Sie sind auf Seite 1von 12

Toxicologic Pathology, 37: 617-628, 2009

Copyright # 2009 by The Author(s)


ISSN: 0192-6233 print / 1533-1601 online
DOI: 10.1177/0192623309339502

Qualification of Cardiac Troponin I Concentration in Mouse


Serum Using Isoproterenol and Implementation in Pharmacology
Studies to Accelerate Drug Development
STEVEN K. ENGLE,1 WILLIAM H. JORDAN,1 MICHAEL L. PRITT,1 ALAN Y. CHIANG,1 MYRTLE A. DAVIS,1,2
JOHN L. ZIMMERMANN,1,3 DANIEL G. RUDMANN,1 KATHLEEN M. HEINZ-TAHENY,1 ARMANDO R. IRIZARRY,1 YUMI YAMAMOTO,1
DAVID MENDEL,1 A. ERIC SCHULTZE,1 PAUL D. CORNWELL,1 AND DAVID E. WATSON1
1
Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, Indiana, 46285, USA
2
National Cancer Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
3
Vet Path Services, Inc., Mason, Ohio, 45040, USA
ABSTRACT

Cardiac troponin I is a useful biomarker of myocardial injury, but its use in mice and application to early drug discovery are not well described. The
authors investigated the relationship between cTnI concentration in serum and histologic lesions in heart tissue from mice treated with isoproterenol
(ISO). Cardiac TnI concentrations in serum increased in a dose-dependant manner and remained increased twenty-four to forty-eight hours after a single
administration of isoproterenol. Increased cTnI concentration was of greater magnitude and longer duration than increased fatty acid binding protein 3
concentration, aspartate aminotransferase activity, and creatine kinase activity in serum. Isoproterenol-induced increases in cTnI concentrations were
both greater and more sustained in BALB/c than in CD1 mice and correlated with incidence and severity of lesions observed in heart sections from both
strains. In drug development studies in BALB/c mice with novel kinase inhibitors, cTnI concentration was a reliable stand-alone biomarker of cardiac
injury and was used in combination with measurements of in vivo target inhibition to demonstrate an off-target contribution to cardiotoxicity. Additional
attributes, including low cost and rapid turnaround time, made cTnI concentration in serum invaluable for detecting cardiotoxicity, exploring structure–
activity relationships, and prioritizing development of compounds with improved safety profiles early in drug discovery.

Keywords: troponin; biomarker; cardiac; necrosis; drug development; mouse.

INTRODUCTION compared to measurement of a serum-based biomarker. Electron


microscopy, while more sensitive than histology, is even more
Cardiac toxicity is an unintended and undesirable consequence
time-consuming and expensive. In contrast, analysis for serum-
of exposure to many drugs, including medications used to treat
based biomarkers is cost-effective and rapid. A sufficiently sen-
cancer, retroviral infection, diabetes mellitus, chronic obstructive
sitive and specific biomarker can be used as a stand-alone safety
pulmonary disease, inflammation, and psychoses (O’Brien
endpoint to prioritize compounds at early stages in drug develop-
2006). Preclinical safety assessment in animals has demonstrated
ment. Unfortunately, traditional indicators of cardiac injury, such
high predictivity of human cardiac toxicities, with up to 80%
as aspartate aminotransferase (AST) and creatine kinase (CK)
concordance between preclinical and clinical studies (Olson
activities in serum, have limited sensitivity and lack specificity
et al. 2000). Histological observation of heart lesions provides
for cardiac injury.
indisputable evidence of cardiac injury in drug development and
Cardiac troponin I is a specific, sensitive, and quantitative
preclinical studies and is routinely utilized during safety assess-
measure of cardiac injury in humans (Adams et al. 1993;
ment. However, it is not commonly employed during pharmacol-
Babuin and Jaffe 2005) and veterinary species (O’Brien et al.
ogy studies because it is relatively time-consuming and expensive
2006). Concentrations in serum are increased as a result of
direct myocardial injury, myocardial ischemia, ventricular
Address correspondence to: Steven K. Engle or David E. Watson, Lilly strain caused by disease (Fromm 2007), or drug-induced toxi-
Corporate Center, Indianapolis, IN 46285, USA; phone: 317-655-2121 or city (Babuin and Jaffe 2005; Adamcova et al. 2007). Cardiac
317-433-2155; e-mail: Englesk@lilly.com or Davewatson@lilly.com. TnI has thus become the biomarker of choice for monitoring
Financial Disclosure: The studies described in this article were funded by cardiac injury in humans (Collinson and Gaze 2007).
Eli Lilly and Company. The authors have not declared any other conflicting
interests.
Cardiac TnI is a sarcomeric protein in the troponin complex
Abbreviations: AST, aspartate aminotransferase (also known as serum of cardiomyocytes. The complex consists of troponins I (cTnI),
glutamic oxaloacetic transaminase); CK, creatine kinase; CMC, the inhibitory subunit; T (cTnT), the tropomyosin binding
carboxymethylcellulose; cTnC, cardiac troponin calcium binding subunit; subunit; and C (cTnC), the calcium binding subunit. Upon
cTnI, cardiac troponin inhibitory subunit (Tnni3); cTnT, cardiac troponin membrane disruption, intracellular cTnI leaks into the extra-
tropomyosin binding subunit (Tnnt2); CV, coefficient of variation; Fabp3,
fatty acid binding protein 3; GenBank, Protein symbols shown in
cellular environment as a monomer, part of a binary complex
parentheses.; ISO, isoproterenol; IVTI, in vivo target inhibition; LLQ, lower with cTnC, or in a ternary complex of cTnT-cTnI-cTnC
limit of quantitation; sc, subcutaneous; ULQ, upper limit of quantitation. (Wu et al. 1998). Cardiac troponin I is distinguished from

