Sie sind auf Seite 1von 9

Risk and the Efficacy of Antiinflammatory Agents

Retrospective and Confirmatory Studies of Sepsis


Peter Q. Eichacker, Chantal Parent, Andre Kalil, Claire Esposito, Xizhong Cui, Steven M. Banks,
Eric P. Gerstenberger, Yvonne Fitz, Robert L. Danner, and Charles Natanson
Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland

We investigated whether a relationship between risk of death and


treatment effect could explain the disparate results between the
preclinical and clinical sepsis trials of antiinflammatory agents over
the last decade. A metaregression analysis of cited preclinical studies
showed that the treatment effects of these agents were highly
dependent on risk of death (p 0.0001) and that animals were
studied at significantly higher control mortality rates than humans
(median [25th75th quartile], 88% [7996%] versus 39% [3242%],
p 0.0001). An analysis of the clinical trials showed that antiinflammatory agents were also significantly more efficacious in septic
patients with higher risk of death (p 0.002) and were harmful
in those with low risk. To test this relationship prospectively, we
studied antiinflammatory agents in models employing differing
doses of bacterial challenge to produce the full range of risk of
death. We found that the efficacy of these agents, although very
beneficial at high control mortality rates, was much reduced (p
0.0001) and similar to those in human trials at moderate control
mortality rates (i.e., 30 to 40%). The efficacy of antiinflammatory
agents during sepsis is dependent on the risk of death, an observation that explains the apparent contradiction between preclinical
and clinical trial results. Accounting for this relationship may be
necessary for the safe and effective development of antiinflammatory therapies for sepsis.
Keywords: septic shock; preclinical and clinical trials; metaregression
analysis; antiinflammatory therapies

Despite promising preclinical testing (138) and the expenditure of several billion dollars, antiinflammatory agents designed to inhibit specific host mediators during sepsis failed
to show benefit in 22 clinical trials involving over 10,000
patients (Table 1) (3960). This failure for the global pharmaceutical industry and academic community has been much
discussed but never adequately explained. The underlying
hypothesis of this therapeutic approachthat the intense
host inflammatory response becomes pathogenic in severe
sepsis and septic shockhas been questioned but not refuted
(6165). Even if this hypothesis is incorrect or its successful
application requires the blockade of different or multiple targets in the host response to infection, it would not explain
the marked disparity in the efficacy of these mediator-specific
antiinflammatory agents comparing animal models of sepsis to
infected patients. Identifying factors underlying this difference
between clinical and preclinical trials would likely strengthen

(Received in original form April 9, 2002; accepted in final form July 16, 2002)
Correspondence and requests for reprints should be addressed to Peter Q. Eichacker, M.D., Critical Care Medicine Department, National Institutes of Health,
Building 10, Room 7D43, 10 Center Drive, MSC 1662, Bethesda, MD 208921662.
E-mail: peichacker@nih.gov
This article has an online data supplement, which is accessible from this issues
table of contents online at www.atsjournals.org
Am J Respir Crit Care Med Vol 166. pp 11971205, 2002
Originally Published in Press as DOI: 10.1164/rccm.200204-302OC on July 19, 2002
Internet address: www.atsjournals.org

the development and application not only of antiinflammatory


agents but also of other new therapies for sepsis.
Clinical investigators have suggested that the severity of
the underlying septic process and its associated risk of death
may be linked to the treatment effect of antiinflammatory
agents in sepsis (66). If preclinical and clinical testing was
performed at very different control group mortality rates,
then a strong relationship between risk of death and efficacy
could explain why animal studies and patient trials of antiinflammatory agents produced such divergent results. For example, a drug with this profile might be effective only at high
and not at low risks of death. The results of preclinical testing
would be misleading unless clinical studies were conducted
at a risk of death similar to those found in clinical trials.
Also, at some levels of risk, agents might become harmful,
leading to excess deaths in clinical trials. Such a relationship
between the treatment effect of an agent and risk of death
has been previously proposed for several other potentially
fatal conditions (67) such as antiarrhythmics for sudden death
(68), magnesium for myocardial infarction (69), cholesterollowering drugs for atherosclerosis (70), and tocolytic agents
(71) for premature birth.
To test the potential relationship between risk of death
and the efficacy of antiinflammatory agents in sepsis, we first
examined the published preclinical trials used to support
testing mediator-specific antiinflammatory agents in septic
patients. None of these individual animal studies examined
this relationship. We then analyzed the treatment effect across
published studies in a metaregression analysis to see whether
it changed as risk of death varied. This led us to perform
a similar analysis of the clinical trials themselves, looking
separately at those trials that stratified patients treatment
effect based on their predicted risk of death and then those
that did not. Finally, we prospectively tested in our own
laboratories the influence of risk of death on antiinflammatory agents by altering the dose of bacterial-challenge animals
received and thereby mortality rates. This allowed us to examine animal studies at lower risks of death more similar to
human sepsis studies. In addition, the route and type of
challenge and type of antiinflammatory agent studied were
varied because these factors had also been proposed as a
basis for the differing effects of antiinflammatory agents (61
65). As a control, we tested the influence of risk of death on
fluid therapy, a conventional treatment for sepsis not known
to have antiinflammatory effects.
Our metaregression analyses of published preclinical and
clinical sepsis trials and confirmatory prospective animal sepsis studies each showed similarly that the risk of death influences the effects of antiinflammatory agents in sepsis. This
relationship explains the contradictory results noted in prior
preclinical and clinical trials. More importantly, accounting
for this relationship may be necessary for the safe and effective development of new agents for the treatment of sepsis
and septic shock.

1198

AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 166 2002
TABLE 1. RESULTS OF CLINICAL TRIALS OF MEDIATOR-SPECIFIC ANTIINFLAMMATORY AGENTS
TESTED IN SEPTIC PATIENTS

Antiinflammatory Agent (Company) (Reference)


Anti-TNF antibodies
MAK 195F (Knoll) (39)
CDP571 (Celltech) (40)
CA2 (Centacor) (45)
CB006 (Celltech) (41)
MAK 195F (Knoll) (42)
MAK 195F (Knoll) (46)
BAYx1351 (Bayer/Miles) (43)
BAYx1351 (Bayer/Miles) (44)
BAYx1351 (Bayer/Miles) (47)
TNF-soluble receptors
P80 (Immunex) (48)
P55 (Hoffman) (49)
P55 (Hoffman) (50)
IL-1ra
Antril (Synergen) (51)
Antril (Synergen) (52)
Antril (Synergen) (53)
PAFra
BN 52021 (Ipsen) (54)
BN 52021 (Ipsen) (55)
Antiprostaglandin
Ibuprofen (Upjohn) (56)
Ibuprofen (Upjohn) (57)
Ibuprofen (Upjohn) (58)
Antibradykinin
CPO127 (Cortech) (59)
CPO127 (Cortech) (60)

