You are on page 1of 17

DRUG DISPOSITION Clin Pharmacokinet 2002; 41 (15): 1229-1245

0312-5963/02/0015-1229/$25.00/0

© Adis International Limited. All rights reserved.

Pharmacokinetics and
Pharmacodynamics of Tenecteplase
in Fibrinolytic Therapy of Acute
Myocardial Infarction
Paul Tanswell,1 Nishit Modi,2,3 Dan Combs2 and Thierry Danays4
1 Department of Pharmacokinetics and Metabolism, Boehringer Ingelheim Pharma KG,
Biberach, Germany
2 Department of Pharmacokinetics and Metabolism, Genentech Inc, South San Francisco,
California, USA
3 ALZA Corporation, Mountain View, California, USA
4 Department of Medical and Regulatory Affairs, Boehringer Ingelheim, Reims, France

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1229
1. Biochemistry and Pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1231
1.1 Mechanism of Action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1231
1.2 Protein Engineering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1231
1.3 Preclinical Efficacy and Clot-Selectivity Measures . . . . . . . . . . . . . . . . . . . . . . . . . 1232
2. Pharmacokinetic Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1232
2.1 Plasma Assays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1232
2.2 Data Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1233
3. Preclinical Pharmacokinetics and Pharmacodynamics . . . . . . . . . . . . . . . . . . . . . . . . 1234
3.1 Distribution, Biotransformation and Elimination in Animal Studies . . . . . . . . . . . . . . . . 1234
3.2 Mechanism of Hepatic Elimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1234
3.3 Pharmacokinetics and Pharmacodynamics in Animals . . . . . . . . . . . . . . . . . . . . . . 1235
4. Clinical Pharmacokinetics and Pharmacodynamics . . . . . . . . . . . . . . . . . . . . . . . . . . 1236
4.1 Pharmacokinetics in Patients, Phase I . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1236
4.2 Pharmacokinetics in Patients, Phase II . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1237
4.3 Sources of Pharmacokinetic Variability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1238
4.4 In Vivo Markers of Pharmacodynamic Action . . . . . . . . . . . . . . . . . . . . . . . . . . . 1240
4.5 Pharmacokinetic-Pharmacodynamic Relationships . . . . . . . . . . . . . . . . . . . . . . . . 1242
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1243

Abstract Tenecteplase is a novel fibrinolytic protein bioengineered from human tissue


plasminogen activator (alteplase) for the therapy of acute ST-segment elevation
myocardial infarction. Specific mutations at three sites in the alteplase molecule
result in 15-fold higher fibrin specificity, 80-fold reduced binding affinity to the
physiological plasminogen activator inhibitor PAI-1 and 6-fold prolonged plasma
half-life (22 vs 3.5 minutes). Consequently, tenecteplase can be administered as
1230 Tanswell et al.

a single intravenous bolus of 30–50mg (0.53 mg/kg bodyweight) over 5–10 sec-
onds, in contrast to the 90-minute accelerated infusion regimen of alteplase.
Tenecteplase plasma concentration-time profiles have been obtained from a
total of 179 patients with acute myocardial infarction. Tenecteplase exhibited
biphasic disposition; the initial disposition phase was predominant with a mean
half-life of 17–24 minutes, and the mean terminal half-life was 65–132 min. Over
the clinically relevant dose range of 30–50mg, mean clearance (CL) was 105
ml/min. The mean initial volume of distribution V1 was 4.2–6.3L, approximating
plasma volume, and volume of distribution at steady state was 6.1–9.9L, suggest-
ing limited extravascular distribution or binding. Bodyweight and age were found
to influence significantly both CL and V1. Total bodyweight explained 19% of
the variability in CL and 11% of the variability in V1, and a 10kg increase in total
bodyweight resulted in a 9.6 ml/min increase in CL. This relationship aided the
development of a rationale for the weight-adjusted dose regimen for tenecteplase.
Age explained only a further 11% of the variability in CL.
The percentage of patients who achieved normal coronary blood flow was
clearly related to AUC. More than 75% of patients achieved normal flow at 90
minutes after administration when their partial AUC2–90 exceeded 320 μg •
min/ml, corresponding to an average plasma concentration of 3.6 μg/ml. Systemic
exposure to tenecteplase at all times after bolus administration of 30–50mg was
higher than for alteplase 100mg.
Tenecteplase has demonstrated equivalent efficacy and improved safety com-
pared with the current gold standard alteplase in a large mortality trial (ASSENT-
2). This suggests that the reduced clearance, greater fibrin specificity and higher
PAI-1 resistance of tenecteplase allow higher plasma concentrations and thus a
more rapid restoration of coronary patency to be attained, while providing a
reduction in major non-cerebral bleeding events.

The approval for clinical use of recombinant hu- sue Plasminogen Activator for Occluded Coronary
man tissue plasminogen activator (rt-PA, alte- Arteries (GUSTO I) mortality trial.[2] Neverthe-
plase) in 1987 represented a landmark in the less, it was soon recognised that further potential
pharmacological treatment of acute ST-segment existed for improvement in pharmacological throm-
elevation myocardial infarction. Alteplase is a bolysis, in particular ease of administration, more
fibrin-specific thrombolytic agent, derived from rapid and complete coronary artery reperfusion,
natural human t-PA, that is administered by an ac- reduction of bleeding complications and reduction
celerated intravenous infusion regimen because of of the incidence of reocclusion and reinfarction.[4]
its short half-life of 3.5 minutes.[1] Alteplase rap- Tenecteplase has been specifically bioengine-
idly dissolves blood clots in occluded coronary ar- ered for improved properties compared with al-
teries and significantly reduces morbidity and mor- teplase. These are: 15-fold higher fibrin specific-
tality compared both to placebo and to the previous ity, 80-fold reduced binding to the physiological
standard of treatment, streptokinase.[2] Streptoki- plasminogen activator inhibitor PAI-1[5] and 6-
nase, a non-fibrin-specific plasminogen activator fold prolonged plasma half-life, enabling single
of bacterial origin, activates clot-bound and sys- bolus administration.[6-8] These improved proper-
temic plasminogen indiscriminately, thus limiting ties were characterised extensively in preclinical
its efficacy and safety.[3] The superiority of alte- models and have been shown to translate into clini-
plase to streptokinase was demonstrated in the cal benefit.
large Global Utilization of Streptokinase and Tis- In the large phase III Assessment of the Safety