617
618 ENGLE ET AL. TOXICOLOGIC PATHOLOGY

skeletal muscle isoforms of TnI by a cardiac-specific N- METHODS


terminal sequence (Larue et al. 1992), which is the basis for its
Animals
diagnostic specificity for myocardial damage.
Release of cTnI from myocardium was proportional to the Animal protocols were approved by Eli Lilly and Com-
size and extent of tissue injury in several animal models of car- pany’s Institutional Animal Care and Use Committee. Female
diotoxicity (O’Brien et al. 2006) and has been reported to cor- CD-1 and BALB/c mice were obtained at nine to eleven weeks
relate well with systolic function (Adamcova et al. 2007). of age from Charles River Laboratories (Wilmington, MA) or
Challenges remain with regard to preclinical applications, Harlan Sprague Dawley, Inc. (Indianapolis, IN). Female mice
including assessment of a suitable cutoff for irreversible myo- were chosen for consistency with pharmacology studies in drug
cardial injury, optimal timing of sample collection (Adamcova development, in which female BALB/c mice are used. Mice
et al. 2005), relatively limited data sets for animals, and lack of were housed singly with access to food and water ad libitum
standardization between various immunoassays available to and were allowed to acclimate for at least one week to caging,
measure cTnI concentration. Important differences exist in feeding, and watering conditions before any handling. Blood
species-specific cross-reactivity of the antibodies used in the was collected under isoflurane anesthesia via retro-orbital sinus
assays available for the measurement of cTnI concentrations or abdominal vena cava. Mice were humanely euthanized
in laboratory rats, dogs, and monkeys, as well as the precision either by exsanguination under anesthesia followed by organ
and accuracy of the measurements (Apple et al. 2008). There is removal or by carbon dioxide inhalation followed by cervical
little information available regarding the use of these same dislocation.
assays in mice. These challenges contribute to the limited use
of cTnI in toxicology studies, even those involving compounds
with previously identified cardiotoxic potential. Furthermore, Isoproterenol Studies
strains of mice used in discovery biology studies often differ
ISO (Sigma-Aldrich, St. Louis, MO) was dissolved in 0.9%
from those used in toxicology studies, contributing to uncer-
sodium chloride and administered in a single, sc administration
tainty in the interpretation of toxicology measurements in
at 0.1, 1, 10, or 50 mg/kg to BALB/c and CD-1 mice. Control
‘‘nonstandard’’ strains of rodent.
mice were treated with 0.9% sodium chloride. Dose volume
To extend the application of cTnI as a biomarker of cardiac
was 5 ml/kg. In a series of seven studies, blood was collected
injury in preclinical species, we qualified cTnI concentration in
from anesthetized mice via abdominal vena cava 1, 4, 8, 24,
serum as a biomarker of cardiotoxicity in CD-1 and BALB/c
48, or 72 hours after injection and processed for serum. Hearts
mice treated with a nonspecific b-adrenergic agonist, isoproter-
were collected for histopathologic examination 24, 48, or 72
enol (ISO), a well-described model of cardiac necrosis (Zhang
hours after injection.
et al. 2008). Mice, because they require relatively small
amounts of compound, are a preferred species for in vivo
assessment of target inhibition and efficacy. In these studies,
Histopathologic Examinations in Isoproterenol Studies
we explored the kinetics of alterations in cTnI concentrations
in dose response and time course studies, as well as the rela- Hearts were fixed for histological examination in 10% neu-
tionship of cTnI concentrations to fatty acid binding protein tral buffered formalin, bisected longitudinally through both
3 (Fabp3) concentrations, CK and AST activities, and severity ventricles, processed, embedded (both halves) in paraffin, sec-
of myocardial injury detected by histopathological examina- tioned at a thickness of 5 mm, and stained with hematoxylin and
tion. Fabp3, also known as heart-type fatty acid binding pro- eosin. Tissue sections were examined by light microscopy and
tein, is a marker of tissue injury (Pelsers, Hermens, and Glatz evaluated for necrosis and inflammation (cellular infiltration,
2005) with reported utility as a biomarker of cardiac necrosis primarily mononuclear). Lesions were scored as follows: 0 ¼
(Ruzgar et al. 2006), but its use has not been described in mice. normal, 1 ¼ minimal (very few, small foci affected, approx.
CD-1 and BALB/c mice were chosen because of their common 2 to 5 myofibers per histological section), 2 ¼ slight (identifi-
use in toxicology and pharmacology studies, respectively. The able at low magnification but estimated to involve < 8% of tis-
comparison of increased cTnI concentration in serum from sue), 3 ¼ moderate (easily identifiable, 8% to 20% of tissue
both strains with histology provides assurance that the results affected), 4 ¼ marked (20% to 50% of tissue affected), and
of cTnI screening in BALB/c mice will be comparable to the 5 ¼ severe (>50% affected). Interpretation was based on
results of a toxicology study in CD-1 mice. changes in individuals within each group, but for presentation
Cardiac TnI concentration in serum was then deployed as a purposes, necrosis severity scores were also averaged into a
stand-alone safety marker to detect myocardial injury caused group score. Following initial evaluations to establish the prin-
by novel kinase inhibitors in early drug discovery studies using cipal changes, tissue sections were evaluated independently by
BALB/c mice. In vivo target inhibition was assessed concur- two board-certified veterinary pathologists without knowledge
rently in these studies. This strategy resulted in the identification of strain, time, or dose. The final scores represented the consen-
of unacceptably cardiotoxic compounds very early in the discov- sus of both evaluators. Photomicrographs were extracted at 8X
ery process and enabled prioritization of compounds that had a and 20X from digital images taken on a ScanScope XT (Aperio
favorable balance between target inhibition (potency) and safety. Technologies, Inc., Vista, CA) using a 20X objective.
Vol. 37, No. 5, 2009 IMPLEMENTATION OF cTnI IN MOUSE PHARMACOLOGY STUDIES 619