Odds Ratio
(95% Confidence Interval
for Survival )*

Number of
Patients Enrolled

Control
Mortality Rate

39
42
56
80
122
446
553
971
1,878

50%
60%
39%
32%
41%
57%
40%
33%
43%

2.86
0.90
1.16
0.58
0.79
1.12
1.10
1.13
1.11

141
498
1,362

30%
39%
28%

0.52 (0.23, 1.20)


1.02 (0.69, 1.53)
1.12 (0.88, 1.43)

99
893
906

44%
34%
36%

2.44 (0.94, 6.33)


1.19 (0.89, 1.60)
1.10 (0.84, 1.45)

262
608

51%
49%

1.44 (0.89, 2.35)


1.11 (0.81, 1.53)

29
30
455

31%
43%
40%

0.35 (0.07, 1.61)


3.25 (0.63, 16.79)
1.12 (0.77, 1.64)

251
504

29%
41%

0.68 (0.38, 1.20)


1.07 (0.71, 1.61)

(0.69,
(0.21,
(0.39,
(0.20,
(0.34,
(0.77,
(0.76,
(0.85,
(0.92,

11.84)
3.85)
3.44)
1.73)
1.83)
1.63)
1.59)
1.51)
1.33)

Definition of abbreviations: IL-1ra interleukin one receptor antagonist; PAFra platelet-activating factor receptor antagonist;
TNF tumor necrosis factor.
* We averaged over dose of drug in each trial to increase our ability to find significant effects because there were no significant
effects of dose on survival rates with the exception that the high and middle dose of P-80 TNF-soluble receptor significantly
increased mortality rates compared with low dose and controls (48) (modified and updated from Zeni and colleagues [72]).

METHODS
Analysis of Published Preclinical Studies
Twenty-two clinical trials (3960) were conducted between January
1986 and January 2000 that tested the effects of seven different nonsteroidal mediator-specific antiinflammatory agents (i.e., anti-TNF [tumor
necrosis factor] antibodies, P-55 and P-80 TNF-soluble receptors [sometimes referred to as the P-75 TNF-soluble receptor], interleukin-1 receptor antagonist, platelet-activating factor receptor antagonist, antibradykinin, and antiprostaglandin) (see online data supplement) (72). To
investigate the basis for the differing effects that these agents were
reported to have in animal models versus human trials, all preclinical
studies cited to support their use in clinical trials were reviewed for
this metaregression analysis (138). All reported experiments in these
studies were included provided that survival rates of the treatment
agent were directly compared with a survival rate of a placebo or lower
dose treatment group. In addition, in all experiments, the type and
route of septic challenge, the regimen of treatment employed, including
both timing and dose, the species studied, and the duration of observation were recorded for analysis.
For each experiment, treatment mortality rate was plotted on the
y axis and the corresponding control mortality rate on the x axis, yielding
a graph on the unit square in which each axis represents fully independent measures. To avoid constraining data at the extremes of the mortality rate ranges, we reparameterized the unit square to occupy the whole
plane (73). The y axis was transformed to become the log of the odds
of treatment mortality and the x axis to become the log of the odds of
control mortality. A weighted linear regression based on the number
of animals in each study was performed. Although the analysis was
always done with the y axis expressed as the log of the odds of treatment
mortality, for ease of presentation, the results are presented with the

y axis expressed as the log odds ratio of survival. The odds ratio of
survival is the odds of survival in the treatment group divided by the
odds of survival in the control group. The odds ratio of survival is
mathematically equivalent and is a commonly used measure of efficacy
in clinical trial literature. No other factor examined (i.e., type or route
of septic challenge, regimen of treatment, including timing and dose,
duration of observation, or species studied) significantly changed (all
p 0.20) the relationship between treatment effect and control mortality rate (74).

Analysis of Published Clinical Trials


We assessed the consistency of the clinical trials by determining whether
the odds ratios of the 22 human sepsis trials fell within the 95% confidence interval for the mean regression line calculated with the preclinical animal studies. In addition, two trials stratified patients based on
the predicted risk of death, and these trials, along with the remaining
20 studies, were analyzed in a manner similar to the published preclinical
studies.

Prospective Preclinical Studies


To examine prospectively the relationship between risk of death and
treatment effect with antiinflammatory agents in sepsis, Sprague-Dawley rats (n 1,296) were randomized to be challenged intravenously
or intrabronchially with varying doses of Escherichia coli 0111:B4,
Staphylococcus aureus (0.25 to 100 109 CFU kg1 body weight), or
E. coli 0111:B4 endotoxin (0.75 to 2 g kg1; Sigma, St. Louis, MO).
These differing bacterial doses, types, and routes of challenge were
studied to attempt to simulate the diverse types of infection that would
be present in a population of septic patients producing a wide range
of mortality rates. We studied antiinflammatory agents with different

Eichacker, Parent, Kalil, et al.: Risk Alters Antiinflammatory Therapies

1199

mechanisms of action to examine prospectively whether the effect of


risk of death is dependent on one type of immunomodulator. After
challenge, one group of animals (n 653) was treated with high molecular weight TNF-soluble receptor (P-80 TNF-soluble receptor; 100 g
kg1 intravenously; Immunex, Seattle, WA) or placebo. This agent was
significantly harmful in a clinical sepsis trial (48) after animal studies
had shown it to be beneficial in a highly lethal gram-negative bacterial
or bacterial toxin model (23). Another group of animals (n 638)
received a tyrosine kinase inhibitor (7581) (tyrphostin AG556, 2.5 g
kg1, intraperitoneally) or placebo given immediately and 6 hours after
challenge to examine an antiinflammatory agent that blocks the release
of multiple inflammatory mediators simultaneously (7581) rather than
directly neutralizing circulating TNF. To investigate whether risk of
death affected the efficacy of a conventional sepsis treatment, an additional group of rats (n 192) was challenged with varying doses of E.
coli and then treated with or without normal saline (20 ml kg1h1
intravenously for 24 hours) (82). All animals received daily antibiotics
for 3 days, and after 7 days, animals were considered survivors as
previously described (83).
The relationship between risk of death and treatment efficacy was
examined in a manner similar to the published animal studies, and
these relationships were not significantly changed (p 0.20) by any of
the other variables studied. The consistency of this data with published
preclinical and clinical trials was assessed as previously described.
All animal protocols used in these studies were approved by the
Animal Care and Use Committee of the Clinical Center of the National
Institutes of Health. Every effort was made to minimize animal suffering
during these protocols. The research protocols required the veterinarian
staff or principle investigators to euthanize any animal that experienced
unexpected severe pain or distress.