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
Tenecteplase 1231

and Efficacy of a New Thrombolytic (ASSENT-2) incidence of intracranial and non-cerebral bleed-
trial,[9] patients with acute myocardial infarction ing complications and blood transfusions.
who were treated with a single-bolus, bodyweight- Tenecteplase received marketing approval as
tiered dose of tenecteplase between 30 and 50mg TNKase®1 (Genentech, Inc.) in the USA in 1999
(equivalent to approximately 0.53 mg/kg) showed and as Metalyse® (Boehringer Ingelheim) in Eur-
equivalent 30-day mortality and incidence of in- ope in 2000. Clinical therapeutic trials of tenec-
tracranial haemorrhage compared with those teplase have recently been reviewed.[13-17] This re-
treated with a 90-minute alteplase infusion. Of a view therefore focuses on the pharmacokinetic and
total of 16 949 randomised patients in 1 021 hos- pharmacodynamic characteristics of tenecteplase
pitals, covariate-adjusted 30-day mortality rates and their significance for efficacy and safety in
acute myocardial infarction in comparison to al-
were almost identical for the two groups: 6.18%
teplase.
for tenecteplase and 6.15% for alteplase. The 95%
one-sided upper boundaries of the absolute and rel-
1. Biochemistry and Pharmacology
ative differences in 30-day mortality were 0.61%
and 10%, respectively, which met the prespecified
1.1 Mechanism of Action
criteria of equivalence (1% absolute or 14% rela-
tive difference, whichever proved smaller). Rates The blood fibrinolytic enzyme system is shown
of intracranial haemorrhage were similar (0.93% schematically in figure 1. Fibrinolysis is the pro-
for tenecteplase and 0.94% for alteplase). How- cess of dissolution of clots (thrombi), that were
ever, there were fewer non-cerebral bleeding com- formed by the conversion of soluble fibrinogen
plications (26.4% vs 29.0%, p = 0.0003) and less into insoluble fibrin as a result of physiological or
need for blood transfusion (4.25% vs 5.49%, p = pathological activation of the coagulation sys-
0.0002) with tenecteplase. Further, the patient sub- tem.[18] Plasminogen is a circulating plasma pro-
group with a time to treatment of greater than 4 tein that also binds to the solid-phase fibrin com-
hours (22.4% of patients) showed a lower mortal- ponent of a coronary thrombus. It is activated
ity with tenecteplase compared with alteplase physiologically or pharmacologically on the clot
surface by t-PA to plasmin, a serine protease. Plas-
(7.0% vs 9.2%, p = 0.018).
min digests the fibrin network of the clot, resulting
Reteplase is a further fibrinolytic agent with
in restoration of vessel patency. Fibrinolysis is
prolonged half-life that has recently been mar-
regulated by the inhibitors α2-antiplasmin and
keted for treatment of acute myocardial infarction,
plasminogen activator inhibitor 1 (PAI-1).[18] The
and is administered as two bolus doses 30 minutes mechanism of action of tenecteplase is identical to
apart.[10] Its structure is derived from t-PA by that of alteplase, with the enhancements described
deletion of amino acids 4–175 from the N-terminal below.
sequence; it is not glycosylated and is not fibrin-
specific.[3,11] Tenecteplase and reteplase have not 1.2 Protein Engineering
been compared directly in any clinical trial. How-
ever, reteplase was compared with alteplase in Extensive basic research was required to iden-
15 059 patients in the Global Use of Strategies to tify specific molecular changes in alteplase that
Open Occluded Coronary Arteries (GUSTO-III) might improve therapeutic performance. Initially,
study.[12] Reteplase did not provide any additional investigators modified only single amino acids or
deleted entire protein domains. Such mutations
survival benefit compared with alteplase; mortal-
yielded a desired reduction of plasma clearance,
ity rates at 30 days were 7.47 and 7.24%, respec-
tively. Also, there was no difference in safety be- 1 Use of tradenames is for product identification only and
tween reteplase and alteplase, as measured by the does not imply endorsement.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
1232 Tanswell et al.

Tenecteplase and alteplase had the same activity


Plasmin PAI-1
t-PA1c t-PA2c t-PA:PAI
(50% effective concentration, EC50, 3.1 μg/ml) in
an in vitro 125I-labelled human plasma clot lysis
α2-AP model.[23] The second-order rate constant for bind-
Plasmin:
ing of tenecteplase to the inhibitor PAI-1 was re-
Plasminogen Plasmin
antiplasmin duced 80-fold as compared with alteplase.[6] Be-
Fibrin
cause PAI-1 is present in high concentrations in the
platelet-rich clots which may occur in coronary
Thrombin Fibrin thrombi, it was expected that the considerably re-
Fibrinogen Fibrin degradation duced interaction of tenecteplase with PAI-1
products
would translate into greater fibrinolytic efficacy
towards platelet-rich clots compared with al-
Fig. 1. Mechanism of action of alteplase (t-PA) and ten-
ecteplase, corresponding to the physiological fibrinolytic sys- teplase. This hypothesis was confirmed in vivo in
tem. 1c and 2c indicate the one- and two-chain molecular forms a rabbit arteriovenous shunt thrombolytic model.[6]
of the plasminogen activator, respectively. PAI-1 = plasminogen The fibrin specificity of tenecteplase, expressed as
activator inhibitor type 1; α2-AP = α 2-antiplasmin.
the ratio of its catalytic activity in the presence
of fibrin as compared to fibrinogen, was 15-fold
but also resulted in impaired fibrinolytic activity higher than for alteplase.[8] As an in vivo correlate,
or fibrin specificity.[19] To overcome these limita- tenecteplase caused less peripheral bleeding than
tions, a high resolution functional analysis of the alteplase at equipotent thrombolytic doses in a rab-
alteplase protein sequence was performed,[20] as a bit model of carotid artery thrombosis.[24]
result of which the tenecteplase (TNK-tPA) mole-
cule was developed.[6] Tenecteplase is a single 2. Pharmacokinetic Methods
chain glycoprotein with 527 amino acids, 17 disul-
fide bridges and 65kD molecular mass, and is pro- 2.1 Plasma Assays
duced by recombinant DNA technology in chinese
hamster ovary cells. The enhanced properties rela- For pharmacokinetic studies, immunoreactive
tive to native alteplase were achieved by modifica- tenecteplase and alteplase were analysed in human
tion at three molecular sites, and are summarised and animal plasma using sensitive and validated
in figure 2 and table I. Like alteplase, tenecteplase two-site enzyme-linked immunosorbent assays
also exhibits type I and type II glycoforms.[21] Type (ELISA).[25,26] The lower limit of quantification
I has three carbohydrate structures at asparagine was 8 ng/ml for both analytes, and alteplase was
residues 103, 184 and 448, whereas type II lacks found to exhibit 100% immunoreactivity in the ten-
the carbohydrate at asparagine 184. Tenecteplase ecteplase ELISA. The alteplase ELISA had been
carbohydrates are all complex oligosaccharides shown to correlate well with a functional fibrino-
and no high mannose structure is present. Thus, the lytic activity assay.[1] Blood samples for ELISA
rapid clearance mediated by the hepatic mannose analysis were collected in presence of EDTA anti-
receptor observed for alteplase[22] does not occur coagulant and the protease inhibitors D-Phe-Pro-
in tenecteplase. Arg-chloromethyl ketone (PPACK) and aprotinin.
PPACK irreversibly inhibits the serine protease ac-
1.3 Preclinical Efficacy and tivity of tenecteplase or alteplase and thus prevents
Clot-Selectivity Measures artifactual in vitro formation of complexes with
inhibitor proteins in plasma that are non-immuno-
The fibrin binding affinity of tenecteplase (dis- reactive in the ELISA.[27,28] PPACK and aprotinin
sociation constant, KD, 0.35 μmol/L or 23 ng/ml) also prevent artifactual in vitro consumption of
was similar to that of t-PA (KD 0.21 μmol/L).[6] plasminogen and degradation of α2-antiplasmin

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
Tenecteplase 1233

and fibrinogen, [28] which are important in vivo 2.2 Data Analysis
biomarkers of tenecteplase and alteplase effects on
blood coagulation. Assays of clinical serum sam- Tenecteplase and alteplase pharmacokinetic
ples to assess potential antibody formation against parameters were derived from individual plasma
tenecteplase were performed using a validated concentration profiles using either noncompart-
radioimmuno-precipitation technique.[29] mental methods or multicompartmental intrave-

