the use of epinephrine in
patients with sepsis because this agent has been shown to
cause a significant increase in the serum lactate concentration with an associated decrease in the serum pH
(129,130). This rise in lactate has not been demonstrated
with the use of the other commonly used inotropic
agents. The increased lactate production may be due to
increased glycogenolysis or a maldistribution of blood
flow (131). There are limited data on the hemodynamic
effect of phenylephrine in patients with sepsis (63,132).
Although this agent increases blood pressure and systemic vascular resistance, it usually lowers cardiac output.
Low-dose vasopressin (0.01 0.04 units/min) may be
considered in patients with refractory shock despite adequate fluid resuscitation and high-dose conventional
vasopressors (16,133,134). During early septic shock,
vasopressin concentrations are elevated; however, with
continued shock, concentrations decrease to normal
range in the majority of patients between 24 and 48 h
resulting in relative vasopressin deficiency (135). Because vasopressin also has been shown to increase serum
cortisol levels via stimulation of ACTH release, a relative vasopressin deficiency state during septic shock
may predispose to a relative adrenal insufficiency and
increased mortality (136 138). Therefore, it is conceivable that supplementation of a physiologic dose of steroids and vasopressin improves survival. Pending the
outcome of ongoing trials, vasopressin is not recommended as a replacement for norepinephrine as a firstline agent. Vasopressin is a direct vasoconstrictor without inotropic or chronotropic effects and may result in
decreased cardiac output and hepatosplanchnic flow
(139). Doses of vasopressin 0.04 units/min have been
associated with myocardial ischemia, significant decreases in cardiac output, and cardiac arrest (137).

GOALS OF HEMODYNAMIC SUPPORT


The primary goal of sepsis resuscitation is to expedite
the correction of tissue hypoxia and oxygen debt to
prevent sequential organ dysfunction (9). Various end-

Management of Sepsis

points have been proposed to confirm the adequacy of


resuscitation, including normalization of values for
mixed venous oxygen saturation ( 70%), central
venous pressure ( 8 12 mm Hg), arterial lactate
concentration, base deficit, urine output and pH (9,16).
However, these global markers may fail to detect
tissue hypoxia in vital vascular beds. For instance,
central venous pressure and pulmonary artery occlusion pressure (PAOP) have been shown to be poor
indicators of intravascular volume status and preload
responsiveness (52). In addition, alterations in central
venous pressure and PAOP associated with changes in
circulating volumes do not correlate significantly with
changes in end diastolic volume and stroke volume
(140 142). This lack of correlation between pressure
and volume indexes of preload has been ascribed to
several causes, including, most prominently, variations in ventricular compliance specific to the patient
groups studied (143). During sepsis, mixed venous
oxygen saturation (SmvO2) and, by inference, central
venous oxygen saturation (ScvO2), have been postulated to be a surrogate marker for the adequacy of
oxygen delivery and an endpoint of resuscitation
(9,16). However, during sepsis, mixed venous oxygen
saturation has been shown to poorly correlate with
cardiac output and indices of tissue oxygenation due to
microcirculatory flow maldistribution and shunting
(22,56,144,145). When patients present with hypovolemia and normal or low cardiac output, mixed venous
oxygen saturation is frequently low and is likely to
increase with resuscitation. However, although a low
SmvO2 indicates inadequate oxygen delivery, a normal SmvO2 does not exclude a tissue oxygen debt,
especially in sepsis, possibly due to shunting (56,144).
The endpoint of the initial phase of resuscitation is
to achieve a mean arterial pressure of 70 to 80 mm Hg
as rapidly as possible. A decrease in the mean arterial
pressure to less than the renal autoregulatory range
(mean arterial pressure, 70 mm Hg) leads to an
abrupt decrease in glomerular filtration rate (GFR)
(146). Serial measurements of other indices of perfusion such as urine output and lactate levels may help
to identify patients with occult persistent shock. In
patients receiving vasopressor agents, sublingual capnography provides useful information as to the adequacy of splanchnic oxygen utilization and to assess
the adequacy of volume and pressor resuscitation
(21,144,145). Currently, there is insufficient evidence
for routine consideration of invasive hemodynamic
monitoring (pulmonary artery catheters and oximetric
central venous catheters) in patients with septic shock
(142,147150).

191

ANTI-INFECTIVE THERAPY
The importance of the early and accurate diagnosis of
suspected infection, to select initial empirical anti-microbial therapy and to achieve source control, cannot be
overemphasized (Figure 1). Sepsis remains the primary
diagnosis in over 50% of patients who die in the ICU
(4,151). Recent studies have clearly demonstrated that
delays in initiating appropriate anti-infective therapy for
bactremia and other serious infections is associated with
increased mortality rates (15,37,152156).
Most patients presenting with severe sepsis have a
pulmonary, genitourinary, primary blood stream, intraabdominal, or intravenous catheter as a source of infection, and two-thirds of patients with severe bacterial
sepsis have negative blood cultures (157159). To optimize identification of causative organisms, at least two
blood cultures (at least 20 mL) should be obtained with
at least one drawn percutaneously and one drawn
through each vascular access device, unless the device
was recently ( 48 h) inserted (152,160). Cultures of
other sites such as urine, cerebrospinal fluid, wounds,
respiratory secretions, or other body fluids should be
obtained before antibiotic therapy is initiated, as the
clinical situation dictates. For patients presenting with
severe sepsis and septic shock secondary to communityacquired pneumonia, who are mechanically ventilated,
ED-based bronchoalveolar lavage technique allows for
early identification of pathogens, which leads to changes
in antibiotic therapy (161). Non-bronchoscopic protected
bronchoalveolar lavage, when successfully done by the
emergency physician, offers the potential advantage of
evaluating patients who are antibiotic nave without negatively affecting their care.

ANTIMICROBIAL THERAPY
Antimicrobial therapy remains the cornerstone of treatment in patients with sepsis. Empiric intravenous antibiotic therapy should be started within the first hour of
recognition of severe sepsis, after appropriate cultures
have been obtained. Selection of antimicrobial therapy
for patients presenting with severe sepsis and septic
shock depends on a number of factors including the
source or focus of infection, whether the infection is
community- or hospital-acquired, knowledge of local
antimicrobial resistance patterns, presence of indwelling
catheters, and immunologic status of the patient. Initial
empirical anti-infective therapy should include one or
more drugs that have activity against the likely pathogens (bacterial or fungal) and that penetrate into the
presumed source of sepsis.