The differing control mortality rates reported for these subgroups allowed us to examine the relationship between risk of
death and treatment effect over a wider range. In a phase III
trial of interleukin-1 receptor antagonist (53, 66) and a phase
III trial of P-55 TNF-soluble receptor (50), patients were categorized into groups with increasing risks of death as determined
by physiology-based scoring systems (modified Acute Physiology
and Chronic Health Evaluation III score and Stratified Acute
Physiology score, respectively) (Figures 3A and 3C). Stratifying
by the risk of death score, we found that the treatment effect of
these two antiinflammatory agents was significantly (p 0.0002)
related to the risk of death (Figures 3A and 3C). The effect of
these antiinflammatory agents in septic humans at higher risks
of death was very similar to the effects seen with these agents
in published animal studies with the same high risks (Figure 1A).
The 20 remaining clinical trials studied control mortality rates
over a significantly narrower range of overall control mortality
rates than the two trials categorizing patients by risk of death
(29 to 60% versus 10 to 78%, p 0.0001) (Figures 3A and 3B).
Despite this, the relationship between risk of death and the
treatment effect for these 20 remaining clinical trials had an
identical slope and intercept to the one based on patients segregated by risk of death in the other two trials (both p 0.85
comparing the two groups of trials) (Figures 3A and 3B). Moreover, even within the narrower range of control mortality rates
studied in these other 20 trials, the slope for this relationship
between risk of death and treatment effect approached being
significant (p 0.07).

RESULTS

Prospective Preclinical Studies with Antiinflammatory


Therapies or Fluids

Published Preclinical Studies of Antiinflammatory Agents

In 22 human sepsis trials of antiinflammatory agents (Table 1)


(3960), a total of 38 animal sepsis studies were cited (95 individual experiments; Figures 1A1C) (138). Animals were studied
at much higher control mortality rates than humans (median
[25th75th quartile] 88% [7996%] versus 39% [3243%], p
0.0001) (Figures 1B and 1C). Regardless of the type of challenge,
timing or dose of treatment, species studied, or duration of
observation, the odds ratio of survival with all mediator-specific
antiinflammatory agents diminished as the risk of death (control
mortality rate studied) decreased (p 0.0001) (Figure 1). Notably, anti-TNF antibodies were found to be significantly harmful
in two animal studies cited with very low (less than 15%) control
mortality rates that were below those studied in most clinical
and preclinical sepsis trials (17, 18). Each of these two studies
significantly increased the slope of the relationship between risk
of death and treatment. Although shown with other data, because these two studies were overly influential, they were excluded from analysis; however, inclusion would make this relationship even stronger. Among all factors examined, risk of death
explained the majority of the variability (70%) in the treatment
effects of antiinflammatory agents comparing these 95 experiments.
Published Clinical Trials of Mediator-Specific
Antiinflammatory Therapies

Control mortality rates in clinical trials were lower and studied


over a narrower range than in preclinical trials (Figures 1B and
1C). At comparable control mortality rates, the effects of mediator-specific antiinflammatory therapies in 19 (3941, 4346, 48
54, 5660) of the 22 (3960) clinical trials fit within the 95%
confidence interval for the odds ratio for survival predicted from
the 95 published preclinical experiments (Figure 2A). However,
two clinical trials provided data that categorized patients at study
entry into subgroups based on predicted risk of death (50, 53).

In prospective experiments testing P-80 TNF-soluble receptor


and tyrphostin AG556 in rats, increasing the dose of each type
of challenge (E. coli, S. aureus, or endotoxin) caused mortality
rates to increase (combined; r 0.83; p 0.0001) (data not
shown). Regardless of the type or route of challenge or the type
of antiinflammatory agent used, as the control mortality rates
increased, the odds ratio of survival with antiinflammatory therapy increased (combined, p 0.0002) (Figures 4A and 4C).
For our prospective and previously unpublished studies
(Figure 4A) and the published preclinical studies from other
laboratories (Figure 1A) of antiinflammatory therapies, the
slopes of this regression relationship comparing risk of death
and treatment effect were similar (0.40 versus 0.48, respectively,
p NS). However, the y intercept was greater in the published
compared with the prospective preclinical studies (2.95 versus
1.17, respectively, p 0.0001) (Figures 1A and 4A). The greater
beneficial effects and increases in y intercept in published versus
prospective studies can be partially explained as positive results
are more likely to be published, causing an overestimation of
treatment effects (84). All 22 clinical trials fit within the 95%
confidence interval for the odds of survival in these prospective
preclinical trials of antiinflammatory agents (Figure 2B).
In this preclinical model, fluid therapy had a significantly
different relationship (i.e., slope and intercept) between risk of
death and treatment effect (Figure 4B) compared with antiinflammatory agents in prospective studies (p 0.008 for both
slope and intercept) (Figure 4A). After E. coli challenges, fluids
produced significant beneficial effects that were similar regardless of control mortality rate. At comparable control mortality
rates, only 4 (39, 45, 51, 57) of the 22 (3960) clinical trials fit
within the 95% confidence interval for the effects of fluid therapy
in animals (Figure 2C). Notably, more of the 22 clinical trials
fit within the 95% confidence intervals for survival with antiinflammatory therapies than with fluid therapy in animals (22 of
22 versus 4 of 22, respectively; p 0.0001) (Figures 2B and 2C).

1200

AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 166 2002

Figure 1. (AC) The weighted regression


line in A shows the relationship between
control odds and the odds ratio of survival
with treatment for 95 experiments (open circles) in 38 published animal studies cited
(138) in 22 clinical sepsis trials of mediatorspecific antiinflammatory agents (Table 1)
(3960). B is the same as A but also shows
the control odds and odds ratio for survival
for the 22 clinical trials (open diamonds) (39
60) testing mediator-specific antiinflammatory agents. C divides the data in B by type
of antiinflammatory agent. The circle size in
each panel is adjusted for the number of
animals in each experiment, but not the diamond size in B and C because the number
of patients in each trial was not used to calculate the weighted regression line.

Comparison of Time of Observation Used to Calculate Survival


Rates in Animals and Human Sepsis Studies

In this meta-analysis, we analyzed survival rates and not survival


times because this was the only outcome measure universally
reported. Differences in follow-up times within species or differing life spans across species could potentially produce differences in control mortality rates and thereby alter the relationship
between control mortality rate and treatment effect. The median
(range) duration of follow-up for determining survival rates in
published animal studies was 168 hours (24 to 288 hours) in rats,
75 hours (24 to 360 hours) in mice, 72 hours (24 to 336 hours)
in primates, and 24 hours (5 to 168 hours) in rabbits. For these
previously published animal studies, there was no significant
correlation between control mortality rate and the length of
follow-up time within or across these species (r values, 0.30 to
0.14; p values, 0.40 to 0.98). Therefore, in the published animal
studies, differences in follow-up times cannot explain the significant relationship found between increasing control mortality
rate and the treatment effects of antiinflammatory agents. All
of our prospective animal studies were based on 7-day survival
rates, and all of the human clinical sepsis trials were based on
28- to 30-day survival rates. Therefore, differences in follow-

up times also cannot explain the significant relationship found


between control mortality rate and treatment effects of antiinflammatory agents in either our animal studies or the human
sepsis trials. Finally, because the time of observation after treatment was actually much longer in human sepsis trials than all
of the preclinical sepsis studies examined, it is unlikely that this
variable caused the significantly higher control mortality rates
found in animal compared with human sepsis studies.