G A S C L
P Y S E S P
W
A Q K G R T N
A L R P L L V H CW S
S D S K M
S A P I
Q H N K
I L
W 117 Kringle 1 R T R A I
N L G R D G
T P K S D G R L G K
C W R D L Y T D
C P V
E P G A W
Y N R N Y
A
V C Y N HN S E
R T
G F G Y A
C Kringle 2
S K K Y S S E F C N 184 C
Y Q
E A G G D
S N N
A T F V
T 103 P H P A S H
SW P Y S S I P
N G R Y S I G Q A C K D W
C G A
D C D L Q A Q
E S E S S G L A F
Y G N A
T L
C C G A
T G G I
A P
L C
V L I K HK A F
G K C R Q F R R
A R P T Y S Q P Q R A
C E T A R 275 A A 296
F I D D A
G
V
G P S 299
E P C Q L V R E
C
V Q S L I G G C L F
F I
L S S P P H H
L R P Growth R C F D Q E R F
SW V P N S P E S C F
L
T
Q L factor F D Q C A H V
E G A A I
H R Y W W I L S L
S G C
Q S T L T G
Q Finger C E R D N D R
N S CQ Q A S I Y
Y K C S L A T T
R D S K L Q L
I V P V D N L E D Y
V S G C L D R
M R V
Q E H S D V
T Y C M L G F
K Q T P Y
G Protease E
V
E C A K H P
L G K
D W R G 478 E A L G
R C V S P F H
N G G S Y S E
C S I D E
I
S
* G R V
E
I W Q L I
V G C K Y E
Q L A K Q
G D E
Y C H A E V E F K
S G L
Q N H
1 K A V
D Q
P R
V G
P G L
G S Y
V R P
Y T S
T D S
K G
A R
527 V C
P T C
L T
R N S
M Y M Q
N L N H
DR D D L
I W
T 448 L
V T R N

Fig. 2. Secondary structure of tenecteplase showing the domains, disulfide bridges, glycosylation sites and the plasmin cleavage
site at Arg-275, which generates the two-chain molecular form. The protease domain (amino acid residues 276–527) cleaves
plasminogen to plasmin (see figure 1). The remaining domains (finger, growth factor, kringle 1 and kringle 2) mediate other ten-
ecteplase biological functions such as fibrin binding, fibrin specificity and hepatic clearance. The bioengineering modifications that
were performed relative to wild-type tissue plasminogen activator (alteplase) are summarised in table 1.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
1234 Tanswell et al.

Table I. Description of the molecular modifications in tenecteplase (TNK-tPA) compared to alteplase (t-PA), as depicted in figure 2
Designation Amino acid substitution Effect
T Thr-103 →Asn Adds a new glycosylation site on kringle-1, which decreases the rate of
clearance but decreases fibrin binding
N Asn-117→ Gln Removes the existing glycosylation site on kringle-1, which decreases
the rate of clearance and restores fibrin binding in combination with
Thr-103 → Asn
K Lys-His-Arg-Arg (296-299) → Increases fibrin specificity and makes the molecule more resistant to the
Ala-Ala-Ala-Ala naturally occurring inhibitor PAI-1
K = lysine; N = asparagine; PAI = plasminogen activator inhibitor; T = threonine.

nous disposition models using the TOPFIT, marrow). The tissue-to-blood ratios for these or-
PCNonlin or WinNonlin software.[1,7,26] Pharmaco- gans in rank order were 3.8, 3.7, 3.6, 2.5, 2.4, 2.1,
kinetic parameters were derived from the models 1.3 and 1.2, respectively. No radioactivity was de-
using standard methods.[30] Because the tenecte- tected in the brain. It appeared that tenecteplase
plase ELISA used in the clinical pharmacokinetic distributed to similar tissues as alteplase,[35] but
studies did not distinguish between endogenous t- with different kinetics. Gradient gel electrophore-
PA and tenecteplase, concentrations detected prior sis was performed on plasma and urine samples in
to tenecteplase administration and post-dose con- order to characterise the nature of circulating and
centrations of less than 30 ng/ml were not included excreted radioactivity.[33]
in pharmacokinetic analyses.[26] So far, no pharma- No low-molecular-weight polypeptides derived
cokinetic analyses have been reported using the from 125I-tenecteplase or 125I-alteplase were ob-
population approach and nonlinear mixed effects served in plasma, although there was evidence of
modelling,[31] but such work is currently ongoing. the formation of higher molecular weight com-
plexes with other plasma proteins. It is likely that
3. Preclinical Pharmacokinetics and these correspond to an association between the
Pharmacodynamics tenecteplase serine protease domain and several
plasma protein inhibitors, which have been well
3.1 Distribution, Biotransformation and characterised for alteplase.[36] Radioactivity ex-
Elimination in Animal Studies creted in urine was primarily trichloroacetic acid
soluble, and no intact (62–65kD) test material was
A whole body autoradiography study was con- observed. A radiolabelled degradation fragment of
ducted in rats[32,33] to assess tissue distribution fol- 28–30kD, believed to be the protease domain, was
lowing a single intravenous bolus injection of ten- detected at very low levels in urine (<2% of the
ecteplase labelled with the novel active site reagent radioactive dose) following administration of ei-
125I-tyrosyl-prolyl-arginyl chloromethyl ketone[34] ther 125I-tenecteplase or 125I-alteplase.
compared to alteplase. The results clearly demon- Overall, the results were consistent with hepatic
strated that the liver was the major clearance organ uptake of tenecteplase followed by catabolism to
for both 125I-tenecteplase and 125I-alteplase. The small peptides, as described for alteplase.[37]
liver was the only organ that had significant uptake
of radioactivity 20 minutes after administration of 3.2 Mechanism of Hepatic Elimination
125I-tenecteplase, with a tissue-to-blood ratio of 2.0

(2.7% of dose/g liver). By 90 minutes, the liver-to- Strong evidence for the hepatic elimination of
blood ratio increased to 9.1 (6.0% of dose/g liver) tenecteplase in humans is provided by the afore-
and relatively little uptake of radioactivity in other mentioned rat studies, the observation that al-
highly perfused organs was noted (spleen, intes- teplase is cleared in human liver,[37,38] and the
tine, thyroid, bladder, kidney, adrenal, lung, bone structural similarity of tenecteplase and alteplase.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
Tenecteplase 1235

However, it is not precisely known which hepatic for tenecteplase than for alteplase. Plasma clear-
receptor systems are responsible for tenecteplase ance of tenecteplase was reduced significantly in
elimination. Tenecteplase, like alteplase, is a gly- all species by a factor of 4–7.5 compared with al-
coprotein but with differing oligosaccharide struc- teplase, as summarised in table II. Clearance data
tures. The hepatic mannose receptor, which is im- in patients from the Thrombolysis in Myocardial
portant for alteplase elimination,[22] cannot be Infarction (TIMI) 10B phase II clinical trial are
involved in tenecteplase clearance because tenec- also included in table II and demonstrate a similar
teplase does not contain high-mannose oligosac- 4-fold decrease in clearance of tenecteplase com-
charide. This glycosylation site was eliminated by pared with alteplase. The initial volume of distri-
mutation of amino acid residue Asn-117 to Gln. bution (V1) of tenecteplase in the animal species
The hepatic low-density lipoprotein receptor re- generally approximated plasma volume, and steady-
lated protein (LRP)[39] could be involved in clear- state volume of distribution (Vss) was approxi-
ance of both alteplase and tenecteplase via a non- mately 1.5- to 2-fold greater than V1 (data not
carbohydrate-mediated mechanism. In addition, shown). Similar pharmacokinetic characteristics
the hepatic asialoglycoprotein receptor, which were found in humans, and are described in section
binds terminal galactose,[40] may also mediate 4. Thus overall, the pharmacokinetics of ten-
clearance of alteplase and tenecteplase. ecteplase can be regarded as highly consistent
across animal species and humans.
3.3 Pharmacokinetics and
The principal experimental pharmacological
Pharmacodynamics in Animals
model used for investigating the fibrinolytic po-
The animal species mouse, rat, rabbit, dog and tency of tenecteplase in vivo consisted of rabbits
monkey were used in safety pharmacology, indi- that were fitted with an extracorporal arteriove-
cation-specific pharmacology and toxicity studies nous shunt containing clots made from whole
of tenecteplase. Extensive pharmacokinetic stud- blood or platelet-enriched plasma.[6] It was found
ies were performed in these species to define ten- that bolus tenecteplase induced 50% lysis in one-
ecteplase systemic exposure for comparison with third of the time required by the same dose of al-
clinical therapy.[33] teplase by infusion, and that in rabbits tenecteplase
Tenecteplase, like alteplase,[41] demonstrated was 8- and 13-fold more potent on a mg/kg basis
multiexponential, three-compartment pharmaco- than alteplase to whole blood and platelet-enriched
kinetics in animals, which did not allow the assign- clots, respectively. This superior potency was at-
ment of a principal plasma half-life. However, tributable to pharmacokinetics (reduced clear-
mean residence time was always markedly longer ance) of tenecteplase in the whole blood clot