192

In recent years there has been a tremendous increase


in infections caused by Gram-positive organisms such as
methicillin-resistant Staphylococcus aureus (MRSA)
(162166). Methicillin resistance has an important impact on antimicrobial options due to the lost efficacy of
anti-staphylococcal penicillin and early generation cephalosporins (167). Increasingly, MRSA infections have
their origin in the general outpatient community, a trend
that is, in part, the result of prior antimicrobial exposure
(168 170). Vancomycin is generally the first-line antimicrobial therapy in these patients.
The initial selection of an empirical antimicrobial
regimen should be broad enough, covering all likely
pathogens because there is little margin for error in
critically ill patients. There is currently ample evidence
that failure to initiate appropriate therapy promptly (i.e.,
therapy that is active against the causative pathogen) has
adverse consequences on outcome (15,37). Patients with
severe sepsis or septic shock warrant broad-spectrum
therapy until the causative organism and its antibiotic
susceptibilities are defined.
Once a pathogen is isolated, monotherapy is adequate for most infections (171). Indications for double-antimicrobial therapy include suspected or proven
Pseudomonas aeruginosa infections, enterococcal infections, the treatment of febrile-neutropenic patients,
and severe intra-abdominal infections (172,173). In
patients with culture-negative sepsis, continuation of
the initial empiric combination is warranted. Additional antibiotics or a change in antibiotics may be
required in patients with culture-negative sepsis who
do not seem to be responding to the initial empiric
regimen. Dosing regimens should take into account
whether the antibiotic kills by time-dependent kinetics (e.g., -lactam antibiotics, vancomycin) or concentration-dependent kinetics (e.g., aminoglycoside)
(174,175). Recent data suggest that the clinical effectiveness of -lactam antibiotics and vancomycin is
optimal when the concentration of the antimicrobial
agent at the site of infection exceeds the minimum
inhibitory concentration of the infecting organism for
a prolonged period (176,177). In addition, antibiotic
dosing should also take into account the patients
hepatic and renal function. The antimicrobial regimen
should be reassessed after 48 72 h on the basis of
microbiological and clinical data with the aim of using
a narrow spectrum antibiotic to prevent the development of resistance, to reduce toxicity, and to reduce
costs. Once a causative pathogen is identified, there is
no evidence that combination therapy is more effective than monotherapy. The duration of therapy should
typically be 710 days and guided by clinical response
(16).

M. Raghavan and P. E. Marik

SOURCE CONTROL OF SEPSIS


Every patient presenting with severe sepsis should be
evaluated for the presence of a focus on infection amenable to source control measures. Specifically, this includes the drainage of an abscess or local focus on
infection, debridement of infected necrotic tissue, removal of a potentially infected device, or the definitive
control of a source of ongoing microbial contamination.
When a focus of infection amenable to source control
measures such as an intra-abdominal abscess, a gastrointestinal perforation, cholangitis, or intestinal ischemia
has been identified as the cause of severe sepsis or septic
shock, source control measures should be instituted as
soon as possible after initial resuscitation (16). If intravascular access devices are potentially the source of
severe sepsis or septic shock, they should be promptly
removed after establishing other vascular access. When
patients develop sepsis of unknown source, it may be
reasonable to leave vascular access devices in place until
the source of infection can be determined. However,
when patients have severe sepsis or septic shock of
unknown source, clinicians should consider removal and
replacement of vascular access devices to be a priority,
even if the device is tunneled or surgically implanted
(178).

CORTICOSTEROIDS
In patients with vasopressor-dependent septic shock,
there has been a great deal of interest regarding the
assessment of adrenal function and the indications for
adrenal replacement therapy (12,13,138,179,180). Although the exact incidence of adrenal insufficiency varies with the diagnostic test and cortisol level used to
diagnose the disorder, in one series approximately 61%
of critically ill septic shock patients had adrenal insufficiency when a baseline cortisol concentration of 25
g/dL was used as the diagnostic threshold (13). Therefore, any patient who presents with septic shock should
be considered for treatment with stress doses of corticosteroids (hydrocortisone, 100 mg i.v. q. 8 hourly) pending the results of diagnostic testing (random cortisol
level) (Figure 2). During septic shock, treatment with
stress-level doses of hydrocortisone has been demonstrated to improve hemodynamic status, down-regulate
the proinflammatory response, and improve mortality;
however, what currently remains controversial is the
diagnosis of this disorder (12,181186). In a highly
stressed patient with severe sepsis and septic shock, a
random cortisol level assesses the integrity of the entire
HPA axis (187). Dysfunction at the hypothalamic, pituitary, or adrenal level will result in low circulating cor-

Management of Sepsis

Figure 2. Strategy for evaluation of adrenocortical function


and treatment of patients with vasopressor-dependent severe sepsis and septic shock.

tisol levels ( 25 g/dL), and therefore a stress random


cortisol level of 25 g/dL in a patient with refractory
hypotension warrants a trial of low-dose glucocorticoids
(179).
Marik and Zaloga studied whether a baseline (random) cortisol concentration 25 g/dL was a better
discriminator of adrenal insufficiency than the standard
(250 g) and the low-dose (1 g) corticotropin stimulation tests, in 59 patients with septic shock (13). Following baseline cortisol level, patients were given 1 g of
corticotrophin (low dose), followed 60 min later by an
injection of 249 g of corticotropin (high-dose test).
Cortisol concentrations were obtained 30 and 60 min
after administration of low- and high-dose corticotropin.
All patients were administered hydrocortisone (100 mg
every 8 h) for the first 24 h while awaiting results of
cortisol assessment. Patients were considered steroid responsive if the pressor agent could be discontinued
within 24 h of the first dose of hydrocortisone. Sixty-one
percent of patients met the criteria of adrenal insufficiency when a baseline cortisol concentration of 25
g/dL was used. Ninety-five steroid-responsive patients
had a baseline cortisol concentration 25 g/dL. Receiver operating characteristic curve analysis revealed
that a stress cortisol concentration of 23.7 g/dL was the
most accurate diagnostic threshold for determination of
the hemodynamic response to glucocorticoid therapy.
The sensitivity of a baseline cortisol 25 g/dL in
predicting steroid responsiveness was 96%, compared
with 54% for the low-dose test and 22% for the high-

193

dose test. Therefore, a random cortisol concentration of


25 g/dL in a highly stressed patient is a very useful
diagnostic threshold for the diagnosis of adrenal insufficiency (13).
Annane and colleagues, in a placebo-controlled, randomized, double-blind, multicenter study, enrolled 300
adult patients with septic shock after undergoing a short
high-dose (250g) corticotropin test (138). Patients were
randomly assigned to receive either hydrocortisone (50
mg i.v. every 6 h) and fludrocortisone (50 g tablet once
daily) (n 151) or matching placebos (n 149) for 7
days. The mortality was 53% in the corticosteroid group
and 63% in the placebo group. Vasopressor therapy was
withdrawn in 40% of patients who received placebo and
in 57% in the corticosteroid group (hazard ratio, 1.91;
95% confidence interval [CI] 1.29 2.84; p 0.001). The
number of patients needed to treat to safe one additional
life at day 28 was 8.
From a practical point of view it is reasonable to
initiate treatment with low dose steroids in the ED or
ICU in any patient presenting with septic shock and
refractory hypotension pending the results of random
cortisol (Figures 1, 2) (16). Due to poor sensitivity of
low-dose and high-dose corticotrophin stimulation testing, these tests are not recommended in the severely
stressed vasopressor-dependant septic shock patient
(13,188 190). Glucocorticoids should be continued in
those patients with a stress cortisol level of 25 g/dL
and in those patients with a level 25 g/dL who have
demonstrated a hemodynamic response (lesser vasopressor requirement) to glucocorticoid replacement (13,188).
We believe this to be a useful (although not the only)
approach to the diagnosis of adrenal failure in the critically ill septic patient until more specific diagnostic tests
become available that can quantitate glucocorticoid activity at the cellular or nuclear level.