DISCUSSION
In a retrospective analysis of clinical and preclinical trials and
confirmatory animal studies, we found that risk of death altered
the efficacy of antiinflammatory agents during sepsis. This relationship provides an explanation for the contradictory results
noted with this therapeutic approach in preclinical (138) and
clinical sepsis trials (3960) over the past decade. Antiinflammatory agents had greater treatment effects in animal models compared with clinical trials because preclinical studies were done
at significantly higher risks of death (Figure 1). Furthermore,
human trials of antiinflammatory therapies failed because they
enrolled patients at a level of risk where these agents have
minimal effects (Figure 1 and Table 1). Animal studies we con-

Eichacker, Parent, Kalil, et al.: Risk Alters Antiinflammatory Therapies


Figure 2. A compares the
treatment effect of mediator-specific antiinflammatory agents in 22 clinical trials (3960) (open diamonds)
(Table 1) to the 95% confidence interval (gray bar)
for the regression line describing their effects in
cited animal studies (138)
from Figure 1A. B and C
compare these same 22
clinical trials to the 95%
confidence interval (gray
bars) for the regression line
describing their effects in
prospective animal studies
testing either antiinflammatory agents from Figure
4A or fluid support in animals from Figure 4B, respectively. Because preclinical trials included fewer
subjects than clinical ones,
for ease of presentation,
these confidence intervals
are adjusted for the median
number of patients in clinical trials. However, all comparisons in the
results section were based on actual numbers of animals or patients in
each trial.

ducted at low risks of death produced the same results seen in


septic patients (Figure 2B). A small number of published animal
studies conducted at low control mortality rates (Figure 1) (17,
18) demonstrated that antiinflammatory agents in sepsis have
the potential to be harmful in less severely ill subjects. This
effect is consistent with the adverse outcome seen after P-80
TNF-soluble receptor administration in a subgroup of patients
with gram-positive infection that had a low control mortality
rate (13%) (48) (Figure 4C).
Two clinical trials provided data that categorized patients
into groups based on their predicted risk of death (50, 53, 66).
These trials allowed us to evaluate the effects of two different
agents over a range of control mortality rates similar to the

1201

animal studies. Just as in animal studies, these agents were most


beneficial in those groups of patients with high risks of death
and were harmful in those with low risks (Figure 3A). The other
20 sepsis trials (3949, 51, 52, 5460), studied over a more limited
range of control mortality rates, showed a very similar relationship between risk of death and treatment effects (Figure 3B).
We believe that this consistency among preclinical studies from
different laboratories and clinical trials strongly supports our
hypothesis that risk of death alters the effects of antiinflammatory agents during sepsis.
Results of a phase III sepsis trial of activated protein C (APC),
an agent with antiinflammatory and antithrombotic properties,
could be an exception to our findings because of a highly significant (p 0.005) beneficial effect. Therefore, this agent deserves
comparison to these clinical trials even though published after
completion of this metaregression analysis (Figure 5A) (85, 86).
Although APC significantly improved survival rates compared
with standard sepsis therapies, its treatment effect was not significantly different compared with the combined effects of all
these previously studied mediator-specific antiinflammatory
agents (n 22) (p 0.10) (Figure 5A) or the phase III trials
of these agents (n 9) (odds ratio [95% confidence interval];
1.36 [1.10, 1.68] versus 1.13 [1.03, 1.24], respectively, p 0.20)
(Table 1) (3960). Furthermore, a prospective analysis divided
the 1,690 patients in the clinical APC trial into four equal groups
based on Acute Physiology and Chronic Health Evaluation II
scores (8587). Similar to clinical trials of the other antiinflammatory agents, APC was less beneficial (odds ratio of survival
1.56, 1.82, 1.19, and 0.77, respectively) as control mortality rates
(49, 36, 26, and 12%, respectively) and Acute Physiology and
Chronic Health Evaluation scores decreased (5230, 2925, 24
20, 193, respectively) (Figure 5B). Accordingly, APC was licensed for use by the Food and Drug Administration for only
patients with severe sepsis (sepsis associated with acute organ
dysfunction) and a high risk of death (e.g., as determined by
Acute Physiology and Chronic Health Evaluation II scores) (87).
Because of the consistency of the APC data to our findings
(Figure 5) examining other antiinflammatory agents, it must be
considered that in patients with a low risk of death, APC may
not only be ineffective but also harmful.
Even though all of these agents may work through different
biologic pathways, we found that the slopes for the relationship
between risk of death and treatment efficacy were similar (0.32
to 0.48). One possible explanation for this consistency is that

Figure 3. (AC) In one phase III trial of interleukin-1 receptor antagonist and one of P-55 TNF-soluble receptors, septic patients were
categorized at study entry into groups based on their predicted
risks of death, as determined by a modified Acute Physiology and
Chronic Health Evaluation II or Stratified Acute Physiology score,
respectively (open diamonds) (50, 53, 66). Using the same format
as Figure 1, A shows the observed control odds and odds ratio of
survival with antiinflammatory treatment for these risk categories
as well as the weighted regression line for the relationship. B shows
the relationship between the overall control odds and odds ratio
of survival for the remaining 20 trials of antiinflammatory agents
(3949, 51, 52, 5460) that did not provide segregated data like
that shown in A. C divides the data from A by type of antiinflammatory agent. The size of the diamonds in each panel is adjusted to
the number of patients in each subgroup (A and C ) or trial (B )
because a weighted regression analysis was done.

1202

AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 166 2002

Figure 4. (AC) This format is similar to Figure 1


except that here each circle represents an experiment in which we prospectively challenged animals
with a dose of bacterial challenge designed to produce a range of mortality rates and then treated
them with an antiinflammatory agent or placebo
(A ). In B, the format is similar to A except now
each circle represents an experiment in which we
prospectively challenged animals with a differing
dose of bacteria and then either did or did not treat
with fluids. C divides the data in A by type of antiinflammatory agent used as well as by the type and
route of bacterial challenge.