AST, CK, Fabp3, and cTnI administrations of a range of dose levels of each compound.
Limited blood volume in BALB/c mice precluded measurement
AST activity and total CK activity in serum were analyzed
of percentage IVTI in toxicology studies. However, plasma com-
using a Roche/Hitachi 917 automated analyzer (Indianapolis,
pound concentrations were measured at 1, 2, 4, 8, 16, and 24
IN) according to the manufacturer’s protocol. Fabp3 concentra-
hours (n ¼ 2 per time) after the first administration and used
tion in serum was measured by a mouse/rat-specific ELISA
to interpolate percentage IVTI from the previously established
(Cat# HK403 Hycult Biotechnology, Uden, the Netherlands)
relationship between plasma compound concentration and per-
as previously described (Pritt et al. 2008). Cardiac TnI concen-
centage IVTI (preceding paragraph). We then calculated per-
tration in serum was measured by a mouse-specific ELISA
centage target inhibition  hour as area under the curve and
(Cat# 2010-1-HS, Life Diagnostics, Inc., West Chester, PA).
plotted these data against group mean cTnI concentration in
Standard and sample volumes were reduced to 25 mL from the
serum to assess the relationship between target inhibition and
manufacturer’s recommended 100 mL per well to conserve lim-
cardiotoxicity.
ited sample volume. Cardiac troponin I, purified from mouse
hearts by ion-exchange chromatography in conjunction with
calcium-dependent affinity chromatography on troponin-C Screening for Improved Margin of Safety in
agarose (Cat# 7110, Life Diagnostics, Inc., West Chester, Pharmacology Studies
PA), was used to construct a standard curve after dilution in In cardiotoxicity screening studies, female BALB/c mice
naı̈ve mouse serum provided with the kit. Concentration of pur- were treated orally with 150 mg/kg of novel kinase inhibitors
ified cTnI for standards was verified by amino acid analysis as sharing a common target. Test articles were administered in a
previously described (van Wandelen and Cohen 1997). The 1% carboxymethylcellulose (Thermo Fisher Scientific, Wal-
cTnI ELISA was qualified according to published criteria tham, MA) vehicle at 10 mL/kg. Blood was drawn from
(DeSilva et al. 2003). Specifically, the quantifiable range anesthetized mice 4 hours after the second daily administration
was determined in mouse serum spiked with cTnI for which via retro-orbital sinus from 5 mice per compound, processed
total error (mean bias þ interrun covariance) was less than for serum, and assayed for cTnI concentration. Comparisons
30%. Samples from ISO studies with cTnI concentration > were made across compounds using both IVTI and cTnI data.
10 ng/mL were diluted 1:2 and reanalyzed. A subset of compounds were advanced into more extensive
toxicology studies to test the ability of cTnI measurements in
Baseline cTnI Concentration screening studies to predict cardiotoxicity. In these studies,
female BALB/c mice were treated daily with a range of doses
Sera from vehicle-treated mice were assayed for cTnI con-
for 4 days. Twenty-four hours after the fourth dose, hearts were
centration to assess baseline concentrations and what effect,
collected for histologic examination.
if any, dosing and blood collection routes had. Blood was col-
lected from anesthetized mice via retro-orbital sinus or abdom-
Measurement of cTnI and Fabp3 Concentrations in
inal vena cava. Mice received a single sc administration of
vehicle (0.9% saline) in studies with ISO or 1 to 4 daily oral Mouse Hearts
vehicle (1% carboxymethylcellulose or 10% acacia) adminis- Left ventricles were collected at necropsy from mice and
trations in drug development studies. homogenized as previously described (Pritt et al. 2008). Total
protein concentration was measured by Bradford assay (Pierce
Discrimination of On- versus Off-Target Toxicity in cat# 23236, Thermo Fisher Scientific, Rockford, IL) of each tis-
Pharmacology Studies sue supernatant sample. Cardiac TnI and Fabp3 concentrations
in supernatant were determined by ELISA (described above) and
In vivo target inhibition (IVTI) was measured 2 hours after a expressed per microgram of total protein.
single oral administration of a novel kinase inhibitor (dose
volume ¼ 10 mL/kg) at a range of dose levels in BALB/c mice
Statistical Analysis
(n ¼ 3 to 4 mice per dose). IVTI was measured as phosphoryla-
tion status of a signal transduction protein in peripheral blood Quantitative results for cTnI concentration were analyzed
mononuclear cells (PBMCs) using phospho-specific antibodies using a three-factor analysis of variance. Factors in the model
and flow cytometry procedures similar to those previously included treatment, strain, time, and their interactions. Treat-
described (Krutzik and Nolan 2003; Krutzik et al. 2004). Com- ment effect for each strain at each time point was evaluated
pound concentrations in plasma were measured using mass spec- using a t-test at the .05 significance level. The mean difference
trometry. A nonlinear, sigmoidal fit of percentage IVTI versus in cTnI concentration in strain (CD-1 vs. BALB/c mice) was
plasma compound concentrations (Prism, GraphPad Software, also tested at the .05 significance level at each time point.
Inc., La Jolla, CA) provided a quantitative relationship between Shapiro-Wilk’s test (Shapiro and Wilk 1965) for normality was
IVTI and plasma compound concentration that was used to inter- performed at the .01 significance level to detect outlying obser-
pret serum cTnI data from toxicology studies (see below). vations, and Levene’s test (Levene 1960) for homogeneity of
Cardiotoxicity was assessed using cTnI concentrations in variance was performed at the .01 significance level to assist
serum (n ¼ 3 to 12) 4 hours after the last of 4 daily, oral in interpretation of the effects.
620 ENGLE ET AL. TOXICOLOGIC PATHOLOGY