ACTIVATED PROTEIN C
Recombinant Human Activated Protein C (rhAPC) is the
first novel agent that has proved effective in the treatment of sepsis (11,191,192). Protein C is a vitamin
K-dependent blood factor that is synthesized in the liver
and secreted as a zymogen (193). Conversion of protein
C to its active form (APC) occurs when it complexes
with thrombin and endothelial cell thrombomodulin. Activated protein C is an endogenous anticoagulant that
inactivates factors Va and VIIIa, thereby preventing the
generation of thrombin and microvascular thrombi formation (11,194). While the anti-thrombotic and profibrinolytic activity of APC preserves and restores microcirculatory blood flow, the anti-inflammatory
property prevents tissue injury and organ dysfunction

194

(195,196). Other anti-inflammatory effects of APC include inhibition of TNF-, macrophage migration inhibitory factor, leukocyte adhesion to Selectins, limiting the
inflammatory response induced by thrombin within the
microvasculature, and decreasing nuclear factor B activation in target cells (197201).
Bernard et al., in a randomized multicenter, doubleblind trial (PROWESS), studied the efficacy of APC on
28-day all-cause mortality rate in patients with severe
sepsis (11). Patients were randomly assigned to receive a
continuous infusion of APC at 24 g/kg/h or placebo
intravenously for 96 h. Subjects were randomized within
24 h of onset of sepsis-related organ failure, such that the
maximum time window for receipt of study drug was
48 h. A total of 1690 randomized patients were treated
(840 in the placebo group and 850 in the APC group).
The mortality rate was 30.8% in the placebo group and
24.7% in the APC group. APC was associated with a
19.4% (95% CI 6.6 30.5) relative risk reduction and
6.1% absolute reduction in the risk of death (p 0.005).
The number of patients needed to treat to save one
additional life was 16. The incidence of serious bleeding
was higher in the APC group than in the placebo group
(3.5% vs. 2.0%, respectively, p 0.06). A post hoc
analysis revealed that the benefit was observed among
subjects who had APACHE II scores of 25 or more (a
13% absolute reduction), whereas the subjects with
lower risk showed no benefit from activated protein C.
Other subgroups in which APC was beneficial included
patients older than 50 years of age, patients with more
than one organ system dysfunction, and in patients who
had shock at the time of the infusion.
In a separate preliminary analysis of the subset of
patients with single organ dysfunction and recent surgery
from a separate, randomized, placebo-controlled study
(ADDRESS) of septic patients at lower risk of death
(APACHE II score 25 or single sepsis-induced organ
failure at any APACHE II score), all-cause mortality was
found to be higher in the APC group (28-day: 67/323;
in-hospital: 76/325) compared with the placebo group
(28-day: 44/313; in-hospital: 62/314). Therefore, APC
may be harmful in less ill patients with single organ
dysfunction alone (APACHE score 25) and cannot be
recommended.
Based on the PROWESS study, recombinant APC is
currently recommended in patients at high risk of death
(APACHE II 25, sepsis-induced multiple organ failure, septic shock, or sepsis-induced acute respiratory
distress syndrome [ARDS]) if there are no absolute contraindications related to bleeding risk or relative contraindication that outweigh the potential benefit of rhAPC
(Figure 1) (16). Based on the PROWESS study, some
authors have advocated the use of APACHE- II scores to
risk stratify patients for whom administration of APC

M. Raghavan and P. E. Marik

would be beneficial (202). However, this approach is


flawed for several reasons. First, APACHE score was
developed for use to include patients with homogenous
severity of illness in clinical trials, and not for decisions
in individual patients (203). Second, APACHE II as
obtained in the PROWESS trial was based on data obtained within the 24 h immediately before study randomization, not the first 24 h in the intensive care unit
(11,193). Third, the inter-observer and intra-observer
variability in the determination of APACHE II scores
among experienced intensive care physicians may be as
high as 10% to 20% (204,205). Fourth, it is unclear
whether or not patients should receive APC if their
APACHE II score falls from, say, 26 to 23 as they are
resuscitated with fluids and vasopressors, before the infusion of the drug has begun, and finally, the use of
APACHE II scores in this new manneras a screening
test to determine which patients should receive a new
therapy and which patients should not has not been
validated.
Currently, it is reasonable that a risk assessment is
best determined by bedside clinical evaluation and judgment. Given the uncertainty of risk assessment and the
potential for rapid deterioration of patients with severe
sepsis and septic shock, the decision to give APC to a
high-risk patient must be made early in the initial golden
hours of onset of sepsis, based on the number of failing
organs rather than scores (Figure 1, Table 1).

MULTIDISCIPLINARY SEPSIS TEAMS


Early recognition and treatment of severe sepsis poses a
considerable problem in the ED (29,51). As early recognition and aggressive resuscitation have been shown to
decrease mortality from septic shock, it would be optimal
to recognize the syndrome as soon as a patient presents
to the ED. Because this is often difficult for practical
purposes, several hospitals, like the University of Pittsburgh Medical Center in Pittsburgh, and Thomas Jefferson University Hospital in Philadelphia, Pennsylvania,
have implemented a multidisciplinary sepsis team for
screening and providing continuous early goal-directed therapy from the ED to the ICU for patients
presenting with sepsis. The team has a specific written
protocol for sepsis management. The team is also responsible for monitoring and improving physician and nurse
compliance with the protocol, as well as continuous
quality assessment, and follow-up of all patients receiving sepsis therapy. These teams currently perform several important tasks such as: (1) to quantify the number
of patients meeting entry criteria for early goal-directed
therapy; (2) to identify barriers to care, such as logistics,
communication, equipment, and disposition from the

Management of Sepsis

ED; (3) to provide a consistent level of response from the


team to the ED, 24 h a day, 7 days a week, including a
critical care medicine physician and extra nursing support; and (4) to facilitate prompt and quick physical
transfer of septic shock patients to the intensive care unit
from the ED, thereby decreasing waiting times.
CONCLUSION
Significant advances have been made in our understanding of sepsis in the last decade, with newer promising
therapies that decrease mortality. Nevertheless, early recognition of severe sepsis in the initial golden hours,
with intervention directed towards physiologic optimization, continues to be a problem in practice. Implementing
measures such as hospital-based sepsis teams, specific
written sepsis protocols, early stabilization and aggressive resuscitation in the emergency department with
early transfer to the ICU may aid in improved outcome
of these patients. In addition, future investigations should
consider timing of intervention and quality of life as a
consequence of delays in institution of antisepsis as an
important endpoint variable. T2
REFERENCES
1. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of
sepsis in the United States from 1979 through 2000. N Engl
J Med 2003;348:1546 54.
2. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo
J, Pinsky MR. Epidemiology of severe sepsis in the United States:
analysis of incidence, outcome, and associated costs of care. Crit
Care Med 2001;29:130310.
3. Padkin A, Goldfrad C, Brady AR, Young D, Black N, Rowan K.
Epidemiology of severe sepsis occurring in the first 24 hrs in
intensive care units in England, Wales, and Northern Ireland. Crit
Care Med 2003;31:2332 8.
4. Vincent JL, Bihari DJ, Suter PM, et al. The prevalence of nosocomial infection in intensive care units in Europe. Results of the
European Prevalence of Infection in Intensive Care (EPIC) Study.
EPIC International Advisory Committee. JAMA 1995;274:639
44.
5. Balk RA. Severe sepsis and septic shock. Definitions, epidemiology, and clinical manifestations. Crit Care Clin 2000;16:179
92.
6. Friedman G, Silva E, Vincent JL. Has the mortality of septic
shock changed with time. Crit Care Med 1998;26:2078 86.
7. Teres D, Rapoport J, Lemeshow S, Kim S, Akhras K. Effects of
severity of illness on resource use by survivors and nonsurvivors
of severe sepsis at intensive care unit admission. Crit Care Med
2002;30:24139.
8. Heyland DK, Hopman W, Coo H, Tranmer J, McColl MA.
Long-term health-related quality of life in survivors of sepsis.
Short Form 36: a valid and reliable measure of health-related
quality of life. Crit Care Med 2000;28:3599 605.
9. 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 77.
10. Van Den BG, Wouters P, Weekers F, et al. Intensive insulin
therapy in the critically ill patients. N Engl J Med 2001;345:
1359 67.