regardless of the type of immunomodulator, when the overwhelming majority of subjects are going to die from sepsis, inhibiting an excessive and lethal inflammatory response improves
outcome. Conversely, if the majority of subjects with sepsis are
likely to survive, disrupting an inflammatory response that is
working well is harmful. In this case, an immunosuppressive
effect may disrupt well-regulated inflammatory pathways and
weaken protective host defense mechanisms. When a mixture
of subjects either destined to survive or die from sepsis are
treated, harm caused by inhibiting inflammation in subjects that
would have otherwise survived may negate the potential benefit
of inhibiting inflammation in subjects that would have died. This
would explain the smaller treatment effects evident with all of

these agents in our analysis when control mortality rates were


in the mid-range.
Our model also has a term predicting the treatment efficacy
of an agent at a 50% control mortality rate. If this value had
actually been very high for the agents we analyzed, then the
slopes for the regression lines reported might have been of little
consequence. In this case, although the efficacy of antiinflammatory agents might decrease as risk of death decreased, it would
still be beneficial throughout the entire range of control mortality
rates. However, in most of our analyses, these terms were relatively small. With such small treatment effects at a 50% control
mortality rate (odds ratio, 1.17 to 1.48), any significant change in
efficacy as control mortality rate decreases becomes worrisome

Figure 5. (A and B ) This figure shows the results of the clinical


(85) and cited preclinical (86) sepsis trial of APC (open diamond
and circle in A ) as well as the effects of APC in patients categorized
based on their predicted risk of death (87) (open diamonds in
B ). For comparison, in A, data are included from Figure 1 (gray
diamonds and circles) and in B from Figure 3 (gray diamonds).

Eichacker, Parent, Kalil, et al.: Risk Alters Antiinflammatory Therapies

because at low risks of death (i.e., less than a 50% control


mortality rate) these agents become harmful. It would be ideal
to find an antiinflammatory agent for sepsis that functions as
fluids did, with a relatively large treatment effect at a 50% control
mortality and a relatively small slope. Such an agent would be
beneficial over the full range of control mortality rates.
For a highly lethal disease such as sepsis, use of preclinical
studies conducted over a wide range of risk may provide a transformative method for the preclinical testing of new agents. They
permit an estimation of an agents treatment effect at a 50%
mortality rate and can demonstrate the influence risk of death
may have on the agents efficacy. Together, these data suggest
whether there is a level of risk below which an agent may produce
harm. Ideally, drugs taken into clinical testing would be those
that have significant beneficial effects over a wide range of competing risks. However, a different approach must be considered
if preclinical or later clinical testing indicates that an agent is
beneficial above, but harmful below, a particular risk of death.
Subsequent clinical trials should then incorporate entry and exclusion criteria such as risk prediction scores to ensure that
patients likely to benefit are included and that those likely to
be harmed are excluded. Examining this relationship between
risk of death and treatment effect preclinically and then using
this information to optimally design the clinical trial are likely
to reduce the size and expense of human research in highly
lethal disease and may also help minimize the risk of drugrelated fatalities.

1203

13.

14.

15.

16.

17.

18.

19.

20.

21.

References
1. Beutler B, Milsark IW, Cerami AC. Passive immunization against
cachectin/tumor necrosis factor protects mice from lethal effect of
endotoxin. Science 1985;229:869871.
2. Suitters AJ, Foulkes R, Opal SM, Palardy JE, Emtage JS, Rolfe M,
Stephens S, Morgan A, Holt AR, Chaplin LC, et al. Differential effect
of isotype on efficacy of anti-tumor necrosis factor alpha chimeric
antibodies in experimental septic shock. J Exp Med 1994;179:849856.
3. Bagby GJ, Plessala KJ, Wilson LA, Thompson JJ, Nelson S. Divergent
efficacy of antibody to tumor necrosis factor-alpha in intravascular
and peritonitis models of sepsis. J Infect Dis 1991;163:8388.
4. Mathison JC, Wolfson E, Ulevitch RJ. Participation of tumor necrosis
factor in the mediation of gram negative bacterial lipopolysaccharideinduced injury in rabbits. J Clin Invest 1988;81:19251937.
5. Fiedler VB, Loof I, Sander E, Voehringer V, Galanos C, Fournel MA.
Monoclonal antibody to tumor necrosis factor-alpha prevents lethal
endotoxin sepsis in adult rhesus monkeys. J Lab Clin Med 1992;120:
574588.
6. Opal SM, Cross AS, Sadoff JC, Collins HH, Kelly NM, Victor GH,
Palardy JE, Bodmer WM. Efficacy of antilipopolysaccharide and antitumor necrosis factor monoclonal antibodies in a neutropenic rat
model of Pseudomonas sepsis. J Clin Invest 1991;88:885890.
7. Stack AM, Saladino RA, Thompson C, Sattler F, Weiner DL, Parsonnet
J, Nariuchi H, Siber GR, Fleisher GR. Failure of prophylactic and
therapeutic use of a murine anti-tumor necrosis factor monoclonal
antibody in Escherichia coli sepsis in the rabbit. Crit Care Med 1995;
23:15121518.
8. Tracey KJ, Fong Y, Hesse DG, Manogue KR, Lee AT, Kuo GC, Lowry
SF, Cerami A. Anti-cachectin/TNF monoclonal antibodies prevent
septic shock during lethal bacteraemia. Nature 1987;330:662664.
9. Cross AS, Opal SM, Palardy JE, Bodmer MW, Sadoff JC. The efficacy
of combination immunotherapy in experimental Pseudomonas sepsis.
J Infect Dis 1993;167:112118.
10. Opal SM, Cross AS, Kelly NM, Sadoff JC, Bodmer MW, Palardy JE,
Victor GH. Efficacy of a monoclonal antibody directed against tumor
necrosis factor in protecting neutropenic rats from lethal infection
with Pseudomonas aeruginosa. J Infect Dis 1990;161:11481152.
11. Hinshaw LB, Emerson TE Jr, Taylor FB Jr, Chang AC, Duerr M, Peer
GT, Flournoy DJ, White GL, Kosanke SD, Murray CK, et al. Lethal
Staphylococcus aureus-induced shock in primates: prevention of death
with anti-TNF antibody. J Trauma 1992;33:568573.
12. Hinshaw LB, Tekamp-Olson P, Chang AC, Lee PA, Taylor FB Jr, Murray
CK, Peer GT, Emerson TE Jr, Passey RB, Kuo GC. Survival of pri-