TABLE 1.—Cardiac TnI concentration in vehicle-treated mice. concentration (n ¼ 5) was 1.1 ng/mL. In CD-1 mice treated
with ISO at 0.1 or 50 mg/kg, cTnI concentration returned to
cTnI concentration (ng/mL)a
baseline by 24 hours. Cardiac TnI concentration in CD-1 mice
<0.14 0.14–0.29 0.3–1 >1 treated with ISO at 1 or 10 mg/kg returned to baseline by 48
hours postdose. Maximum concentration of cTnI occurred 1
Strain Route Number of administrations Number of mice in range
hour postdose for CD-1 mice administered ISO at 0.1, 1, or
CD-1 scb 1 34 1 —d — 50 mg/kg and 1 to 4 hours postdose at 10 mg/kg.
BALB/c sc 1 51 — 1 — In BALB/c mice, mean cTnI concentration in serum
poc 1 39 — 3 — increased in a dose-dependant manner after treatment with ISO
po 2 23 1 — —
at 0.1 to 10 mg/kg ISO during the first 24 hours after treatment
po 4 54 1 3 1
(Figure 1B). Within 24 hours of dosing, cTnI concentration was
a
Blood sampling for cTnI analysis was done 2 to 4 hours after the second greater than LLQ in 42 of 43 animals treated with ISO at 1 or
administration.
b 10 mg/kg. The lowest concentration was 0.36 ng/ml (24 hours
sc vehicle was 0.9% saline.
c
po vehicle was 1% CMC or 10% acacia. postdose), and 37 were greater than 1 ng/ml. In BALB/c mice
d
Zero observations in this range: —. treated with ISO at 0.001 and 0.01 mg/kg, cTnI was below
LLQ at both times tested (4 and 24 hr). Maximum concentra-
RESULTS tion of cTnI occurred 4 hours postdose in BALB/c mice admi-
nistered ISO at 0.1 and 10 mg/kg and 4 to 24 hours postdose
Qualification of cTnI Assay at 1 mg/kg; the lowest concentration after 1 mg/kg was
The lower limit of quantitation (LLQ) of the ELISA used 0.86 ng/ml and occurred 4 hours postdose. Cardiac TnI con-
for detection of purified mouse cTnI in mouse serum was centration in serum from BALB/c mice treated with ISO at 1
0.14 ng/mL; the upper limit of quantitation (ULQ) was  or 10 mg/kg returned to baseline by 72 hours postdose.
10 ng/mL. Spike recovery samples were measured in triplicate for BALB/c mice were not treated with ISO at 50 mg/kg.
4 to 5 runs at 10, 5, 2.5, 1.25, 0.63, 0.31, 0.16, and 0.14 ng/mL;
mean bias was 0.02, 0.27, –0.56, 0.35, 0.93, –0.23, –1.68, and Histological Observations in Hearts
6.85%; interassay variability assessed as coefficient of Isoproterenol treatment-related changes in hearts included
variation (%CV) was 4.3, 5.5, 4.0, 4.1, 6.8, 7.6, 9.0, and dose-, time-, and strain-dependent incidence and severity of
10.5, respectively. myocardial necrosis and inflammation. Aside from one section
at 48 hours (n ¼ 5) with minimal inflammation, no important
Baseline cTnI Concentration changes occurred in CD-1 mice at 0.1 mg/kg (not tabulated;
hearts were not collected from BALB/c mice given 0.1 mg/
Cardiac TnI concentration in serum was less than the LLQ kg). Changes were poorly dose-responsive at 1 and 10 mg/kg
in 94.5% of vehicle-treated mice (Table 1), and no difference in both strains (Figure 2). Evidence for active myocardial
in baseline cTnI concentration based on blood collection route necrosis and inflammation peaked at 48 and 72 hours, respec-
or vehicle was detectable. The few examples of quantifiable tively, and were generally less prominent in CD-1 than BALB/
cTnI concentration in vehicle-treated mice may have been due c mice (Figure 3A-B). Lesion severity ranged from minimal
to spontaneous myopathies or to the stress of handling. With (grade 1), represented by loss of striations (coagulative necro-
such low incidence of detectable serological cTnI concentra- sis) in a few myofibers, to moderate (grade 3), which affected
tions in vehicle-treated mice, most serum concentrations of
multiple clusters of myofibers (Figure 4; additional details of
cTnI above the LLQ in treatment groups were likely to be grading scheme in Methods section). Necrosis and inflamma-
treatment-related. tion occurred most frequently near the lumen of the left ventri-
cle, especially near the apex, but were found in any portion of
Cardiac TnI Concentration in Isoproterenol Studies the left ventricle and rarely in other portions of the heart.
Vacuolation was a minor feature and likely represented a com-
In CD-1 mice, mean cTnI concentration in serum increased
bination of a degenerative change and evidence for edema as a
in a dose-dependant manner from 0.1 mg/kg to 10 mg/kg ISO
component of inflammation. Inflammation was characterized
during the first 24 hours after treatment (Figure 1A). The max-
by increased numbers of mononuclear leukocytes, many of
imum mean cTnI concentration after treatment with 50 mg/kg
which were macrophages. Increased cellularity was likely
(5.9 ng/mL) was not significantly greater than the maximum
enhanced by early regenerative activities of cardiac myocytes.
after treatment with 10 mg/kg. Thirty-five of 39 CD-1 mice
treated with ISO at 1 or 10 mg/kg had cTnI concentrations in
Mean Fabp3 Concentration in Isoproterenol Studies
excess of LLQ in the first 24 hours after dosing. The lowest
measurable increase was 0.18 ng/mL (8 hours postdose), and Mean Fabp3 concentration in serum was measured in mice
21 were in excess of 1 ng/mL. After administration of ISO at treated with vehicle (saline) and ISO at 10 mg/kg. Fabp3 con-
1 and 10 mg/kg, maximum cTnI concentration in serum centration was below the lower limit of quantitation (1 ng/mL)
occurred 1 to 4 hours after dosing, and in both cases, the lowest in 13 out of 15 vehicle-treated CD-1 mice. Fabp3 concentration
Vol. 37, No. 5, 2009 IMPLEMENTATION OF cTnI IN MOUSE PHARMACOLOGY STUDIES 621

FIGURE 1.—Cardiac TnI concentration in serum from isoproterenol (ISO)–treated mice. Dose response and time course studies after a single sub-
cutaneous administration of vehicle (saline) or ISO at 10, 1.0, or 0.1 mg/kg in (A) CD1 and (B) BALB/c mice. Data points and bars represent mean
+ SEM (n ¼ 3–10) cTnI concentration in serum. Blood was collected via retro-orbital sinus in isoflurane anesthetized mice. *Significantly
greater than control (p  .006).

FIGURE 2.—Necrosis and inflammation in histologic sections of hearts from isoproterenol studies. Histopathologic data from heart sections from
CD1 (open triangles) and BALB/c (closed triangles) mice at 24 (A, B), 48 (C, D), and 72 (E, F) hours after a single subcutaneous administration of
ISO. Necrosis was most severe 24 to 48 hours after treatment, and inflammation was most severe 48 to 72 hours after treatment.
622 ENGLE ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 3.—Comparison of cardiac necrosis detected by histopathology and cTnI concentrations in serum. Severity of cardiac necrosis (0 ¼ normal,
1 ¼ minimal, 2 ¼ slight, 3 ¼ moderate) in histologic heart sections and cTnI concentration in serum from BALB/c (dashed lines) and CD1 (solid
lines) mice after treatment with 1 mg/kg (A, C) and 10 mg/kg (B, D) ISO. Data points represent mean + SEM (n ¼ 3–17). BALB/c significantly
different from CD1. *p ¼ .04, **p < .001.