195
11. Bernard GR, Vincent JL, Laterre PF, et al. Efficacy and safety of
recombinant human activated protein C for severe sepsis. N Engl
J Med 2001;344:699 709.
12. Annane D, Sebille V, Charpentier C, et al. Effect of treatment
with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA 2002;288:86271.
13. Marik PE, Zaloga GP. Adrenal insufficiency during septic shock.
Crit Care Med 2003;31:1415.
14. The Acute Respiratory Distress Syndrome Network. Ventilation
with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress
syndrome. N Engl J Med 2000;342:1301 8.
15. Ibrahim EH, Sherman G, Ward S, Fraser VJ, Kollef MH. The
influence of inadequate antimicrobial treatment of bloodstream
infections on patient outcomes in the ICU setting. Chest 2000;
118:146 55.
16. Dellinger RP, Carlet JM, Masur H, et al. Surviving sepsis campaign guidelines for management of severe sepsis and septic
shock. Crit Care Med 2004;32:858 73.
17. Marik PE, Varon J. Sepsis: state of the art. Dis Mon 2001;47:
465532.
18. Bone RC. Inhibitors of complement and neutrophils: a critical
evaluation of their role in the treatment of sepsis. Crit Care Med
1992;20:891 8.
19. Bone RC, Grodzin CJ, Balk RA. Sepsis: a new hypothesis for
pathogenesis of the disease process. Chest 1997;112:235 43.
20. Bone RC, Sibbald WJ, Sprung CL. The ACCP-SCCM consensus
conference on sepsis and organ failure. Chest 1992;101:14813.
21. Marik PE, Varon J. The hemodynamic derangements in sepsis:
implications for treatment strategies. Chest 1998;114:854 60.
22. De Backer D, Creteur J, Preiser JC, Dubois MJ, Vincent JL.
Microvascular blood flow is altered in patients with sepsis. Am J
Respir Crit Care Med 2002;166:98 104.
23. Hinshaw LB. Sepsis/septic shock: participation of the microcirculation: an abbreviated review. Crit Care Med 1996;24:1072 8.
24. Ince C, Sinaasappel M. Microcirculatory oxygenation and shunting in sepsis and shock. Crit Care Med 1999;27:1369 77.
25. Fink MP. Bench-to-bedside review: cytopathic hypoxia. Crit Care
2002;6:4919.
26. Fink MP. Cytopathic hypoxia. Is oxygen use impaired in sepsis as
a result of an acquired intrinsic derangement in cellular respiration? Crit Care Clin 2002;18:16575.
27. Beal AL, Cerra FB. Multiple organ failure syndrome in the 1990s.
Systemic inflammatory response and organ dysfunction. JAMA
1994;271:226 33.
28. Rivers EP, Nguyen HB, Huang DT, Donnino M. Early goaldirected therapy. Crit Care Med 2004;32:314 5.
29. Lundberg JS, Perl TM, Wiblin T, et al. Septic shock: an analysis
of outcomes for patients with onset on hospital wards versus
intensive care units. Crit Care Med 1998;26:1020 4.
30. Nguyen HB, Rivers EP, Havstad S, et al. Critical care in the
emergency department: a physiologic assessment and outcome
evaluation. Acad Emerg Med 2000;7:1354 61.
31. Phillip DR, Parrillo JE. Mediator modulation therapy of severe
sepsis and septic shock: does it work? Crit Care Med 2004;32:
282 6.
32. Natanson C, Esposito CJ, Banks SM. The sirens songs of confirmatory sepsis trials: selection bias and sampling error. Crit
Care Med 1998;26:192731.
33. Eichacker PQ, Parent C, Kalil A, et al. Risk and the efficacy of
antiinflammatory agents: retrospective and confirmatory studies
of sepsis. Am J Respir Crit Care Med 2002;166:1197205.
34. Dellinger RP. Severe sepsis trials: why have they failed? Minerva
Anestesiol 1999;65:340 5.
35. Marik PE, Raghavan M. Stress-hyperglycemia, insulin and immunomodulation in sepsis. Intensive Care Med 2004;30:748 56.
36. Minneci PC, Deans KJ, Banks SM, Eichacker PQ, Natanson C.
Meta-analysis: the effect of steroids on survival and shock during
sepsis depends on the dose. Ann Intern Med 2004;141:4756.
37. Kollef MH, Sherman G, Ward S, Fraser VJ. Inadequate antimi-

196

38.

39.

40.
41.

42.

43.

44.

45.

46.
47.

48.
49.
50.
51.
52.
53.
54.

55.
56.
57.
58.
59.
60.
61.

M. Raghavan and P. E. Marik


crobial treatment of infections: a risk factor for hospital mortality
among critically ill patients. Chest 1999;115:46274.
Kreger BE, Craven DE, McCabe WR. Gram-negative bacteremia.
IV. Re-evaluation of clinical features and treatment in 612 patients. Am J Med 1980;68:344 55.
Marik PE, Zaloga GP. Hypothermia and cytokines in septic
shock. Norasept II Study Investigators. North American study of
the safety and efficacy of murine monoclonal antibody to tumor
necrosis factor for the treatment of septic shock. Intensive Care
Med 2000;26:716 21.
Al Nawas B, Krammer I, Shah PM. Procalcitonin in diagnosis of
severe infections. Eur J Med Res 1996;1:3313.
Luzzani A, Polati E, Dorizzi R, Rungatscher A, Pavan R, Merlini
A. Comparison of procalcitonin and C-reactive protein as markers
of sepsis. Crit Care Med 2003;31:1737 41.
Muller B, Becker KL, Schachinger H, et al. Calcitonin precursors
are reliable markers of sepsis in a medical intensive care unit. Crit
Care Med 2000;28:977 83.
Lind L, Bucht E, Ljunghall S. Pronounced elevation in circulating
calcitonin in critical care patients is related to the severity of
illness and survival. Intensive Care Med 1995;21:63 6.
Nguyen HB, Rivers EP, Knoblich BP, et al. Early lactate clearance is associated with improved outcome in severe sepsis and
septic shock. Crit Care Med 2004;32:1637 42.
Moomey CB Jr, Melton SM, Croce MA, Fabian TC, Proctor KG.
Prognostic value of blood lactate, base deficit, and oxygen-derived variables in an LD50 model of penetrating trauma. Crit Care
Med 1999;27:154 61.
Vitek V, Cowley RA. Blood lactate in the prognosis of various
forms of shock. Ann Surg 1971; 173:308 13.
Grzybowski M. Systemic inflammatory response syndrome criteria and lactic acidosis in the detection of critical illness among
patients presenting to the emergency department. Chest 1996;
110:145S.
James JH Luchette FA, McCarter FD, Fischer JE. Lactate is an
unreliable indicator of tissue hypoxia in injury or sepsis. Lancet
1999;354:505 8.
Bakker J, Gris P, Coffernils M, Kahn RJ, Vincent JL. Serial blood
lactate levels can predict the development of multiple organ
failure following septic shock. Am J Surg 1996;171:221 6.
Broder G, Weil MH. Excess lactate: an index of reversibility of
shock in human patients. Science 1964;143:14579.
Rivers EP, Nguyen HB, Amponsah D. Sepsis: a landscape from
the emergency department to the intensive care unit. Crit Care
Med 2003;31:968 9.
Marik PE, Varon J. Goal-directed therapy for severe sepsis.
N Engl J Med 2002;346:1025 6.
Levy MM, Macias WL, Russell JA, et al. Failure to improve
during first day of therapy is predictive of 28-day mortality in
severe sepsis. Chest Meet Abstr 2003;124:120S.
Shapiro NI, Wolfe RE, Moore RB, Smith E, Burdick E, Bates
DW. Mortality in Emergency Department Sepsis (MEDS) score:
a prospectively derived and validated clinical prediction rule. Crit
Care Med 2003;31:670 5.
Rackow EC, Astiz ME. Pathophysiology and treatment of septic
shock. JAMA 1991;266:548 54.
Marik PE. Gastric intramucosal pH. A better predictor of multiorgan dysfunction syndrome and death than oxygen-derived variables in patients with sepsis. Chest 1993;104:2259.
Cowan BN, Burns HJ, Boyle P, Ledingham IM. The relative
prognostic value of lactate and haemodynamic measurements in
early shock. Anaesthesia 1984;39:750 5.
Tuchschmidt J, Fried J, Swinney R, Sharma OP. Early hemodynamic correlates of survival in patients with septic shock. Crit
Care Med 1989;17:719 23.
Maynard N, Bihari D, Beale R, et al. Assessment of splanchnic
oxygenation by gastric tonometry in patients with acute circulatory failure. JAMA 1993;270:120310.
Ognibene FP. Hemodynamic support during sepsis. Clin Chest
Med 1996;17:279 87.
Ognibene FP, Parker MM, Natanson C, Shelhamer JH, Parrillo