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

mates in LD100 septic shock following therapy with antibody to tumor


necrosis factor (TNF alpha). Circ Shock 1990;30:279292.
Jesmok G, Lindsey C, Duerr M, Fournel M, Emerson T Jr. Efficacy of
monoclonal antibody against human recombinant tumor necrosis factor in E. coli-challenged swine. Am J Pathol 1992;141:11971207.
Silva AT, Bayston KF, Cohen J. Prophylactic and therapeutic effects of
a monoclonal antibody to tumor necrosis factor-alpha in experimental
gram-negative shock. J Infect Dis 1990;162:421427.
Eskandari MK, Bolgos G, Miller C, Nguyen DT, DeForge LE, Remick
DG. Anti-tumor necrosis factor antibody therapy fails to prevent lethality after cecal ligation and puncture or endotoxemia. J Immunol
1992;148:27242730.
Jin H, Yang R, Marsters SA, Bunting SA, Wurm FM, Chamow SM,
Ashkenazi A. Protection against rat endotoxic shock by p55 tumor
necrosis factor (TNF) receptor immunoadhesion: comparison with
anti-TNF monoclonal antibody. J Infect Dis 1994;170:13231326.
Echtenacher B, Falk W, Mannel DN, Krammer PH. Requirement of
endogenous tumor necrosis factor/cachectin for recovery from experimental peritonitis. J Immunol 1990;145:37623766.
Mastroeni P, Arena A, Costa GB, Liberto MC, Bonina L, Hormaeche
CE. Serum TNF alpha in mouse typhoid and enhancement of a Salmonella infection by anti-TNF alpha antibodies. Microb Pathog 1991;11:
3338.
Ashkenazi A, Marsters SA, Capon DJ, Chamow SM, Figari IS, Pennica D,
Goeddel DV, Palladino MA, Smith DH. Protection against endotoxic
shock by a tumor necrosis factor receptor immunoadhesion. Proc Natl
Acad Sci USA 1991;88:1053510539.
Van Zee KJ, Kohno T, Fischer E, Rock CS, Moldawer LL, Lowry SF.
Tumor necrosis factor soluble receptors circulate during experimental
and clinical inflammation and can protect against excessive tumor
necrosis factor alpha in vitro and in vivo. Proc Natl Acad Sci USA
1992;89:48454849.
Lesslauer W, Tabuchi H, Gentz R, Brockhaus M, Schlaeger EJ, Grau
G, Piguet PF, Pointaire P, Vassalli P, Loetscher H. Recombinant
soluble tumor necrosis factor receptor proteins protect mice from
lipopolysaccharide-induced lethality. Eur J Immunol 1991;21:2883
2886.
Russell DA, Tucker KK, Chinookoswong N, Thompson RC, Kohno T.
Combined inhibition of interleukin-1 and tumor necrosis factor in
rodent endotoxemia: improved survival and organ function. J Infect
Dis 1995;171:15281538.
Mohler KM, Torrance DS, Smith CA, Goodwin RG, Stremler KE, Fung
VP, Madani H, Widmer MB. Soluble tumor necrosis factor (TNF)
receptors are effective therapeutic agents in lethal endotoxemia and
function simultaneously as both TNF carriers and TNF antagonists. J
Immunol 1993;151:15481561.
Evans TJ, Moyes D, Carpenter A, Martin R, Loetscher H, Lesslauer W,
Cohen J. Protective effect of 55- but not 75-kD soluble tumor necrosis
factor receptor-immunoglobulin G fusion proteins in an animal model
of gram-negative sepsis. J Exp Med 1994;180:21732179.
Ohlsson K, Bjork P, Bergenfeldt M, Hageman R, Thompson RC.
Interleukin-1 receptor antagonist reduces mortality from endotoxin
shock. Nature 1990;348:550552.
Wakabayashi G, Gelfand JA, Burke JF, Thompson RC, Dinarello CA.
A specific receptor antagonist for interleukin 1 prevents Escherichia
coli-induced shock in rabbits. FASEB J 1991;5:338343.
Fischer E, Marano MA, Van Zee KJ, Rock CS, Hawes AS, Thompson
WA, DeForge L, Kenney JS, Remick DG, Bloedow DC, et al.
Interleukin-1 receptor blockade improves survival and hemodynamic
performance in Escherichia coli septic shock, but fails to alter host
responses to sublethal endotoxemia. J Clin Invest 1992;89:15511557.
Myers AK, Robey JW, Price RM. Relationships between tumour necrosis
factor, eicosanoids and platelet-activating factor as mediators of endotoxin-induced shock in mice. Br J Pharmacol 1990;99:499502.
Chang SW, Feddersen CO, Henson PM, Voelkel NF. Platelet-activating
factor mediates hemodynamic changes and lung injury in endotoxintreated rats. J Clin Invest 1987;79:14981509.
Etienne A, Hecquet F, Guilmard C, Soulard C, Braquet P. Inhibition of
rat endotoxin-induced lethality by BN 52021 and BN 52063, compounds with PAF antagonistic effect and protease-inhibitory activity.
Int J Tissue React 1987;9:1926.
Rabinovici R, Yue TL, Farhat M, Smith EF III, Esser KM, Slivjak M,
Feuerstein G. Platelet activating factor (PAF) and tumor necrosis
factor-alpha (TNF alpha) interactions in endotoxemic shock: studies
with BN 50739, a novel PAF antagonist. J Pharmacol Exp Ther 1990;
255:256263.
Feuerstein G, Leader P, Siren AL, Braquet P. Protective effect of a PAF-

1204

33.

34.

35.
36.