was increased in CD-1 mice 4 hours after treatment with ISO interference. No significant alterations in the activity of AST
(p ¼ .05) and returned to control values by 8 hours postdose were measured at any time in BALB/c mice (n ¼ 3-8). Total
(n ¼ 5; Figure 5C). CK activity in ISO-treated CD-1 mice (n ¼ 4-5) was increased
In contrast, Fabp3 concentration was above LLQ in 5 out of 1 hour postdose (p  .001) and was not different from baseline
6 vehicle-treated BALB/c mice. Mean Fabp3 concentration values at 8 or 24 hours after dosing (Figure 5G). Activity of CK
was increased in BALB/c mice after treatment with ISO; max- in BALB/c mouse serum was markedly increased at 1 hour
imum concentration occurred 1 hour after dosing and was sta- postdose, suggestive of myocyte injury. At 8 and 24 hours post-
tistically significant compared to control at 1 and 4 hours after dose, individual mice had marked increases in the activity of
dosing (n ¼ 5, p < .003; Figure 5C). Eight hours after dosing, CK, but group mean was not different from baseline (n ¼ 1-5).
mean Fabp3 concentration in serum had decreased but had not
returned to baseline.
Strain Comparison for Biomarkers
AST and CK Activities in Isoproterenol Studies
Consistent with histological observations (Figure 3A-B),
AST and total CK activities, classical markers of cardiac concentrations of cTnI in serum were significantly greater in
injury, were measured in the serum of mice treated with vehicle BALB/c than CD-1 mice at 4, 24, and 48 hours and remained
or ISO at 10 mg/kg. Vehicle-treated control mice from all increased above LLQ longer in BALB/c (48 hours) than CD-1
CD-1 (n ¼ 28) and BALB/c (n ¼ 24-26) studies were used to mice (24 hours) after treatment with ISO at 1 and 10 mg/kg
establish baselines for AST and CK. Baseline AST and CK (Figure 3C-D). Increases in Fabp3 concentration compared to
activities were comparable between strains. AST activities were control groups were both greater and more sustained in
minimally increased at 1 and 8 hours after treatment with ISO at BALB/c mice than CD-1 mice (Figure 5D) but were not as
10 mg/kg (n ¼ 4-5, p  .003) and were not different from base- great or sustained as increases in cTnI concentration in either
line values at 24 hours postdose (Figure 5E) in CD1 mice. Anal- strain (Figure 5B). There was no apparent strain difference in
yses of AST and CK activities in BALB/c mice were increases of AST or CK activities, and both were of lesser mag-
compromised by hemolysis, which is known to cause positive nitude than changes in cTnI (Figure 5F, H).
Vol. 37, No. 5, 2009 IMPLEMENTATION OF cTnI IN MOUSE PHARMACOLOGY STUDIES 623

FIGURE 4.—Distribution and microscopic features of myocardial changes in isoproterenol studies. Photomicrographs of H&E stained sections of
mouse hearts including a slightly affected CD-1 mouse (A, C) and a moderately affected BALB/c mouse (B, D) 48 hours after treatment with
1 mg/kg ISO. In the left ventricle (LV), multiple foci of pallor (arrows) and increased cellularity were apparent at low (8X) magnification (A,
B). Changes were rarely found in the right ventricle (RV). Normal myocardium (NM) was interspersed with foci of coagulative necrosis (arrows)
and increased cellularity at both low and high (20X, C, D) magnification. Consistent with greater cTnI values in the BALB/c mouse, coagulative
necrosis was more severe in that strain than in the CD-1 mouse.

Cardiac TnI and Fabp3 Concentrations in Left Ventricles bridge between these studies, we measured plasma compound
of BALB/c and CD-1 Mice concentrations in both.
In the event cardiotoxicity were target-mediated, little varia-
Concentrations of cTnI and Fabp3 in supernatants from
bility should occur across compounds in plots of percentage
homogenized left ventricles of BALB/c mice (2.5 + 0.3 and
IVTI versus serum cTnI. If the cardiotoxicity had a significant
13.4 + 0.2 ng/mg total protein, n ¼ 5) were not different from
off-target component, significant variability would occur
those from CD-1 mice (2.6 + 0.5 and 14.6 + 1.6 ng/mg total
across compounds in plots of percentage IVTI versus serum
protein, n ¼ 5). Consequently, an inherent difference in tissue
cTnI. A clear difference was observed across compounds
concentration cannot account for either strain-based differ-
(Figure 6), suggesting a significant off-target component to the
ences in the response of these biomarkers in serum from the
acute cardiotoxicity caused by these kinase inhibitors and jus-
two strains or for the differences in baseline serological Fabp3
tifying further exploration of structure-activity relationships to
concentration.
retain pharmacological activity while minimizing compound-
related cardiotoxicity.
Discrimination of On- versus Off-Target Toxicity in
Pharmacology Studies Using BALB/c Mice Screening for Improved Margin of Safety in
Pharmacology Studies
To discriminate between on-target and off-target mechan-
isms of cardiotoxicity, we explored the quantitative relation- Mice treated with kinase inhibitors in screening studies dis-
ship between IVTI and serum cTnI in BALB/c mice treated played a range of cTnI concentrations in serum that were not
with kinase inhibitors sharing a common target. Due to limited consistently related to target inhibition. Compounds were
blood volume in BALB/c mice, independent studies were con- binned into high risk, moderate risk, and low risk by consider-
ducted for IVTI and toxicological response (serum cTnI). To ing both the IVTI and the serum cTnI data (Table 2). Increased
624 ENGLE ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 5.—Comparison of biomarkers of cardiac necrosis measured in serum from isoproterenol studies. CD1 (white columns) and BALB/c
(black columns) mice treated with 10 mg/kg ISO. Bars represent mean values (+ SEM) for cTnI (A), Fabp3 (C), AST (E), and CK (G) in serum,
and fold change from control for cTnI (B), Fabp3 (D), AST (F) and CK (H). *p  .05 vs. control.

cTnI concentrations in sera from screening studies were predic- DISCUSSION


tive of cardiac necrosis after extended dosing (Table 3). Com-
Biomarkers of target organ toxicity can be used as stand-
pounds that caused cTnI concentrations in excess of 1 ng/mL
alone safety endpoints in discovery biology studies during
in serum from one or more mice after 2 days caused cardiac
early drug development. In these studies, cTnI concentration
lesions observable through histopathology after 4 days of
in serum was qualified in BALB/c and CD-1 mice and then
treatment.
Vol. 37, No. 5, 2009 IMPLEMENTATION OF cTnI IN MOUSE PHARMACOLOGY STUDIES 625

TABLE 2.—Cardiotoxicity screening in BALB/c mice treated


orally with novel kinase inhibitors.