62.

63.

64.

65.

66.
67.
68.
69.

70.
71.

72.
73.

74.

75.

76.
77.

78.
79.
80.

81.
82.
83.

JE. Depressed left ventricular performance. Response to volume


infusion in patients with sepsis and septic shock. Chest
1988;93:90310.
Rackow EC, Falk JL, Fein IA, et al. Fluid resuscitation in circulatory shock: a comparison of the cardiorespiratory effects of
albumin, hetastarch, and saline solutions in patients with hypovolemic and septic shock. Crit Care Med 1983;11:839 50.
Flancbaum L, Dick M, Dasta J, Sinha R, Choban P. A doseresponse study of phenylephrine in critically ill, septic surgical
patients. Eur J Clin Pharmacol 1997;51:4615.
Haupt MT, Gilbert EM, Carlson RW. Fluid loading increases
oxygen consumption in septic patients with lactic acidosis. Am
Rev Respir Dis 1985;131:912 6.
Finfer S, Bellomo R, Boyce N, French J, Myburgh J, Norton R.
A comparison of albumin and saline for fluid resuscitation in the
intensive care unit. N Engl J Med 2004;350:224756.
Marik PE. Assessment of intravascular volume: a comedy of
errors. Crit Care Med 2001;29:1635 6.
Weil MH. Crystalloids, colloids, and fluid compartments. Crit
Care Med 1999;27:3.
Patey R, Wilson G, Hulse T. Albumin controversy continues.
Meta-analysis has affected use of albumin. BMJ 1999;318:464.
Human albumin administration in critically ill patients: systematic review of randomised controlled trials. Cochrane Injuries
Group Albumin Reviewers. BMJ 1998;317:235 40.
Marik PE, Iglesias J. Would the colloid detractors please sit
down! Crit Care Med 2000;28:2652 4.
Ernest D, Belzberg AS, Dodek PM. Distribution of normal saline
and 5% albumin infusions in septic patients. Crit Care Med
1999;27:46 50.
Sibbald WJ, Fox G, Martin C. Abnormalities of vascular reactivity in the sepsis syndrome. Chest 1991;100:155S9S.
Sprung CL, Isikoff SK, Hauser M, et al. Comparison of measured
and calculated colloid osmotic pressure of serum and pulmonary
edema fluid in patients with pulmonary edema. Crit Care Med
1980;8:6135.
Barclay SA, Bennett D. The direct measurement of plasma colloid osmotic pressure is superior to colloid osmotic pressure
derived from albumin or total protein. Intensive Care Med 1987;
13:114 8.
Kellum JA. Fluid resuscitation and hyperchloremic acidosis in
experimental sepsis: improved short-term survival and acid-base
balance with Hextend compared with saline. Crit Care Med
2002;30:300 5.
Kellum JA, Bellomo R, Kramer DJ, Pinsky MR. Etiology of
metabolic acidosis during saline resuscitation in endotoxemia.
Shock 1998;9:364 8.
Rehm M, Orth V, Scheingraber S, Kreimeier U, Brechtelsbauer
H, Finsterer U. Acid-base changes caused by 5% albumin versus
6% hydroxyethyl starch solution in patients undergoing acute
normovolemic hemodilution: a randomized prospective study.
Anesthesiology 2000;93:1174 83.
Scheingraber S, Rehm M, Sehmisch C, Finsterer U. Rapid saline
infusion produces hyperchloremic acidosis in patients undergoing
gynecologic surgery. Anesthesiology 1999;90:126570.
Brill SA, Stewart TR, Brundage SI, Schreiber MA. Base deficit
does not predict mortality when secondary to hyperchloremic
acidosis. Shock 2002;17:459 62.
Wilkes NJ, Woolf R, Mutch M, et al. The effects of balanced
versus saline-based hetastarch and crystalloid solutions on acidbase and electrolyte status and gastric mucosal perfusion in elderly surgical patients. Anesth Analg 2001;93:811 6.
Wilcox CS. Regulation of renal blood flow by plasma chloride.
J Clin Invest 1983;71:726 35.
Ekseth K, Abildgaard L, Vegfors M, Berg-Johnsen J, Engdahl O.
The in vitro effects of crystalloids and colloids on coagulation.
Anaesthesia 2002;57:1102 8.
Gan TJ, Bennett-Guerrero E, Phillips-Bute B, et al. Hextend, a
physiologically balanced plasma expander for large volume use in
major surgery: a randomized phase III clinical trial. Hextend
Study Group. Anesth Analg 1999;88:992 8.