Table II. Comparison of plasma clearance of tenecteplase and alteplase in various animal species and humans
Speciesa Tenecteplase Alteplase Clearance ratio
(alteplase/tenecteplase)
dose (mg/kg) CL (ml/min/kg) dose (mg/kg) CL (ml/min/kg)
Mouse 0.3 4.9 0.3 29 5.9
Rat 0.3 4.7 0.3 27 5.7
Rabbit 0.3 1.9 0.3 16.1 7.5
Dog 0.3 4.1 0.3 23 5.6
Rhesus monkey 0.3 2.4 0.3 9.5 4.0
Humanb 0.5 1.4 1.22 5.5 4.0
a Single dose animal pharmacokinetic data.[33]
b Clinical phase II, TIMI 10B study, 40mg dose group.[7]
CL = total body clearance; TIMI = Thrombolysis in Myocardial Infarction.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
1236 Tanswell et al.

model, and to a combination of pharmacokinetics In contrast to alteplase,[44] no pharmacokinetic


and pharmacodynamics (enhanced PAI-1 resis- studies were performed with tenecteplase in healthy
tance) in the platelet-enriched clot model. In phase volunteers because of ethical concerns arising
II clinical studies, tenecteplase 40mg was found to from its increased fibrinolytic potency and longer
achieve approximately equivalent efficacy and half-life. The pharmacokinetics of tenecteplase
safety to alteplase 100mg.[42,43] This indicated a were studied in 179 patients with acute myocardial
relative potency of about 2.5. While not exhibiting infarction during clinical development up to regu-
the same potency increase compared with alteplase latory approval. Comparative data with alteplase
as in the rabbit model, the effective total clinical were obtained concurrently in an additional 53 pa-
tients. Table III indicates the clinical studies for
dose of bolus tenecteplase was nevertheless con-
which pharmacokinetic data for tenecteplase or al-
siderably lower than that of infused al-
teplase were obtained. Two large pharmacokinetic
teplase.[9,42,43]
substudies were integrated into the TIMI 10A
phase I dose-ranging clinical trial[26,45] and the
4. Clinical Pharmacokinetics and TIMI 10B phase II angiographic clinical trial.[7,42]
Pharmacodynamics No pharmacokinetic data were obtained in the
phase II safety trial (ASSENT-1)[43] and in the
Adequate blood sampling for pharmacokinetic
large phase III trial (ASSENT-2),[9] in order not to
studies of tenecteplase or alteplase in patients with compromise the clinical objectives of these trials.
an acute myocardial infarction is subject to consid- Post-approval clinical trials have been aimed prin-
erable practical constraints because these agents cipally at studying combinations of tenecteplase
are given in an intensive care setting as a single with antithrombotic agents, for example en-
dose only. This greatly restricts the time frame in oxaparin or abciximab in ASSENT-3,[46] and no
which sampling may be performed. During myo- further tenecteplase pharmacokinetic data were
cardial infarction, patients may exhibit fluctua- obtained in those studies.
tions in cardiac output and hepatic blood flow
which increase intra-individual pharmacokinetic 4.1 Pharmacokinetics in Patients, Phase I
variability. The required blood sampling schedule,
including the necessary addition of protease inhib- In the phase I (TIMI 10A) study, pharmacoki-
itors, often coincides with coronary angiography netic parameters were obtained over a wide ten-
or other invasive procedures, which may also af- ecteplase dose range of 5–50mg, administered as
fect the pharmacokinetic results. a single intravenous bolus over 5–10 seconds in

Table III. Summary of tenecteplase clinical trials for regulatory approval and patient subgroups used for pharmacokinetic analysis
Study (objective) Clinical Dose range No. of patients entered PK data No. of patients in PK analysis References
phase (tenecteplase/alteplase) obtained?
tenecteplase alteplase
TIMI 10A I 5–50mg 113/– Yes 80 26,45
(dose ranging)
TIMI 10B II 30, 40, 50mg 555/325 Yes 99 53 7,42
(efficacy)
ASSENT-1 II 30, 40, 50mg 3235/– No 43
(safety)
ASSENT-2 III Bodyweight 8461/8488 No 9
(safety and adjusted: 0.53
efficacy) mg/kg
ASSENT = Assessment of the Safety and Efficacy of a New Thrombolytic; PK = pharmacokinetic; TIMI = Thrombolysis in Myocardial Infarction.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
Tenecteplase 1237

Table IV. Principal pharmacokinetic parameters of tenecteplase in comparison with alteplase in patients with acute myocardial infarction
Dose (mg) and regimen n CL (ml/min) Cmax (μg/ml) V1 (L) Vss (L) Initial t1⁄2 Terminal t1⁄2 Initial AUC
(min) (min) (%)

Tenecteplase bolus, phase I (TIMI 10A)[26]


5 5 216 ± 98 0.70 ± 0.39 8.4 ± 6.3 10 ± 5.3 11 ± 5.2 41 ± 16 31 ± 22
7.5 5 168 ± 66 1.0 ± 0.07 5.8 ± 1.8 8.4 ± 2.8 16 ± 8.5 96 ± 73 58 ± 16
10 5 185 ± 35 1.5 ± 0.30 6.4 ± 1.1 11 ± 5.5 15 ± 8.1 121 ± 100 57 ± 14
15 6 182 ± 76 2.7 ± 1.2 6.7 ± 3.3 9.6 ± 3.7 11 ± 7.9 54 ± 13 42 ± 30
20 7 169 ± 63 2.8 ± 6.5 7.6 ± 2.7 15 ± 7.3 18 ± 6.8 138 ± 84 56 ± 25
30 26 143 ± 45 5.9 ± 3.0 6.3 ± 4.0 9.9 ± 6.0 18 ± 7.7 132 ± 140 64 ± 20
40 18 130 ± 41 9.5 ± 3.4 4.3 ± 1.9 6.3 ± 1.8 17 ± 5.6 88 ± 45 69 ± 15
50 7 125 ± 25 8.8 ± 1.4 5.7 ± 1.4 6.7 ± 1.3 20 ± 5.5 65 ± 25 55 ± 29

Tenecteplase bolus, phase II (TIMI 10B)[7]


30 48 98.5 ± 42 7.5 ± 5.2 4.2 ± 2.6 6.3 ± 3.3 22 ± 8.2 116 ± 63 72 ± 22
40 31 119 ± 49 9.5 ± 8.1 5.4 ± 2.7 8.0 ± 5.9 24 ± 5.5 129 ± 87 75 ± 16
50 20 99.9 ± 32 11.6 ± 4.4 4.7 ± 2.6 6.1 ± 2.4 20 ± 10 90.4 ± 35 66 ± 29