Pharmacology cTnI Concentration (ng/mL)a

Compoundb % IVTIc <0.14 0.14–0.29 0.3–1 >1 Decision


d
1 –10 5 — — —
2 18 4 1 — —
3 31 1 1 2 1
4 52 3 — 2 —
5 54 3 — 2 —
6 58 5 — — —
7 64 — — — 5
8 65 — 1 1 3
9 67 3 1 — 1
10 76 — — — 5
11e 88 — — — 4
FIGURE 6.—Target inhibition and cTnI concentration in serum from 12 90 — 1 3 1
kinase inhibitor studies. Compound concentration in plasma was mea- 13e 91 1 1 — 2
sured 2 hours after a single administration of a novel kinase inhibitor, 14 91 3 1 — 1
and cTnI in serum was measured 4 hours after the fourth daily admin- 15 93 1 3 — 1
istration. Percent in vivo target inhibition (IVTI) was calculated from a 16 93 4 — 1 —
quantitative relationship established between compound concentration 17 94 1 — 4 —
in plasma and target inhibition in peripheral blood mononuclear cells 18 94 — — — 5
in pharmacology studies. Cardiac troponin I concentration in serum 19 95 2 2 1 —
20 96 2 1 1 1
increased with target inhibition after treatment with compounds 7 and
21 99 — — — 5
27, but not with compound 2, and to a lesser extent with compounds 3
22e >100 — — 1 3
and 12. Data for these compounds also appear in Tables 2 and 3. 23 >100 5 — — —
24e >100 — — — 4
25 >100 4 — 1 —
implemented as a biomarker of acute cardiac necrosis in paral-
26 >100 — — 2 3
lel with in vivo target inhibition studies in BALB/c mice. 27f >100 — — 3 2
Benefits of such a strategy included discrimination between
Problematic (high risk—cardiac necrosis expected) ¼&. Potential for safety margin
on- and off-target mechanisms of toxicity and prioritization may exist (medium risk) ¼&. Promising (low risk—cardiac necrosis not expected) ¼
of the development of compounds with improved safety pro- &.
files. This strategy proved to be an efficient and quantitative a
Blood sampling for cTnI analysis was done 4 hours after the second administration;
method of detecting cardiotoxicity after treatment with novel values are number of mice in range.
b
Compounds (see also Figure 6) were administered once daily for cardiotoxicity
kinase inhibitors in a drug discovery program. screening at 150 mg/kg for 2 days.
Cardiac TnI concentration in serum was a sensitive and c
In vivo target inhibition (IVTI) was measured 2 hours after a single administration at
quantitative marker of acute cardiac injury induced by treat- 10 mg/kg in pharmacology studies.
d
Zero observations in this range: —; n ¼ 5 mice per compound.
ment with ISO in both BALB/c and CD-1 mice. Increased e
Sample missing due to early death.
cTnI concentration in serum was progressive with dose of ISO f
cTnI sample 2 hours after 2 administrations at 100 mg/kg.
and occurred concurrently with cardiac necrosis, although
morphological changes detectable by histopathologic exami- in the medium-risk category demonstrated a dose margin in
nation plateaued between 1 and 10 mg/kg. A threshold con- which compound administration did not cause cardiac necrosis.
centration of cTnI in serum was identified, above which an All compounds that caused cTnI concentrations in the high-risk
animal was considered to have experienced adverse cardiac category caused cardiac necrosis in 4-day studies. Given the
injury. This was considered to be the lowest cTnI concentra- lack of uniformity in the assays available to measure cTnI, the
tion in any sample at the time point during which maximum threshold concentrations are likely to vary depending on which
group mean concentration occurred. In both strains, this assay is used.
occurred from 1 to 4 hours postdose and was 0.7 ng/mL Isoproterenol-induced increases in cTnI concentrations
(CD-1) and 0.9 ng/mL (BALB/c). were both greater and more sustained in BALB/c than in
In practice, we found that cTnI concentrations in serum CD-1 mice, consistent with increased incidence and severity
from BALB/c mice after treatment with a series of novel kinase of morphological changes (greater number of muscle fibers
inhibitors could be binned into low-risk (BQL-0.29 ng/mL), involved or greater total volume of injury). The increased
medium-risk (0.3-1.0 ng/mL), and high-risk (>1.0 ng/mL) duration was likely attributable to higher initial concentra-
categories. Compounds used in 2-day screening studies from tions of cTnI in serum, resulting from more extensive injury
which all measured cTnI concentrations fell into the low-risk in BALB/c mice. Differences in cTnI concentration in serum
category were not observed to cause cardiac necrosis in at the same dose across strains could not be explained by dif-
4-day studies. Compounds that produced cTnI concentrations ferent concentrations of cytoplasmic cTnI in the left
626 ENGLE ET AL. TOXICOLOGIC PATHOLOGY