Management of Sepsis
84. Raghavan M, Marik P. Anemia, allogenic blood transfusion and
immune-modulation in the critically ill. Chest 2005;127:295307.
85. Marik PE, Sibbald WJ. Effect of stored-blood transfusion on
oxygen delivery in patients with sepsis. JAMA 1993;269:3024 9.
86. Hebert PC, Wells G, Blajchman MA, et al. A multicenter, randomized, controlled clinical trial of transfusion requirements in
critical care. Transfusion Requirements in Critical Care Investigators, Canadian Critical Care Trials Group. N Engl J Med
1999;340:409 17.
87. Purdy FR, Tweeddale MG, Merrick PM. Association of mortality
with age of blood transfused in septic ICU patients. Can J Anaesth 1997;44:1256 61.
88. De Backer D, Vincent JL. Norepinephrine administration in septic shock: how much is enough? Crit Care Med 2002;30:1398 9.
89. Klinzing S, Simon M, Reinhart K, Bredle DL, Meier-Hellmann
A. High-dose vasopressin is not superior to norepinephrine in
septic shock. Crit Care Med 2003;31:2646 50.
90. 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 8.
91. Yang Y, Qiu HB, Zhou SX, Tan Y, Li SQ. Comparison of
norepinephrine-dobutamine to dopamine alone for splanchnic
perfusion in sheep with septic shock. Acta Pharmacol Sin 2002;
23:1337.
92. Zhou SX, Qiu HB, Huang YZ, Yang Y, Zheng RQ. Effects of
norepinephrine, epinephrine, and norepinephrine-dobutamine on
systemic and gastric mucosal oxygenation in septic shock. Acta
Pharmacol Sin 2002;23:654 8.
93. Martin C, Papazian L, Perrin G, Saux P, Gouin F. Norepinephrine
or dopamine for the treatment of hyperdynamic septic shock?
Chest 1993;103:1826 31.
94. De Backer D, Creteur J, Silva E, Vincent JL. Effects of dopamine,
norepinephrine, and epinephrine on the splanchnic circulation in
septic shock: which is best? Crit Care Med 2003;31:1659 67.
95. Marik PE, Mohedin M. The contrasting effects of dopamine and
norepinephrine on systemic and splanchnic oxygen utilization in
hyperdynamic sepsis. JAMA 1994;272:1354 7.
96. Marik PE. Renal dose norepinephrine! Chest 2004;126:3357.
97. Desjars P, Pinaud M, Bugnon D, Tasseau F. Norepinephrine
therapy has no deleterious renal effects in human septic shock.
Crit Care Med 1989;17:426 9.
98. Third European Consensus Conference in Intensive Care Medicine. Tissue hypoxia: how to detect, how to correct, how to
prevent. Societe de Reanimation de Langue Francaise. The American Thoracic Society. European Society of Intensive Care Medicine. Am J Respir Crit Care Med 1996;154:1573 8.
99. Rudis MI, Basha MA, Zarowitz BJ. Is it time to reposition
vasopressors and inotropes in sepsis? Crit Care Med 1996;24:
52537.
100. Smith LW, Winbery SL, Barker LA, McDonough KH. Cardiac
function and chronotropic sensitivity to beta-adrenergic stimulation in sepsis. Am J Physiol 1986;251:H405H412.
101. Smith LW, McDonough KH. Inotropic sensitivity to beta-adrenergic stimulation in early sepsis. Am J Physiol 1988;255:H699
703.
102. Schreuder WO, Schneider AJ, Groeneveld AB, Thijs LG. Effect
of dopamine vs norepinephrine on hemodynamics in septic shock.
Emphasis
on
right
ventricular
performance.
Chest
1989;95:1282 8.
103. Segal JM, Phang PT, Walley KR. Low-dose dopamine hastens
onset of gut ischemia in a porcine model of hemorrhagic shock.
J Appl Physiol 1992;73:1159 64.
104. Neviere R, Mathieu D, Chagnon JL, Lebleu N, Wattel F. The
contrasting effects of dobutamine and dopamine on gastric mucosal perfusion in septic patients. Am J Respir Crit Care Med
1996;154:1684 8.
105. Olson D, Pohlman A, Hall JB. Administration of low-dose dopamine to nonoliguric patients with sepsis syndrome does not
raise intramucosal gastric pH nor improve creatinine clearance.
Am J Respir Crit Care Med 1996;154:1664 70.

197
106. Santambrogio L, Lipartiti M, Bruni A, Dal Toso R. Dopamine
receptors on human T- and B-lymphocytes. J Neuroimmunol
1993;45:1139.
107. Kouassi E, Li YS, Boukhris W, Millet I, Revillard JP. Opposite
effects of the catecholamines dopamine and norepinephrine on
murine polyclonal B-cell activation. Immunopharmacology 1988;
16:12537.
108. Devins SS, Miller A, Herndon BL, OToole L, Reisz G. Effects
of dopamine on T-lymphocyte proliferative responses and serum
prolactin concentrations in critically ill patients. Crit Care Med
1992;20:1644 9.
109. Bergquist J, Josefsson E, Tarkowski A, Ekman R, Ewing A.
Measurements of catecholamine-mediated apoptosis of immunocompetent cells by capillary electrophoresis. Electrophoresis
1997;18:1760 6.
110. Josefsson E, Bergquist J, Ekman R, Tarkowski A. Catecholamines are synthesized by mouse lymphocytes and regulate
function of these cells by induction of apoptosis. Immunology
1996;88:140 6.
111. Van Den BG, de Zegher F. Anterior pituitary function during
critical illness and dopamine treatment. Crit Care Med 1996;24:
1580 90.
112. Bellomo R, Chapman M, Finfer S, Hickling K, Myburgh J.
Low-dose dopamine in patients with early renal dysfunction: a
placebo-controlled randomised trial. Australian and New Zealand
Intensive Care Society (ANZICS) Clinical Trials Group. Lancet
2000;356:2139 43.
113. Kellum JA, Decker M. Use of dopamine in acute renal failure: a
meta-analysis. Crit Care Med 2001;29:1526 31.
114. Marik PE. Low-dose dopamine: a systematic review. Intensive
Care Med 2002;28:877 83.
115. Jardin F, Fourme T, Page B, et al. Persistent preload defect in
severe sepsis despite fluid loading: A longitudinal echocardiographic study in patients with septic shock. Chest 1999;116:
1354 9.
116. Vieillard-Baron A, Prin S, Chergui K, Dubourg O, Jardin F.
Hemodynamic instability in sepsis: bedside assessment by Doppler echocardiography. Am J Respir Crit Care Med 2003;168:
1270 6.
117. Beaulieu Y, Marik PE. Hand-held ultrasound in the ICU. Part 1
and 2. Chest 2005;128:88195 and 1766 81.
118. Ashrafian H, Bogle RG, Rosen SD, Henein M, Evans TW.
Portable echocardiography. BMJ 2004;328:300 1.
119. Vignon P. Evaluation of fluid responsiveness in ventilated septic
patients: back to venous return. Intensive Care Med 2004;30:
1699 701.
120. Vignon P, Chastagner C, Francois B, et al. Diagnostic ability of
hand-held echocardiography in ventilated critically ill patients.
Crit Care 2003;7:R84 91.
121. Vieillard-Baron A, Augarde R, Prin S, Page B, Beauchet A,
Jardin F. Influence of superior vena caval zone condition on
cyclic changes in right ventricular outflow during respiratory
support. Anesthesiology 2001;95:1083 8.
122. Vieillard-Baron A, Chergui K, Rabiller A, et al. Superior vena
caval collapsibility as a gauge of volume status in ventilated
septic patients. Intensive Care Med 2004;30:1734 9.
123. Feissel M, Michard F, Faller JP, et al. The respiratory variation in
inferior vena cava diameter as a guide to fluid therapy. Intensive
Care Med 2004;30:1834 7.
124. Rodriguez RM, Berumen KA. Cardiac output measurement with
an esophageal doppler in critically ill Emergency Department
patients. J Emerg Med 2000;18:159 64.
125. Weiss SJ, Kulik JP, Calloway E. Bioimpedance cardiac output
measurements in patients with presumed congestive heart failure.
Acad Emerg Med 1997;4:568 73.
126. 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;23:2827.
127. Silverman HJ, Tuma P. Gastric tonometry in patients with sepsis.

198

128.

129.

130.

131.

132.

133.

134.
135.

136.

137.

138.

139.
140.
141.

142.

143.
144.
145.
146.
147.

148.
149.
150.