37.
38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 166 2002

acether antagonist, BN 52021, in trichothecene toxicosis. Toxicol Lett


1987;38:271274.
Whalley ET, Solomon JA, Modafferi DM, Bonham KA, Cheronis JC.
CP-0127, a novel potent bradykinin antagonist, increases survival in
rat and rabbit models of endotoxin shock. Agents Actions Suppl 1992;
38:413420.
Fletcher JR, Ramwell PW. Indomethacin improves survival after endotoxin in baboons. Adv Prostaglandin Thromboxane Res 1980;7:821
828.
Parratt JR, Sturgess RM. E. coli endotoxin shock in the cat: treatment
with indomethacin. Br J Pharmacol 1975;53:485488.
Wise WC, Cook JA, Eller T, Halushka PV. Ibuprofen improves survival
from endotoxic shock in the rat. J Pharmacol Exp Ther 1980;215:160
164.
Jacobs ER, Soulsby ME, Bone RC, Wilson FJ Jr, Hiller FC. Ibuprofen
in canine endotoxin shock. J Clin Invest 1982;70:536541.
Fletcher JR, Ramwell PW. Modification, by aspirin and indomethacin,
of the haemodynamic and prostaglandin releasing effects of E. coli
endotoxin in the dog. Br J Pharmacol 1977;61:175181.
Kay CA. Reducing mortality to patients and suppliers. Paper presented
at: Cambridge Health Institutes Designing Better Drugs and Clinical
Trials for Sepsis/SIRS; 1996; Washington, DC.
Dhainaut JF, Vincent JL, Richard C, Lejeune P, Martin C, Fierobe L,
Stephens S, Ney UM, Sopwith M. CDP571, a humanized antibody to
human tumor necrosis factor-alpha: safety, pharmacokinetics, immune
response, and influence of the antibody on cytokine concentrations in
patients with septic shock: CPD571 Sepsis Study Group. Crit Care
Med 1995;23:14611469.
Fisher CJ Jr, Opal SM, Dhainaut JF, Stephens S, Zimmerman JL, Nightingale P, Harris SJ, Schein RM, Panacek EA, Vincent JL, et al. Influence
of an anti-tumor necrosis factor monoclonal antibody on cytokine
levels in patients with sepsis: the CB0006 Sepsis Syndrome Study
Group. Crit Care Med 1993;21:318327.
Reinhart K, Wiegand-Lohnert C, Grimminger F, Kaul M, Withington S,
Treacher D, Eckart J, Willatts S, Bonza C, Krausch D, et al. Assessment
of the safety and efficacy of the monoclonal anti-tumor necrosis factor
antibody-fragment, MAK 195F, in patients with sepsis and septic
shock: a multicenter, randomized, placebo-controlled, dose-ranging
study. Crit Care Med 1996;24:733742.
Cohen J, Carlet J. INTERSEPT: an international, multicenter, placebocontrolled trial of monoclonal antibody to human tumor necrosis factor-alpha in patients with sepsis: International Sepsis Trial Study
Group. Crit Care Med 1996;24:14311440.
Abraham E, Wunderink R, Silverman H, Perl TM, Nasraway S, Levy
H, Bone R, Wenzel RP, Balk R, Allred R, et al. Efficacy and safety
of monoclonal antibody to human tumor necrosis factor alpha in patients with sepsis syndrome: a randomized, controlled, double-blind,
multicenter clinical trial: TNF-alpha MAb Sepsis Study Group. JAMA
1995;273:934941.
Clark MA, Plank LD, Connolly AB, Streat SJ, Hill AA, Gupta R, Monk
DN, Shenkin A, Hill GL. Effect of a chimeric antibody to tumor
necrosis factor-alpha on cytokine and physiologic responses in patients
with severe sepsis: a randomized, clinical trial. Crit Care Med 1998;26:
16501659.
Reinhart K, Menges T, Gardlund B, Harm Zwaveling J, Smithes M,
Vincent JL, Tellado JM, Salgado-Remigio A, Zimlichman R, Withington S, et al. Randomized, placebo-controlled trial of the anti-tumor
necrosis factor antibody fragment afelimomabin hyperinflammatory
response during sepsis: the RAMSES Study. Crit Care Med 2001;29:
765769.
Abraham E, Anzueto A, Gutierrez G, Tessler S, San Pedro G, Wunderink
R, Dal Nogare A, Nasraway S, Berman S, Cooney R, et al. Doubleblind randomised controlled trial of monoclonal antibody to human
tumour necrosis factor in treatment of septic shock: NORASEPT II
Study Group. Lancet 1998;351:929933.
Fisher CJ Jr, Agosti JM, Opal SM, Lowry SF, Balk RA, Sadoff JC,
Abraham E, Schein RM, Benjamin E. Treatment of septic shock with
the tumor necrosis factor receptor: Fc fusion protein: the Soluble TNF
Receptor Sepsis Study Group. N Engl J Med 1996;334:16971702.
Abraham E, Glauser MP, Butler T, Garbino J, Gelmont D, Laterre PF,
Kudsk K, Bruining HA, Otto C, Tobin E, et al. p55 tumor necrosis
factor receptor fusion protein in the treatment of patients with severe
sepsis and septic shock: a randomized controlled multicenter trial: Ro
45-2081 Study Group. JAMA 1997;277:15311538.
Abraham E, Laterre P-F, Garbino J, Pingleton S, Butler T, Dugernier
T, Margolis B, Kudsk K, Zimmerli W, Anderson P, et al. Lenercept
(p55 tumor necrosis factor receptor fusion protein) in severe sepsis and

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.
62.

63.

64.

65.
66.

67.

68.

early septic shock: a randomized, double-blind, placebo-controlled,


multicenter phase III trial with 1,342 patients. Crit Care Med 2001;39:
503510.
Fisher CJ Jr, Slotman GJ, Opal SM, Pribble JP, Bone RC, Emmanuel
G, Ng D, Bloedow DC, Catalano MA. Initial evaluation of human
recombinant interleukin-1 receptor antagonist in the treatment of sepsis syndrome: a randomized, open-label, placebo-controlled multicenter trial: the IL-1RA Sepsis Syndrome Study Group. Crit Care Med
1994;22:1221.
Opal SM, Fisher CJ Jr, Dhainaut JF, Vincent JL, Brase R, Lowry SF,
Sadoff JC, Slotman GJ, Levy H, Balk RA, et al. Confirmatory
interleukin-1 receptor antagonist trial in severe sepsis: a phase III,
randomized, double-blind, placebo-controlled, multicenter trial: the
Interleukin-1 Receptor Antagonist Sepsis Investigator Group. Crit
Care Med 1997;25:11151124.
Fisher CJ Jr, Dhainaut JF, Opal SM, Pribble JP, Balk RA, Slotman GJ,
Iberti TJ, Rackow EC, Shapiro MJ, Greenman RL, et al. Recombinant
human interleukin 1 receptor antagonist in the treatment of patients
with sepsis syndrome: results from a randomized, double-blind, placebo-controlled trial: Phase III rhIL-1ra Sepsis Syndrome Study
Group. JAMA 1994;271:18361843.
Dhainaut JF, Tenaillon A, Le Tulzo Y, Schlemmer B, Solet JP, Wolff
M, Holzapfel L, Zeni F, Dreyfuss D, Mira JP, et al. Platelet-activating
factor receptor antagonist BN 52021 in the treatment of severe sepsis:
a randomized, double-blind, placebo-controlled, multicenter clinical
trial: BN 52021 Sepsis Study Group. Crit Care Med 1994;22:17201728.
Dhainaut JF, Tenaillon A, Hemmer M, Damas P, Le Tulzo Y, Radermacher P, Schaller MD, Sollet JP, Wolff M, Holzapfel L, et al. Confirmatory platelet-activating factor receptor antagonist trial in patients
with severe gram-negative bacterial sepsis: a phase III, randomized,
double-blind, placebo-controlled, multicenter trial: BN 52021 Sepsis
Investigator Group. Crit Care Med 1998;26:19631971.
Bernard GR, Reines HD, Halushka PV, Higgins SB, Metz CA, Swindell
BB, Wright PE, Watts FL, Vrbanac JJ. Prostacyclin and thromboxane
A2 formation is increased in human sepsis syndrome: effects of
cyclooxygenase inhibition. Am Rev Respir Dis 1991;144:10951101.
Haupt MT, Jastremski MS, Clemmer TP, Metz CA, Goris GB. Effect
of ibuprofen in patients with severe sepsis: a randomized, doubleblind, multicenter study: the Ibuprofen Study Group. Crit Care Med
1991;19:13391347.
Bernard GR, Wheeler AP, Russell JA, Schein R, Summer WR, Steinberg
KP, Fulkerson WJ, Wright PE, Christman BW, Dupont WD, et al.
The effects of ibuprofen on the physiology and survival of patients
with sepsis: the Ibuprofen in Sepsis Study Group. N Engl J Med 1995;
336:912918.
Rodell TC, Scharschmidt LA, Knaus WA. CP-0127 SIRS and Sepsis
Study Group: results of a multi-center randomized, placebo-controlled
trial of CP-0127, a novel bradykinin antagonist, in patients with SIRS
and sepsis. Shock 1995;3:60.
Fein AM, Bernard GR, Criner GJ, Fletcher EC, Good JT Jr, Knaus WA,
Levy H, Matuschak GM, Shaines HM, Taylor RW, et al. Treatment of
severe systemic inflammatory response syndrome and sepsis with a
novel bradykinin antagonist, deltibant (CP-0127): results of a randomized, double-blind, placebo-controlled trial. CP-0127 SIRS and Sepsis
Study Group. JAMA 1997;277:482487.
Stephenson J. Reflecting and regrouping after failed trials, sepsis researchers forge on. JAMA 1996;275:823824.
Piper RD, Cook DJ, Bone RC, Sibbald WJ. Introducing critical appraisal
to studies of animal models investigating novel therapies in sepsis.
Crit Care Med 1996;24:20592070.
Heumann D, Glauser MP. Anticytokine strategies for the treatment of
septic shock: relevance of animal models. Curr Top Microbiol Immunol 1996;216:299311.
From the bench to the bedside: the future of sepsis research: executive
summary of an American College of Chest Physicians, National Institute of Allergy and Infectious Disease, and National Heart, Lung, and
Blood Institute Workshop. Chest 1997;111:744753.
Opal SM, Cross AS. Clinical trials for severe sepsis: past failures and
future hopes. Infect Dis Clin North Am 1999;13:285297.
Knaus WA, Harrell FE, LaBrecque JF, Wagner DP, Pribble JP, Draper
EA, Fisher CJ Jr, Soll L. Use of predicted risk of mortality to evaluate
the efficacy of anticytokine therapy in sepsis: the rhIL-1ra Phase III
Sepsis Syndrome Study Group. Crit Care Med 1996;24:4656.
Schmid CH, Lau J, McIntosh MW, Cappelleri JC. An empirical study
of the effect of the control rates a predictor of treatment efficacy in
meta-analysis of clinical trials. Stat Med 1998;17:19231942.
Boissel JP, Collet JP, Lievre M, Girard P. An effect model for the