TABLE 3.—Cardiac necrosis in studies with kinase inhibitors as the more cardiac specific CK-MB, with no histological
advanced into dose-escalation studies. evidence of cardiac necrosis (Heyndrickx et al. 1985; Vatner,
Heyndrickx, and Fallon 1986).
Compounda Dose (mg/kg) Incidence of Necrosisb Approximately 2% to 4% of total cTnI exists free in the
2 50 0/12 cytosol of ventricular myocytes (Mair et al. 1996; Wu and Feng
2 150 0/12 1998). The rapid, forty- to eighty-fold increases in cTnI con-
4 100 0/3 centration in serum in the first 1 to 8 hours after ISO treatment,
4 300 0/3
4c 1000 1/1
followed by a gradual decline in concentration over the next 48
6 50 0/3 hours, suggested that cytosolic cTnI was released immediately
6 150 0/3 upon injury. Release of sarcomere-bound cTnI may have been
6 500 0/3 inhibited by the gradual process of myocyte cell death and may
7c 30 5/9 not have reached the circulation due to phagocytosis by infil-
7c 100 7/10
12 30 2/3
trating inflammatory cells. Clinical evidence of this has been
12 100 1/3 observed in patients undergoing coronary artery bypass grafts
12 150 2/3 in which cTnI concentration in serum increased in the first
15 50 0/3 20 minutes following reperfusion of the heart, while b-type
15 150 3/3 myosin heavy chain remained within reference intervals. In
15 500 3/3
25 30 0/3
excised atrial appendages from the same patients, 8% of total
25 100 0/3 cTnI content was found free in the cytosol, compared to less
25c 300 0/2 than 0.1% of b-MHC, leading to the conclusion that the imme-
a
Compounds were administered to female BALB/c mice once daily for 4 days (see
diate increase in circulating cTnI was from the free cytosolic
Table 2 for in vivo target inhibition). pool (Bleier et al. 1998).
b
Hearts were collected for histology 24 hours after the fourth dose and assessed for We also compared cTnI changes with other markers of car-
any degree of necrosis. diac injury. Increased Fabp3 concentration in serum suggested
c
Sample(s) missing due to early death.
cell membrane disruption, with considerable magnitude of
change in BALB/c mice. However, the duration of increased
ventricles. Greater cTnI concentration in serum from BALB/c Fabp3 concentration was relatively limited in comparison to
mice is likely attributable to a greater number of injured car- cTnI, and it is also found in skeletal muscle (Pritt et al.
diac muscle fibers. 2008), confounding specific detection of myocardial injury.
The cause(s) for the strain differences in response to cardiac The differences in Fabp3 concentrations between strains, in
injury induced by exposure to ISO was not determined. How- both control and treated mice, were remarkable and unex-
ever, possible causes include desensitization of b-adrenergic pected. They could not be explained by differences in Fabp3
receptors (Perrino et al. 2005), strain-dependent differences concentrations in the ventricles. The small magnitude of
in receptor density, cardiac capacity for aerobic metabolism, increases in Fabp3 in CD-1 mice were insufficient to determine
or adaptive responses (Faulx et al. 2005, 2007). Desensitization whether a difference in clearance rate of Fabp3 from blood
may also account for the lack of dose-response in CD-1 mice exists between mouse strains but in any case cannot explain the
treated with greater than 10 mg/kg ISO. large differences in serum concentrations.
Cardiac TnI concentration in serum was increased in CD-1 Increased AST and CK activities also suggested myocardial
mice treated with 0.1 mg/kg ISO. These increases were injury, but the magnitudes of change in these markers were rel-
below the threshold described above and occurred without atively small and the time for their detection short compared to
histologic evidence of myocardial necrosis. Although small alterations in the concentrations of cTnI. These limitations,
areas of injury could have been missed in histological exam- coupled with a lack of specificity for myocardium, limit their
ination of a single tissue section, it seems more likely that use as stand-alone cardiac biomarkers.
these relatively small increases in cTnI were indicative of Recent evidence supports the use of cardiac troponin con-
myocardial membrane leakage without cell death. Cells centrations in serum, in conjunction with examination of histo-
releasing cTnI likely experienced mechanical stress related logical sections of heart (Santucci et al. 2007) and
to increased contractility, or a transient period of ischemia, echocardiography (Kogan et al. 2007; Adamcova et al. 2007)
not lasting the 10 to 20 minutes required to cause cell death to explore the relationships of compound structure with phar-
(Fromm and Roberts 2001). Conceivably, this resulted in macology and cardiotoxicity in mice. Our experience suggests
temporary membrane disruption and leakage of intracellular that cTnI can be used to reliably and efficiently identify
contents from the cytoplasm. Similar scenarios have been xenobiotic-induced myocardial membrane disruption and
observed in baboons and dogs in which short episodes (15 necrosis without the need for more resource-intensive histolo-
minutes) of myocardial ischemia were induced, leading to gical and imaging-based examinations in short screening
significant increases in plasma activity of total CK as well studies.
Vol. 37, No. 5, 2009 IMPLEMENTATION OF cTnI IN MOUSE PHARMACOLOGY STUDIES 627