M. Raghavan and P. E. Marik


Effects of dobutamine infusions and packed red blood cell
transfusions. Chest 1992;102:184 8.
Duke GJ, Briedis JH, Weaver RA. Renal support in critically ill
patients: low-dose dopamine or low-dose dobutamine? Crit Care
Med 1994;22:1919 25.
Mackenzie SJ, Kapadia F, Nimmo GR, Armstrong IR, Grant IS.
Adrenaline in treatment of septic shock: effects on haemodynamics and oxygen transport. Intensive Care Med 1991;17:36 9.
Day NP, Phu NH, Bethell DP, et al. The effects of dopamine and
adrenaline infusions on acid-base balance and systemic haemodynamics in severe infection. Lancet 1996;348:219 23.
Wilson W, Lipman J, Scribante J, et al. Septic shock: does
adrenaline have a role as a first-line inotropic agent? Anaesth
Intensive Care 1992;20:470 4.
Yamazaki T, Shimada Y, Taenaka N, Oshumi H, Takezawa J,
Yoshiya I. Circulatory responses to afterloading with phenylephrine in hyperdynamic sepsis. Crit Care Med 1982;10:4325.
Dunser MW, Mayr AJ, Ulmer H, et al. Arginine vasopressin in
advanced vasodilatory shock: a prospective, randomized, controlled study. Circulation 2003;107:23139.
Reid IA. Role of vasopressin deficiency in the vasodilation of
septic shock. Circulation 1997;95:1108 10.
Landry DW, Levin HR, Gallant EM, et al. Vasopressin deficiency
contributes to the vasodilation of septic shock. Circulation 1997;
95:11225.
Tucci JR, Espiner EA, Jagger PI, Lauler DP, Thorn GW. Vasopressin in the evaluation of pituitary-adrenal function. Ann Intern
Med 1968;69:191202.
Holmes CL, Patel BM, Russell JA, Walley KR. Physiology of
vasopressin relevant to management of septic shock. Chest 2001;
120:989 1002.
Annane D, Sebille V, Troche G, Raphael JC, Gajdos P, Bellissant
E. A 3-level prognostic classification in septic shock based on
cortisol levels and cortisol response to corticotropin. JAMA 2000;
283:1038 45.
Van Haren FM, Rozendaal FW, van der Hoeven JG. The effect of
vasopressin on gastric perfusion in catecholamine-dependent patients in septic shock. Chest 2003;124:2256 60.
Michard F, Teboul JL. Predicting fluid responsiveness in ICU
patients: a critical analysis of the evidence. Chest 2002;121:
2000 8.
Lichtwarck-Aschoff M, Zeravik J, Pfeiffer UJ. Intrathoracic
blood volume accurately reflects circulatory volume status in
critically ill patients with mechanical ventilation. Intensive Care
Med 1992;18:1427.
Richard C, Warszawski J, Anguel N, et al. Early use of the
pulmonary artery catheter and outcomes in patients with shock
and acute respiratory distress syndrome: a randomized controlled
trial. JAMA 2003;290:271320.
Raper R, Sibbald WJ. Misled by the wedge? The Swan-Ganz
catheter and left ventricular preload. Chest 1986;89:42734.
Marik PE, Bankov A. Sublingual capnometry versus traditional
markers of tissue oxygenation in critically ill patients. Crit Care
Med 2003;31:818 22.
Marik PE. Sublingual capnography: a clinical validation study.
Chest 2001;120:9237.
Brady HR, Brenner BM, Clarkson MR, et al. Acute renal failure.
In: Brenner BM, ed. Brenner & Rectors the kidney. Philadelphia,
PA: WB Saunders; 2000:1201 62.
Kumar A, Anel R, Bunnell E, 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;32:6919.
Sandham JD, Hull RD, Brant RF, et al. A randomized, controlled
trial of the use of pulmonary-artery catheters in high-risk surgical
patients. N Engl J Med 2003;348:514.
Connors AF Jr, Speroff T, Dawson NV, et al. The effectiveness
of right heart catheterization in the initial care of critically ill
patients. SUPPORT Investigators. JAMA 1996;276:889 97.
Iberti TJ, Fischer EP, Leibowitz AB, Panacek EA, Silverstein JH,
Albertson TE. A multicenter study of physicians knowledge of

151.

152.

153.

154.

155.

156.

157.

158.

159.
160.
161.

162.
163.

164.
165.

166.

167.
168.
169.
170.

the pulmonary artery catheter. Pulmonary Artery Catheter Study


Group. JAMA 1990;264:2928 32.
Richards MJ, Edwards JR, Culver DH, et al. Nosocomial infections in combined medical-surgical intensive care units in the
United States. Infect Control Hosp Epidemiol 2000;21:510 5.
Weinstein MP, Towns ML, Quartey SM, et al. The clinical
significance of positive blood cultures in the 1990s: a prospective
comprehensive evaluation of the microbiology, epidemiology,
and outcome of bacteremia and fungemia in adults. Clin Infect
Dis 1997;24:584 602.
Kollef MH. Inadequate antimicrobial treatment: an important
determinant of outcome for hospitalized patients. Clin Infect Dis
2000;31:S131 8.
Osmon S, Ward S, Fraser VJ, Kollef MH. Hospital mortality for
patients with bacteremia due to Staphylococcus aureus or Pseudomonas aeruginosa. Chest 2004;125:60716.
Leibovici L, Shraga I, Drucker M, Konigsberger H, Samra Z,
Pitlik SD. The benefit of appropriate empirical antibiotic treatment in patients with bloodstream infection. J Intern Med 1998;
244:379 86.
Romero-Vivas J, Rubio M, Fernandez C, Picazo JJ. Mortality
associated with nosocomial bacteremia due to methicillin-resistant Staphylococcus aureus. Clin Infect Dis 1995;21:141723.
Rangel-Frausto MS, Pittet D, Costigan M, Hwang T, Davis CS,
Wenzel RP. The natural history of the systemic inflammatory
response syndrome (SIRS). A prospective study. JAMA 1995;
273:11723.
Sands KE, Bates DW, Lanken PN, et al. Epidemiology of sepsis
syndrome in 8 academic medical centers. Academic Medical
Center Consortium Sepsis Project Working Group. JAMA 1997;
278:234 40.
Maki DG. Microbiologic diagnosis of blood culture-negative
sepsis by hemofiltration. Crit Care Med 2004;32:10757.
Mermel LA, Maki DG. Detection of bacteremia in adults: consequences of culturing an inadequate volume of blood. Ann Intern
Med 1993;119:270 2.
Rodriguez RM, Fancher ML, Phelps M, et al. An emergency
department-based randomized trial of nonbronchoscopic bronchoalveolar lavage for early pathogen identification in severe
community-acquired pneumonia. Ann Emerg Med 2001;38:357
63.
Said-Salim B, Mathema B, Kreiswirth BN. Community-acquired
methicillin-resistant Staphylococcus aureus: an emerging pathogen. Infect Control Hosp Epidemiol 2003;24:4515.
Valles J, Rello J, Ochagavia A, Garnacho J, Alcala MA. Community-acquired bloodstream infection in critically ill adult patients: impact of shock and inappropriate antibiotic therapy on
survival. Chest 2003;123:161524.
Wagenvoort JH, Kepers-Rietrae M. Methicillin-resistant Staphylococcus aureus (MRSA) as a community strain. Euro Surveill
1997;2:96 7.
Zinn CS, Westh H, Rosdahl VT. An international multicenter
study of antimicrobial resistance and typing of hospital Staphylococcus aureus isolates from 21 laboratories in 19 countries or
states. Microb Drug Resist 2004;10:160 8.
Frazee BW, Lynn J, Charlebois ED, Lambert L, Lowery D,
Perdreau-Remington F. High relevance of methicillin-resistant
Staphylococcus aureus in Emergency department skin and soft
tissue infections. Ann Emerg Med 2005;45:31120.
Raghavan M, Linden PK. Newer treatment options for skin and
soft tissue infections. Drugs 2004;64:1621 42.
Bukharie HA, Abdelhadi MS, Saeed IA, Rubaish AM, Larbi EB.
Emergence of methicillin-resistant Staphylococcus aureus as a
community pathogen. Diagn Microbiol Infect Dis 2001;40:1 4.
Broom AV, Wolsey DH, Naimi TS, et al. Community-acuired
methicillin-resistant Staphylococcus aureus in a rural American
Indian community. JAMA 2001;286:12015.
Naimi TS, LeDell KH, Boxrud DJ, et al. Epidemiology and
clonality of community-acquired methicillin-resistant Staphylococcus aureus in Minnesota, 1996 1998. Clin Infect Dis 2001;
33:990 6.