Alteplase infusion over 90 minutes


100 (phase II, TIMI 10B[7])a 53 453 ± 170 4.3 ± 3.4 7.2 ± 4.1 28.9 ± 22 144 ± 100
100 (earlier data, TAPS[1]) 10 572 ± 130 4.0 ± 1.0 3.4 ± 1.5 8.4 ± 5.0 3.5 ± 1.4 72 ± 68 85 ± 15
a Initial t1⁄2 of alteplase not sampled, pharmacokinetic data are from noncompartmental analysis.
AUC = area under the concentration-time curve; CL = total body clearance; Cmax = peak plasma concentration; n = number of patients;
TAPS = rt-PA-APSAC Patency Study; TIMI = Thrombolysis in Myocardial Infarction; t1⁄2 = half-life; Vss = volume of distribution at steady
state; V1 = initial volume of distribution.

patients with acute myocardial infarction.[26,45] rationale, based on the considerably reduced
The number of patients for whom pharmacokinetic plasma clearance and prolonged half-life com-
data was obtained in each group varied between 5 pared with alteplase, to support further clinical de-
and 26. Tenecteplase exhibited biphasic (two- velopment of tenecteplase using a single intrave-
compartment) kinetics in 60 patients, and mono- nous bolus rather than an infusion regimen.
phasic kinetics in 20 patients. It is likely that ten-
ecteplase disposition is generally biphasic, but in 4.2 Pharmacokinetics in Patients, Phase II
some patients only a single compartment was de- In the phase II (TIMI 10B) pharmacokinetic
tected. The principal pharmacokinetic parameters study, tenecteplase bolus doses of 30, 40 and 50mg
are listed in table IV. Clearance of tenecteplase were administered and compared with alteplase
decreased from 216 ml/min at the 5mg dose to 125 100mg given as a 90-minute accelerated infusion
ml/min at 50mg (table IV, figure 3), indicating a regimen.[7] The plasma concentration-time pro-
moderate pharmacokinetic nonlinearity.[26] How- files are shown in figure 4, and pharmacokinetic
ever, in the dose range 30–50mg, which was parameters are summarised in table IV.
judged to be clinically relevant based on an- Tenecteplase exhibited two-compartment ki-
giographic efficacy, there was no significant dif- netics in 90 out of 99 patients, and alteplase phar-
ference in clearance (125–143 ml/min). This clear- macokinetics were evaluated by noncompartmen-
ance was 4-fold slower, and the initial half-life of tal analysis. In contrast to the phase I (TIMI 10A)
17–20 min, which corresponded to 55–69% of area study there was no dose dependence of clearance,
under the concentration-time curve (AUC), was 5- probably because of the more restricted dose
fold longer, than reported for alteplase.[1] This range. The mean initial half-life was 22 minutes
study, therefore, provided a clear pharmacokinetic and the mean terminal half-life was 115 minutes.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
1238 Tanswell et al.

400 oligosaccharide moieties of the tenecteplase mol-


ecule and may therefore reduce its clearance by the
350 hepatic asialoglycoprotein receptor.[33,40]
300
The initial half-life of tenecteplase was domi-
nant in both studies, accounting for 55–72% of
CL (ml/min)

250 AUC. Overall, V1 (4.2–6.3L) approximated plas-


ma volume, and Vss was 6.1–9.9L, suggesting
200
mainly intravascular distribution. These parame-
150 ters are consistent with the large molecular mass of
tenecteplase (65kD). In contrast to alteplase,[36] lit-
100 tle information is available on binding of ten-
50
ecteplase to plasma proteins in humans. Recently,
0 10 20 30 40 50 60 slow complex formation between tenecteplase and
Dose (mg) C1 inhibitor was demonstrated in vitro in human
plasma.[47]
Fig. 3. Dependence of tenecteplase plasma clearance (CL) on
dose in the Thrombolysis in Myocardial Infarction (TIMI) 10A
4.3 Sources of Pharmacokinetic Variability
phase I clinical trial in patients with acute myocardial infarction
(reproduced from Modi et al.,[26] with permission).
Interindividual variability of tenecteplase phar-
macokinetic parameters was relatively high (table
IV). For example, overall coefficients of variation
The mean clearance of tenecteplase (105 ml/min)
were 41% for clearance and 58% for V1 in the TIMI
was 4.3-fold slower than that measured for al-
10B study. The intra-individual (residual) variabil-
teplase in TIMI 10B, and the initial (dominant) ity was not quantitatively assessed, and a combined
half-life was 6.3 times longer than that reported analysis of the TIMI 10A and TIMI 10 B studies
for alteplase in the rt-PA-APSAC Patency Study was not performed; both aspects would require fur-
(TAPS),[1] confirming the phase I results. Other ther analysis by population mixed effect model-
pharmacokinetic parameters of alteplase measured
in TIMI 10B were similar to those reported in the
TAPS study.[1] 100 000 30mg tenecteplase (n = 48)
Tenecteplase pharmacokinetics were very simi- 40mg tenecteplase (n = 31)
Plasma concentration (μg/L)

lar in both the TIMI 10A and TIMI 10B studies 50mg tenecteplase (n = 20)
10 000 100mg alteplase (n = 53)
(table IV). The initial plasma half-life was slightly
longer in TIMI 10B (20–24 min) than in TIMI 10A
1 000
(17–20 min), but this may be attributable to the
more sparse blood sampling in TIMI 10B (reduced
from 15 to 8 samples per patient), and the differ- 100

ence is not clinically important. Although ten-


ecteplase clearance was similar at the 40 and 50mg 10
0 60 120 180 240 300 360 420
doses, clearance at the 30mg dose was 31% lower
Time (min)
in TIMI 10B than in TIMI 10A. This may have
been attributable to known minor increases in sia- Fig. 4. Pharmacokinetics in the Thrombolysis in Myocardial In-
lic acid content resulting from manufacturing scale farction (TIMI) 10B phase II clinical trial. Mean immunoreactive
changes and process optimisation of tenecteplase plasma concentration-time profiles for tenecteplase following an
intravenous bolus dose and for alteplase following the acceler-
that occurred after TIMI 10A.[7,33] Increases in sia- ated infusion regimen (100mg in 90 minutes) [reproduced from
lic acid content will block exposed galactose in Modi et al.,[7] with permission].

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
Tenecteplase 1239

ling.[31] In the TIMI 10A and TIMI 10B studies, decrease of clearance with age may reflect reduced
two-stage data analysis approaches were used to expression and/or function of hepatic receptor sys-
identify patient covariates that contributed to inter- tems for tenecteplase catabolism. With initial dis-
individual pharmacokinetic variability. These in- tribution corresponding to plasma volume, the in-
volved stepwise linear regression of pharmacoki- crease of V1 with bodyweight observed in TIMI
netic parameters on covariates after the former had 10B can also be rationalised, because heavier pa-
been computed by model fitting of individual tients will have higher blood volume. Men had
plasma concentration profiles. Demographic data higher mean clearance than women, but the effect
for patients of the TIMI 10 B pharmacokinetic was not statistically significant and may have been
study are given for reference in table V. attributable to the greater bodyweight of male pa-
In the TIMI 10A study, dose followed by total tients. Also, only 28% of the patients with com-
or lean bodyweight, age and gender were signifi- plete demographic data in the pharmacokinetic
cantly correlated with clearance, whereas none in- subset of TIMI 10B were women, which reflected
fluenced V1.[26] Dose was the most important cov- the low female/male ratio in the full trial popula-
ariate but only explained 17% of the variability in tion[42] and acute myocardial infarction studies in
clearance. On including all covariates in the re- general[9] (table V).
gression model, still only 30% of the total variabil- No separate clinical trials were performed to
ity could be accounted for. In TIMI 10B, total or investigate specifically the pharmacokinetics of
lean bodyweight and age were found significantly tenecteplase in special populations such as individ-
to influence both clearance and V1.[7] However, the uals with hepatic and renal insufficiency. Rat stud-
predictive value of these covariates was again ies showed that tenecteplase is not excreted intact
small. Total bodyweight explained 19% of the into the urine.[33] Also, the molecular mass of ten-
variability in clearance and 11% of the variability ecteplase, which is close to that of serum albumin,
in V1. Age explained a further 11% of the variabil- precludes glomerular filtration in any species.
ity in clearance. A 10kg increase in total body- Therefore, it is not expected that renal dysfunction
weight resulted in a 9.6 ml/min increase in plasma will affect pharmacokinetics. The effect of hepatic
clearance. Conversely, a 10-year increase in pa- dysfunction on the pharmacokinetics of ten-
tient age resulted in a 16.1 ml/min decrease in ecteplase in humans was not studied.
plasma clearance. These effects are summarised No separate clinical studies were performed to
graphically in figure 5. evaluate specifically the potential for pharmacoki-
In both studies, clearance increased with body- netic interaction between tenecteplase and other
weight and decreased with age. This is not unex- drugs commonly administered in the treatment of
pected on consideration of the hepatic elimination subjects with acute myocardial infarction. All
route for tenecteplase, because heavier patients study patients included in the pharmacokinetic
might have higher liver weights and therefore in- analyses received aspirin and heparin as concom-
creased capacity for tenecteplase clearance. The itant medication.[42,45] A study in dogs revealed no