Screening for increased cTnI concentration in serum bene- Fromm, R. E., and Roberts R. (2001). Sensitivity and specificity of new serum
fited an early-stage kinase inhibitor drug discovery program. markers for mild cardionecrosis. Curr Prob Cardiol 26, 241–84.
Heyndrickx, G. R., Amano, J., Kenna, T., Fallon, J. T., Patrick, T. A., Manders,
Its use as a stand-alone cardiac safety endpoint gave insight W. T., Rogers, G. G., Rosendorff, C., and Vatner, S. F. (1985). Creatine
into each compound’s safety while assessing its pharmacol- kinase release not associated with myocardial necrosis after short periods
ogy using the same biological model, thus shortening cycle of coronary artery occlusion in conscious baboons. J Am Coll Cardiol 6,
time and reducing associated costs. Cardiac troponin I con- 1299–1303.
centration provided clear evidence of cardiac toxicity as phar- Kogan, N. M., Schlesinger, M., Peters, M., Marincheva, G., Beeri, R., and
Mechoulam, R. A. (2007). A cannabinoid anticancer quinone, HU-331,
macology was being assessed, allowing prioritization of is more potent and less cardiotoxic than doxorubicin: A comparative in
compounds according to potency, bioavailability, and safety. vivo study. J Pharmacol Exp Ther 322, 646–53.
Using cTnI as a stand-alone biomarker in mice allowed for Krutzik, P. O., Irish, J. M., Nolan, G. P., and Perez, O. D. (2004). Analysis of
higher throughput of compounds with less demand on protein phosphorylation and cellular signaling events by flow cytometry:
resources than required to synthesize compounds for toxicol- Techniques and clinical applications. Clin Immunol 110, 206–21.
Krutzik P. O., and Nolan G. P. (2003). Intracellular phospho-protein staining
ogy studies in larger species, such as rat. Without such screen- techniques for flow cytometry: Monitoring single cell signaling events.
ing, the lack of a relationship between target modulation and Cytometry A 55, 61–70.
cardiotoxicity would not have been as quickly evident and Larue, C., Defacque-Lacquemant, H., Calzolari, C., Le Nguyen, D., and Pau,
compound selection for more resource intensive studies not B. (1992). New monoclonal antibodies as probes for human cardiac tro-
as well informed. Subsequent studies revealed potent com- ponin I: Epitopic analysis with synthetic peptides. Mol Immunol 29,
271–8.
pounds with less cardiac liability that were advanced into Levene, H. (1960). Robust tests for equality of variance. In Contributions to
more extensive toxicology studies with a greater chance of Probability and Statistics (I. Olkin, ed.), pp. 278–92. Stanford University
a favorable safety profile. Press, Stanford, CA.
Mair, J., Genser, N., Morandell, D., Maier, J., Mair, P., Lechleitner, P., Calzo-
lari, C., Larue, C., Ambach, E., Dienstl, F., Pau, B., and Puschendorf, B.
(1996). Cardiac troponin I in the diagnosis of myocardial injury and
REFERENCES
infarction. Clin Chim Acta 245, 19–38.
Adamcova, M., Simunek, T., Kaiserova, H., Popelova, O., Sterba, M., Pota- O’Brien, P. J. (2006). Blood cardiac troponin in toxic myocardial injury:
cova, A., Vavrova, J., Malakova, J., and Gersl, V. (2007). In vitro and Archetype of a translational safety biomarker. Expert Rev Mol Diagn
in vivo examination of cardiac troponins as biochemical markers of 6, 685–702.
drug-induced toxicity. Toxicology 237, 218–28. O’Brien, P. J., Smith, D. E. C., Knechtel, T. J., Marchak, M. A., Pruimboom-
Adamcova, M., Sterba, M., Simunek, T., Potacova, A., Popelova, O., Mazur- Brees, I., Brees, D. J., Spratt, D. P., Archer, F. J., Butler, P., Potter, A. N.,
ova, Y., and Gersl, V. (2005). Troponin as a marker of myocardiac dam- Provost, J. P., Richard, J., Snyder, P. A., and Reagan W. J. (2006). Car-
age in drug-induced cardiotoxicity. Expert Opin Drug Saf 4, 457–72. diac troponin I is a sensitive, specific biomarker of cardiac injury in
Adams, J. E., III, Bodor G. S, Dàvila-Romàn, V. G, Delmez, J. A., Apple, F. S., laboratory animals. Lab Anim 40, 153–71.
Ladenson, J. H., and Jaffe, A. S. (1993). Cardiac troponin I: A marker Olson, H., Betton, G., Robinson, D., Thomas, K., Monro, A., Kolaja, G., Lilly,
with high specificity for cardiac injury. Circulation 88, 101–6. P., Sanders, J., Sipes, G., Bracken, W., Dorato, M., Van Deun, K., Smith,
Apple, F. S., Murakami, M. M., Ler, R., Walker, D., and York, M. (2008). Ana- P., Berger, B., and Heller, A. (2000). Concordance of the toxicity of phar-
lytical characteristics of commercial cardiac troponin I and T immunoas- maceuticals in humans and in animals. Regul Toxicol Pharmacol 23,
says in serum from rats, dogs, and monkeys with induced acute 56–67.
myocardial injury. Clin Chem 54, 1982–89. Pelsers, M. M., Hermens, W. T., and Glatz, J. F. (2005). Fatty acid-binding pro-
Babuin, L., and Jaffe, A. S. (2005). Troponin: The biomarker of choice for the teins as plasma markers of tissue injury. Clin Chim Acta 352, 15–35.
detection of cardiac injury. CMAJ 173, 1191–1202. Perrino, C., Naga Prasad, S. V., Schroder, J. N., Hata, J. A., Milano, C., and
Bleir, J., Vorderwinkler, K., Falkensammer, J., Mair, P., Dapunt, O., Rockman, H. A. (2005). Restoration of b-adrenergic receptor signaling
Puschendorf, B., and Mair, J. (1998). Different intracellular compart- and contractile function in heart failure by disruption of the bARK1/
mentations of cardiac troponins and myosin heavy chains: A causal phosphoinositide 3–kinase complex. Circulation 111, 2579–87.
connection to their different early release after myocardial damage. Clin Pritt, M. L., Hall, D. G., Recknor, J., Credille, K. M., Brown, D. D., Yumibe, N.
Chem 44, 1912–18. P., Schultze, A. E., and Watson, D. E. (2008). Fabp3 as a biomarker of
Collinson, P. O., and Gaze, D. C. (2007). Biomarkers of cardiovascular dam- skeletal muscle toxicity in the rat: Comparison with conventional biomar-
age. Med Princ Pract 16, 247–61. kers. Toxicol Sci 103, 382–96.
DeSilva, B., Smith, W., Weiner, R., Kelley, M., Smolec, J., Lee, B., Khan, M., Ruzgar, O., Bilge, A. K., Bugra, Z., Umman, S., Yilmaz, E., Ozben, B.,
Tacey, R., Hill, H., and Celniker, A. (2003). Recommendations for the Umman, B., and Meric, M. (2006). The use of human heart-type fatty
bioanalytical method validation of ligand-binding assays to support phar- acid-binding protein as an early diagnostic biochemical marker of myo-
macokinetic assessments of macromolecules. Pharm Res 20, 1885–1900. cardial necrosis in patients with acute coronary syndrome, and its com-
Faulx, M. D., Chandler, M. P., Zawaneh, M. S., Stanley, W. C., and Hoit, B. D. parison with troponin-T and creatine kinase-myocardial band. Heart
(2007). Mouse strain-specific differences in cardiac metabolic enzyme Vessels 21, 309–14.
activities observed in a model of isoproterenol-induced cardiac hypertro- Santucci, L., Mencarelli, A., Renga, B., Ceccobelli, D., Pasut, G., Veronese, F.
phy. Clin Exp Pharmacol Physiol 34, 77–80. M., Distrutti, E., and Fiorucci, S. (2007). Cardiac safety and antitumoral
Faulx, M. D., Ernsberger, P., Vatner, D., Hoffman, R. D., Lewis, W., Strachan, activity of a new nitric oxide derivative of pegylated epirubicin in mice.
R., and Hoit, B. D. (2005). Strain dependant b-adrenergic receptor func- Anticancer Drugs 18, 1081–91.
tion influences myocardial responses to isoproterenol stimulation in Shapiro, S. S., and Wilk, M. B. (1965). An analysis of variance test for normal-
mice. Am J Physiol Heart Circ Physiol 289, H30–H36. ity (complete samples). Biometrika 52, 591–611.
Fromm, R. E., Jr. (2007). Cardiac troponins in the intensive care unit: Common van Wandelen, C., and Cohen, S. A. (1997). Using quaternary high-
causes of increased levels and interpretation. Crit Care Med 35, 584–88. performance liquid chromatography eluent systems for separating
628 ENGLE ET AL. TOXICOLOGIC PATHOLOGY

6–aminoquinolyl-N-hydroxysuccinimidyl carbamate-derivatized amino Wu, A. H., Feng, Y. J., Moore, R., Apple, F. S., McPherson, P. H., Buechler, K.
acid mixtures. J Chromatogr A 763, 11–22. F., and Bodor, G. (1998). Characterization of cardiac troponin subunit
Vatner, S. F., Heyndrickx, G. R., and Fallon, J. T. (1986). Effects of brief peri- release into serum after acute myocardial infarction and comparison of
ods of myocardial ischemia on regional myocardial function and creatine assays for troponin T and I. Clin Chem 44, 1198–1208.
kinase release in conscious dogs and baboons. Can J Cardiol 2 Suppl A, Zhang, J., Knapton, A., Lipshultz, S. E., Weaver, J. L., and Herman, E. H.
19–24. (2008). Isoproterenol-induced cardiotoxicity in sprague-dawley rats:
Wu, A. H., and Feng, Y. J. (1998). Biochemical differences between cTnT and Correlation of reversible and irreversible myocardial injury with release
cTnI and their significance for diagnosis of acute coronary syndromes. of cardiac troponin T and roles of iNOS in myocardial injury. Toxicol
Eur Heart J 19 Suppl N, 25–29. Pathol 36, 277–88.

For reprints and permissions queries, please visit SAGE’s Web site at http://www.sagepub.com/journalsPermissions.nav

Das könnte Ihnen auch gefallen