Management of Sepsis
171. Cunha BA. Antibiotic treatment of sepsis. Med Clin North Am
1995;79:551 8.
172. Leibovici L, Paul M, Poznanski O, et al. Monotherapy versus
beta-lactam-aminoglycoside combination treatment for gramnegative bacteremia: a prospective, observational study. Antimicrob Agents Chemother 1997;41:112733.
173. Rybak MJ, McGrath BJ. Combination antimicrobial therapy for
bacterial infections. Guidelines for the clinician. Drugs 1996;52:
390 405.
174. Marik PE, Lipman J, Kobilski S, Scribante J. A prospective
randomized study comparing once- versus twice-daily amikacin
dosing in critically ill adult and paediatric patients. J Antimicrob
Chemother 1991;28:753 64.
175. Prins JM, Buller HR, Kuijper EJ, Tange RA, Speelman P. Once
versus thrice daily gentamicin in patients with serious infections.
Lancet 1993;341:3359.
176. Marik PE. Failure of once-daily vancomycin for staphylococcal
endocarditis. Pharmacotherapy 1998;18:650 2.
177. Craig WA, Ebert SC. Continuous infusion of beta-lactam antibiotics. Antimicrob Agents Chemother 1992;36:2577 83.
178. OGrady NP, Alexander M, Dellinger EP, et al. Guidelines for the
prevention of intravascular catheter-related infections. Centers for
Disease Control and Prevention. MMWR Recomm Rep 2002;51:
129.
179. Marik PE, Zaloga GP. Adrenal insufficiency in the critically ill: a
new look at an old problem. Chest 2002;122:1784 96.
180. Cooper MS, Stewart PM. Corticosteroid insufficiency in acutely
ill patients. N Engl J Med 2003;348:72734.
181. Annane D, Bellissant E, Bollaert PE, Briegel J, Keh D, Kupfer Y.
Corticosteroids for treating severe sepsis and septic shock. Cochrane Database Syst Rev 2004;CD002243.
182. Keh D, Boehnke T, Weber-Cartens S, et al. Immunologic and
hemodynamic effects of low-dose hydrocortisone in septic
shock: a double-blind, randomized, placebo-controlled, crossover
study. Am J Respir Crit Care Med 2003;167:51220.
183. Briegel J, Jochum M, Gippner-Steppert C, Thiel M. Immunomodulation in septic shock: hydrocortisone differentially regulates cytokine responses. J Am Soc Nephrol 2001;12:S70 4.
184. Briegel J. Hydrocortisone and the reduction of vasopressors in
septic shock: therapy or only chart cosmetics? Intensive Care
Med 2000;26:1723 6.
185. Briegel J, Forst H, Haller M, et al. Stress doses of hydrocortisone
reverse hyperdynamic septic shock: a prospective, randomized,
double-blind, single-center study. Crit Care Med 1999;27:723
32.
186. Briegel J, Schelling G, Haller M, Mraz W, Forst H, Peter K. A
comparison of the adrenocortical response during septic shock
and after complete recovery. Intensive Care Med 1996;22:894 9.
187. Marik P, Zaloga G. Prognostic value of cortisol response in septic
shock. JAMA 2000;284:308 9.
188. Marik PE. Unraveling the mystery of adrenal failure in the
critically ill: the author replies. Crit Care Med 2004;32:1448.

199
189. Marik PE. Unraveling the mystery of adrenal failure in the
critically ill. Crit Care Med 2004;32:596 7.
190. Mayenknecht J, Diederich S, Bahr V, Plockinger U, Oelkers W.
Comparison of low and high dose corticotropin stimulation tests
in patients with pituitary disease. J Clin Endocrinol Metab 1998;
83:1558 62.
191. Ely EW, Angus DC, Williams MD, Bates B, Qualy R, Bernard
GR. Drotrecogin alfa (activated) treatment of older patients with
severe sepsis. Clin Infect Dis 2003;37:18795.
192. Bernard GR, Margolis BD, Shanies HM, et al. Extended evaluation of recombinant human activated protein C United States
Trial (ENHANCE US): a single-arm, phase 3B, multicenter study
of drotrecogin alfa (activated) in severe sepsis. Chest 2004;125:
2206 16.
193. Bernard GR. Drotrecogin alfa (activated) (recombinant human
activated protein C) for the treatment of severe sepsis. Crit Care
Med 2003;31:S8593.
194. Matthay MA. Severe sepsisa new treatment with both anticoagulant and antiinflammatory properties. N Engl J Med 2001;344:
759 62.
195. Ten Cate H. Pathophysiology of disseminated intravascular coagulation in sepsis. Crit Care Med 2000;28:S9 11.
196. Bajzar L, Nesheim ME, Tracy PB. The profibrinolytic effect of
activated protein C in clots formed from plasma is TAFI-dependent. Blood 1996;88:2093100.
197. Rosenberg RD, Aird WC. Vascular-bed-specific hemostasis and
hypercoagulable states. N Engl J Med 1999;340:1555 64.
198. Schmidt-Supprian M, Murphy C, While B, et al. Activated protein C inhibits tumor necrosis factor and macrophage migration
inhibitory factor production in monocytes. Eur Cytokine Netw
2000;11:40713.
199. Grinnell BW, Hermann RB, Yan SB. Human protein C inhibits
selectin-mediated cell adhesion: role of unique fucosylated oligosaccharide. Glycobiology 1994;4:2215.
200. Joyce DE, Gelbert L, Ciaccia A, DeHoff B, Grinnell BW. Gene
expression profile of antithrombotic protein c defines new mechanisms modulating inflammation and apoptosis. J Biol Chem
2001;276:11199 203.
201. Yan SB, Dhainaut JF. Activated protein C versus protein C in
severe sepsis. Crit Care Med 2001;29:S69 74.
202. Siegel JP. Assessing the use of activated protein C in the treatment of severe sepsis. N Engl J Med 2002;347:1030 4.
203. Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE
II: a severity of disease classification system. Crit Care Med
1985;13:818 29.
204. Polderman KH, Christiaans HM, Wester JP, Spijkstra JJ, Girbes
AR. Intra-observer variability in APACHE II scoring. Intensive
Care Med 2001;27:1550 2.
205. Warren HS, Suffredini AF, Eichacker PQ, Munford RS. Risks
and benefits of activated protein C treatment for severe sepsis.
N Engl J Med 2002;347:102730.

Potrebbero piacerti anche