Table V. Patient demographic data for the pharmacokinetic part of the TIMI 10B study (reproduced from Modi et al.,[7] with permission)
Characteristic Tenecteplase Alteplase
30mg 40mg 50mg 100mg
Male/female 29/18 23/6 17/3 38/15
Age (years) 56.8 ± 11 55.2 ± 11 59.0 ± 11 57.8 ± 12
Weight (kg) 84.7 ± 17 85.4 ± 25 78.8 ± 15 81.8 ± 16
Lean bodyweight (kg) 58.3 ± 11 58.9 ± 9.4 58.5 ± 9.7 57.8 ± 10
TIMI = Thrombolysis in Myocardial Infarction.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
1240 Tanswell et al.

V1 could only partially be explained by the patient


y = 25.331 + 0.961 • x; r2 = 0.185
covariates bodyweight and age and therefore
300
largely reflect random variability in distribution
250 and clearance of tenecteplase in patients with acute
myocardial infarction.
200
Antibodies to tenecteplase were not detected in
CL (ml/min)

150 any patient at 30 days in the TIMI 10A trial, al-


though one patient had a positive titre before treat-
100 ment.[45] In the TIMI 10B trial, antibodies were
50
detected in only 1 out of 364 patients at 30 days
after administration, and no antibodies were de-
0 tected in this patient at 90 days.[42] Thus, ten-
40 60 80 100 120 140 160 180 200 ecteplase is essentially non-antigenic after single
Weight (kg)
administration, and antibody formation can be ex-
cluded as a source of pharmacokinetic or pharma-
300 y = 197.054 − 1.608 • x; r2 = 0.169
codynamic variability.

250
4.4 In Vivo Markers of Pharmacodynamic
200 Action
CL (ml/min)

150
The fibrinolytic enzyme reaction scheme de-
100 picted in figure 1, in which alteplase/tenecteplase
activates plasminogen to plasmin preferentially on
50
the solid-phase fibrin surface, has the following
0 two main features.
30 40 50 60 70 80 90
• Fibrinolysis or clot dissolution is the desired
Age (years)
pharmacological effect, where the fibrin com-
ponent of the blood clot is dissolved by plasmin
Fig. 5. Dependence of tenecteplase plasma clearance (CL) on to form measurable soluble fibrin degradation
bodyweight and age in the Thrombolysis in Myocardial Infarc-
tion (TIMI) 10B phase II trial (reproduced from Modi et al.,[7] with products.
permission). • In systemic lysis, a certain amount of plasmin-
ogen is additionally activated to plasmin in the
fluid phase, resulting in measurable degradation
effect of either aspirin or heparin on the pharma- of coagulation proteins such as fibrinogen and
cokinetics of tenecteplase.[33] Tenecteplase clear- consumption of plasminogen and α2-anti-
ance in the TIMI 10A study was not affected by plasmin. This is undesirable because it reduces
concomitant administration of nitrates or β-block- the coagulability of blood and may be responsi-
ers. [26] Alteplase clearance has been shown to ble for adverse bleeding effects.
exhibit liver blood flow dependence,[48] but the The extent to which systemic lysis occurs for a
substantially reduced hepatic clearance of ten- given amount of fibrinolysis decreases with in-
ecteplase will decrease the likelihood that it would creasing fibrin specificity of the fibrinolytic agent
be influenced by drugs that alter hepatic blood used.[3] Thus, tenecteplase at therapeutically equiva-
flow. lent doses to alteplase results in less consumption
In summary, the relatively high observed inter- of fibrinogen, plasminogen and α2-antiplasmin as
individual variability of tenecteplase clearance and measured in plasma samples by coagulation labo-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
Tenecteplase 1241

ratory methods in clinical trials,[42] as shown in and fibrin degradation products are heterogeneous
figure 6. in nature, posing problems for specific laboratory
Mathematical models for alteplase pharmaco- analysis.[55] Examples of more specific serum
dynamic action in fibrinolysis based on the en- biomarkers that originate from myocardium, and
zymatic reactions of figure 1 have been devel- have been recently explored as indicators of coro-
oped.[49,50] They can be regarded as a type of nary recanalisation or survival, are creatine kinase
indirect pharmacodynamic response model,[51] be- MM subforms[56] and troponin T or I.[57,58] The
cause the therapeutic agent (plasminogen activa- preferred clinical/pharmacological and therapeu-
tor) stimulates the production of an endogenous
tic efficacy measures of tenecteplase and alteplase
enzyme (plasmin) that mediates both the pharma-
include:
cological effect (clot lysis) and adverse effects
• extent of recanalisation of the culprit coronary
(consumption of coagulation/fibrinolytic proteins)
artery determined angiographically according
as a function of plasma concentration of the agent.
to TIMI flow grade;[42,59]
However, although mechanistic pharmacody-
namic models for alteplase and tenecteplase based • corrected TIMI frame count;[60]
on enyzmatic reactions in plasma have been exten- • all-cause mortality at 30 days;[9]
sively used for in vitro studies,[8,52] they have • composite endpoints such as 30-day mortality,
found essentially no application in clinical therapy. in-hospital re-infarction or in-hospital refrac-
With respect to efficacy, this is because fibrin tory ischaemia.[46]
degradation products in plasma are not specific The composite endpoints probably reflect the
biomarkers for the lysis of coronary thrombi.[53] combined effects of fibrinolysis and conjunctive
Fibrin occurs elsewhere in the vascular system,[54] anticoagulation.[3]

30 mg
40 mg
50 mg
Alteplase

α2-Antiplasmin Fibrinogen Plasminogen

0
0
−10 −10

−20 −20
−20
Change (%)

−30
−40
−30
−40
−60
−40 −50

−80 −60

0 1 3 6 0 1 3 6 0 1 3 6
Time post-dose (h)
Fig. 6. Tenecteplase pharmacodynamics showing improved fibrin specificity. Markedly reduced systemic effects occurred during
fibrinolytic therapy with tenecteplase (bolus doses in mg, indicated by the symbols) compared with alteplase infusions of 100mg
over 90 minutes in the Thrombolysis in Myocardial Infarction (TIMI) 10B study, as measured by the time course of median concen-
trations of the proteins of the fibrinolytic system α 2-antiplasmin and plasminogen, and the coagulation protein fibrinogen (reproduced
from Cannon et al.,[42] with permission).

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
1242 Tanswell et al.