Eichacker, Parent, Kalil, et al.: Risk Alters Antiinflammatory Therapies

69.

70.

71.

72.

73.
74.

75.

76.

77.

78.

assessment of drug benefit: example of antiarrhythmic drugs in postmyocardial infarction patients. J Cardiovasc Pharmacol 1993;22:356
363.
Antman EM, Seelig MS, Fleischmann K, Lau J, Kuntz K, Berkey CS,
McIntosh WM. Magnesium in acute myocardial infarction: scientific,
statistical, and economic rationale for its use. Cardiovasc Drugs Ther
1996;10:297301.
Smith GD, Song F, Sheldon TA. Cholesterol lowering and mortality: the
importance of considering initial level of risk. BMJ 1993;306:1367
1373.
Brand R, Kragt H. Importance of trends in the interpretation of an overall
odds ratio in the meta-analysis of clinical trials. Stat Med 1992;11:2077
2082.
Zeni F, Freeman B, Natanson C. Anti-inflammatory therapies to treat
sepsis and septic shock: a reassessment. Crit Care Med 1997;25:1095
1100.
Cox DR, Snell EJ. Analysis of binary data. London: New York: Chapman
and Hall, CRC Press; 1989.
Breslow NE, Day NE. Statistical methods in cancer research, volume II:
the design and analysis of cohort studies. New York: Oxford University
Press; 1994.
Yaish P, Gazit A, Gilon C, Levitzki A. Blocking of EGF-dependent cell
proliferation by EGF receptor kinase inhibitors. Science 1988;242:933
935.
Glaser KB, Asmis R, Dennis EA. Bacterial lipopolysaccharide priming
of P388D1 macrophage-like cells for enhanced arachidonic acid metabolism. Platelet-activating factor receptor activation and regulation of
phospholipase A2. J Biol Chem 1990;265:86588664.
Dong Z, OBrian CA, Fidler IJ. Activation of tumoricidal properties in
macrophages by lipopolysaccharide requires protein-tyrosine kinase
activity. J Leukoc Biol 1993;53:5360.
Guy GR, Chua SP, Wong NS, Ng SB, Tan YH. Interleukin 1 and tumor

1205

79.

80.

81.

82.

83.

84.
85.

86.

87.

necrosis factor activate common multiple protein kinases in human


fibroblasts. J Biol Chem 1991;266:1434314352.
Novogrodsky A, Vanichkin A, Patya M, Gazit A, Osherov N, Levitzki
A. Prevention of lipopolysaccharide-induced lethal toxicity by tyrosine
kinase inhibitors. Science 1994;264:13191322.
Vanichkin A, Patya M, Gazit A, Levitzki A, Novogrodsky A. Late administration of a lipophilic tyrosine kinase inhibitor prevents lipopolysaccharide and Escherichia coli-induced lethal toxicity. J Infect Dis 1996;
173:927933.
Sevransky JE, Shaked G, Novogrodsky A, Levitzki A, Gazit A, Hoffman
A, Elin RJ, Quezado ZM, Freeman BD, Eichacker PQ, et al. Tyrphostin AG 556 improves survival and reduces multiorgan failure in canine
Escherichia coli peritonitis. J Clin Invest 1997;99:19661973.
Natanson C, Danner RL, Reilly JM, Doerfler ML, Hoffman WD, Akin
GL, Hosseini JM, Banks SM, Elin RJ, MacVittie TJ, et al. Antibiotics
versus cardiovascular support in a canine model of human septic shock.
Am J Physiol 1990;259:H1440H1447.
Freeman BD, Correa R, Karzai W, Natanson C, Patterson M, Banks S,
Fitz Y, Danner RL, Wilson L, Eichacker PQ. Controlled trials of rGCSF and CD11b-directed MAb during hyperoxia and E. coli pneumonia in rats. J Appl Physiol 1996;80:20662076.
Hedges LV, Olkin I. Statistical methods for meta-analysis. Orlando:
Academic Press; 1985.
Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, LopezRodriguez A, Steingrub JS, Garber GE, Helterbrand JD, Ely EW, et
al. Efficacy and safety of recombinant human activated protein C for
severe sepsis. N Engl J Med 2001;344:699709.
Taylor FB, Chang A, Esmon CT, DAngelo A, Vigano-DAngelo S,
Blick KE. Protein C prevents the coagulopathic and lethal effects of
Escherichia coli in the baboon. J Clin Invest 1987;79:918925.
Anonymous. FDA Briefing document: anti infective advisory committee
drotrecogin alfa (activated) [recombinant human activated protein C
(rhAPC)]. http://www.fda.gov/ohrms/dockets/ac/01/briefing/3797b1_02_
FDAbriefing.doc 1 A.D.

Das könnte Ihnen auch gefallen