With respect to safety measures, the time 100


courses of concentrations of fibrinogen, plasmino-
gen, α2-antiplasmin and other coagulation/fibri-

TIMI-3 flow at 90 minutes (%)


80

Patients who achieved


nolysis proteins in plasma are useful indicators of
the relative fibrin specificity of tenecteplase com- 60
pared with alteplase and other plasminogen activa-
tors (figure 6).[42,49] However, the preferred clini- 40
cal variables that are used to assess safety in
therapeutic trials are the incidence of intracranial 20
bleeding, non-cerebral major or minor bleeding ep-
isodes and blood transfusion requirements.[9,14,43] 0
Intracranial haemorrhage is the most serious bleed- n = 17 n = 17 n = 19 n = 16 n = 16
<220 220-260 260-320 320-430 >430
ing complication, which is estimated to occur at a
AUC2-90 (mg • min/L)
low frequency of 0.5–1% in large-scale studies of
patients with acute myocardial infarction.[61]
Fig. 7. Tenecteplase pharmacodynamics showing the relation-
ship between the partial area under the concentration-time
4.5 Pharmacokinetic-Pharmacodynamic curve from 2 to 90 minutes (AUC2–90) and the percentage of
Relationships patients who achieved Thrombolysis in Myocardial Infarction
(TIMI) grade 3 coronary artery flow at 90 minutes (reproduced
from Modi et al.,[7] with permission).
Pharmacokinetic-pharmacodynamic modelling
attempts to establish quantitative relationships be-
tween drug dose, plasma concentrations, patient-
specific factors such as age, body size, gender, or- This suggests that the reduced clearance, greater
gan dysfunction or concomitant medication, and fibrin specificity and higher PAI-1 resistance de-
therapeutic effects and adverse events. The objec- signed for tenecteplase allow higher plasma con-
tives of such modelling are to compute the mean centrations and thus a more rapid restoration of
and variability of key model parameters (such as coronary patency to be attained, while providing a
clearance or clinical response) and to aid simula- reduction in serious bleeding events.
tion of new dosage regimens. Although the amount of pharmacokinetic data
As a significant step in this direction, Modi et collected during the clinical development of ten-
al.[7] showed that the percentage of patients achiev- ecteplase was considerably greater than has been
ing TIMI 3 grade coronary artery blood flow reported for other fibrinolytic agents,[1,10,62] it was
yielded a clear AUC-response relationship. It was restricted for the practical reasons explained above
found that >75% of patients achieved TIMI 3 flow to subsets of phase I and early phase II clinical
at 90 minutes after administration when the corre- trials. These comprised 70 and 18% of the TIMI
sponding partial AUC2–90 was >320 μg • min/ml 10A and TIMI 10B patients, respectively. The
(figure 7). This corresponds to an average plasma available pharmacokinetic-pharmacodynamic data
concentration of 3.6 μg/ml during the 90-minute were therefore dwarfed by the amount of dose-
time interval. Systemic exposure to tenecteplase at response data on clinical variables, such as mortal-
all times after bolus administration of 30–50mg ity and bleeding events, that were obtained without
was higher than for alteplase 100mg (figure 4) plasma concentrations in the remainder of the
without compromising clinical safety. For exam- TIMI 10A and 10B trials, the ASSENT-1 phase II
ple, AUC for tenecteplase 40mg, which is close to safety trial and the ASSENT-2 phase III ‘megatr-
the finally adopted clinical dose of 0.53 mg/kg, ial’ (see table III for citations and comparisons of
was 1.8 times higher than for alteplase 100mg, and patient numbers). Therefore, although dose-plasma
peak concentration (Cmax) was 2.2 times higher.[7] concentration-effect data sets are in general con-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
Tenecteplase 1243

siderably more informative than dose-effect data 5. Conclusions


sets without pharmacokinetics, even with intrave-
nous administration, the former were available for The fibrinolytic agent tenecteplase exemplifies
‘only’ 179 tenecteplase patients, whereas the latter the power of rational drug development applied to
therapeutic proteins. By means of a high resolution
were available for approximately 12 100 patients.
structural analysis of the physiological t-PA mol-
This situation was illustrated quite clearly dur-
ecule and combined specific mutagenesis at three
ing the development of the rationale for body-
sites, tenecteplase was developed to exhibit en-
weight-tiered administration of tenecteplase in the hanced biological properties compared with re-
ASSENT-2 phase III study and post-approval use. combinant t-PA (alteplase). These properties were
The rationale was initially derived from data ob- first extensively characterised in preclinical stud-
tained from fixed doses per patient in phase II, and ies and appeared to be confirmed in clinical trials.
could be confirmed in the ASSENT-2 study.[15-17,63] Pharmacodynamically, fibrin specificity was
The bodyweight dependence of tenecteplase clear- increased more than 10-fold and reactivity with
ance shown in figure 5 provided valuable support- PAI-1, which is present in platelet-rich blood clots,
ing data, but the final bolus dosage regimen of was decreased 80-fold relative to alteplase. Phar-
30–50mg (approximately equivalent to 0.53 mg/kg) macokinetically, plasma clearance was reduced 4-
was determined by means of bodyweight-based fold to 105 ml/min and the dominant half life
analysis of clinical variables (corrected TIMI prolonged 6-fold to 22 minutes compared with al-
frame count, TIMI flow grade, intracranial haem- teplase in patients with acute myocardial infarc-
orrhage, stroke and severe bleeding) using multi- tion, with essentially unchanged distribution char-
acteristics. This enabled tenecteplase to be
variate Cox and logistic regression.[15] It is essen-
administered as a 5–10 second intravenous bolus
tial to note that these clinical variables reflect both
dose rather than the 90-minute accelerated infu-
pharmacokinetic and pharmacodynamic variabil- sion regimen of alteplase, while providing equiv-
ity. The former has been defined by the pharmaco- alent efficacy in angiographic studies and less sys-
kinetic studies reviewed above. The latter is criti- temic fibrinogenolysis. Tenecteplase, given as a
cally important but much more difficult to quantify single bodyweight-tiered bolus dose of approxi-
because it is the variability, resulting from a given mately 0.53 mg/kg, is the only fibrinolytic agent
systemic exposure to tenecteplase in different in- that has demonstrated equivalent efficacy and im-
dividuals, both of the susceptibility of coronary proved safety (reduced frequency of major bleed-
thrombi to fibrinolysis and of the susceptibility of ing) compared with alteplase in a large mortality
the vascular system to adverse bleeding events. trial (ASSENT-2).
For example, pharmacodynamic rather than More pharmacokinetic data were obtained for
pharmacokinetic variability is much more likely to tenecteplase than for any other fibrinolytic agent
be responsible for the increased mortality and hitherto (complete concentration-time profiles for
bleeding events in the important patient group 179 patients), which provided excellent estimates
of mean and variability of pharmacokinetic param-
aged >75 years that is observed both for ten-
eters (two-compartment model). The relatively
ecteplase and for other thrombolytic agents.[9,14,16]
high interindividual variability of tenecteplase
The increase in systemic exposure to tenecteplase clearance and initial volume of distribution could,
with age that results from reduced clearance is rel- via regression analysis, be partially explained by
atively modest (figure 5), whereas it is known that the patient covariates bodyweight and age. The in-
hypertension and pathological alterations in blood crease of clearance with bodyweight aided the de-
vessels that may predispose to bleeding events are velopment of a rationale for the weight-tiered dose
more prevalent in elderly patients.[64] regimen for tenecteplase in clinical practice.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
1244 Tanswell et al.

A pharmacokinetic-pharmacodynamic relation- 12. GUSTO III Investigators. A comparison of reteplase with al-
teplase for acute myocardial infarction. N Engl J Med 1997;
ship could be established between partial AUC of 337: 1118-23
tenecteplase and coronary artery recanalisation as 13. Dunn CJ, Goa KL. Tenecteplase: a review of its pharmacology
measured by angiography. Because of its utility in and therapeutic efficacy in patients with acute myocardial
infarction. Am J Cardiovasc Drugs 2001; 1: 51-66
aiding rational dose regimen design, there is poten- 14. Van de Werf F, Barron HV, Armstrong PW, et al. Incidence and
tial for more extensive use of pharmacokinetic- predictors of bleeding events after fibrinolytic therapy with
fibrin-specific agents. Eur Heart J 2001; 22: 2253-61
pharmacodynamic modelling in future clinical tri- 15. Wang-Clow F, Fox NL, Cannon CP, et al. Determination of a
als of fibrinolytic agents and their combinations weight adjusted dose of TNK-tissue plasminogen activator.
with antiplatelet drugs. Am Heart J 2001; 141: 33-50
16. Gibson CM, Marble SJ. Issues in the assessment of the safety
and efficacy of tenecteplase (TNK-tPA). Clin Cardiol 2001;
Acknowledgements 24: 577-84
17. Angeja BG, Alexander JH, Chin R, et al. Safety of the weight-
We wish to thank Drs K. Schleenhain and G. Heusel for adjusted dosing regimen of tenecteplase in the ASSENT trial.
expert help in the preparation of the manuscript. The authors Am J Cardiol 2001; 88: 1240-5
are or were employed by Boehringer Ingelheim or Genetech, 18. Collen D, Lijnen HR. Molecular basis of fibrinolysis, as rele-
Inc. These companies sponsored the clinical trials described vant for thrombolytic therapy. Thromb Haemost 1995; 74:
in the review and also manufacture and market tenecteplase. 167-71
19. Lijnen H, Collen D. Strategies for the improvement of throm-
bolytic agents. Thromb Haemost 1991; 66: 88-110
References 20. Bennett WF, Paoni NF, Keyt BA, et al. High resolution analysis
1. Tanswell P, Tebbe U, Neuhaus KL, et al. Pharmacokinetics and of functional determinants on human tissue-type plasminogen
fibrin specificity of alteplase during accelerated infusions in activator. J Biol Chem 1991; 266: 5191-201
acute myocardial infarction. J Am Coll Cardiol 1992; 19: 21. Parekh RB, Dwek RA, Thomas JR, et al. Cell-type-specific and
1071-5 site-specific N-glycosylation of type I and type II human tis-
2. Topol E. An international, randomized trial comparing four sue plasminogen activator. Biochemistry 1989; 28: 7644-62
thrombolytic strategies for acute myocardial infarction. N 22. Noorman F, Rijken DC. Regulation of tissue-type plasminogen
Engl J Med 1993; 329: 673-82 activator concentrations by clearance via the mannose recep-
3. Sobel BE. Fibrin specificity of plasminogen activators, rebound tor and other receptors. Fibrinolysis Proteolysis 1997; 11:
generation of thrombin, and their therapeutic implications. 173-86
Coron Artery Dis 2001; 12: 232-332
23. McCluskey ER, Keyt BA, Refino CJ, et al. Biochemistry, phar-
4. Collen D, De Bono DP, Lijnen HR, et al. Development of novel
macology and initial clinical experience with TNK-tPA. In:
thrombolytic agents: mini-symposium on the role of the fibri-
Sasahara AA, Loscalzo J, editors. New therapeutic agents in
nolytic system in the pathophysiology and treatment of throm-
thrombosis and thrombolysis. New York: Marcel Dekker,
bosis; 1993 Oct; Leuven. J Intern Med 1994; 236: 433-7
Inc., 1997: 475-93
5. Huber K. Plasminogen activator inhibitor type-I (part two): role
24. Benedict CR, Refino CJ, Keyt BA, et al. New variant of human
for failure of thrombolytic therapy. PAI-1 resistance as a po-
tissue plasminogen activator (TPA) with enhanced efficacy
tential benefit for new fibrinolytic agents. J Thrombosis
Thrombolysis 2001; 11: 195-202 and lower incidence of bleeding compared with recombinant
human TPA. Circulation 1995; 92: 3032-40
6. Keyt B, Paoni N, Refino C, et al. A faster-acting and more
potent form of tissue plasminogen activator. Proc Natl Acad 25. Eppler S, Senn T, Gilkerson E, et al. Pharmacokinetics and
Sci U S A 1994; 91 (1): 3670-4 pharmacodynamics of recombinant tissue-type plasminogen
7. Modi NB, Fox NL, Fong WC, et al. Pharmacokinetics and phar- activator following intravenous administration in rabbits;
macodynamics of tenecteplase: results from a phase II study comparison of three dosing regimens. Biopharm Drug Dispos
in patients with acute myocardial infarction. J Clin Pharmacol 1998; 19: 31-8
2000; 40: 508-15 26. Modi NB, Eppler S, Breed J, et al. Pharmacokinetics of a slower
8. Stewart RJ, Fredenburgh JC, Leslie BA, et al. Identification of clearing tissue plasminogen activator variant, TNK-tPA, in
the mechanism responsible for the increased fibrin specificity patients with acute myocardial infarction. Thromb Haemost
of TNK-tissue plasminogen activator relative to tissue plas- 1998; 79: 134-9
minogen activator. J Biol Chem 2000; 275: 10112-20 27. Mohler MA, Refino CJ, Chen SA, et al. D-Phe-Pro-Arg-chloro-
9. Van de Werf F, Adgey J, Ardissino D, et al. Single bolus ten- methyl ketone: its potential use in inhibiting the formation of
ecteplase compared with front-loaded alteplase in acute myo- in vitro artifacts in blood collected during tissue-type plasmin-
cardial infarction: the ASSENT 2 double-blind randomised ogen activator thrombolytic therapy. Thromb Haemost 1986;
trial. Lancet 1999; 354 (9180): 716-22 56: 160-4
10. Martin U, Kaufmann B, Neugebauer G. Current clinical use of 28. Seifried E, Tanswell P. Comparison of specific antibody, D-
reteplase for thrombolysis: a pharmacokinetic-pharmacody- Phe-Pro-Arg-CH2Cl and aprotinin for prevention of in vitro
namic perspective. Clin Pharmacokinet 1999; 36: 265-76 effects of recombinant tissue-type plasminogen activator on
11. Kohnert U, Rudolph R, Verheijen JH, et al. Biochemical prop- haemostasis parameters. Thromb Haemost 1987; 58: 921-6
erties of the kringle-2 and protease domains are maintained in 29. Reed BR, Chen AB, Tanswell P, et al. Low incidence of anti-
the refolded t-PA deletion variant BM 06.022. Protein Eng bodies to recombinant human tissue-type plasminogen acti-
1992; 5: 95-100 vator in treated patients. Thromb Haemost 1990; 64: 276-80

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)
Tenecteplase 1245

30. Gibaldi M, Perrier D, editors. Pharmacokinetics. 2nd ed. New 49. Sobel BE, Gross RW, Robison AK. Thrombolysis, clot selec-
York: Marcel Dekker, 1982 tivity, and kinetics. Circulation 1984; 70: 160-4
31. Ette EL, Ludden TM. Population pharmacokinetic modeling: 50. Thomaseth K, Boniollo B. Simulation of plasmatic enzymatic
the importance of informative graphics. Pharm Res 1995; 12: reactions during thrombolytic therapy with recombinant tis-
1845-55 sue-type plasminogen activator: from in vitro knowledge to
32. DeGuzman G, Richarson L, Berleau L, et al. Hepatic uptake and new assumptions in vivo. Simulation 1996; 66: 219-28
processing of TNK-tPA [abstract]. Pharm Res 1995; 123: 332 51. Sharma A, Jusko WJ. Characteristics of indirect pharmacody-
33. Tanswell P, Stassen JM, Platz S. Regulatory submission to namic models and application to clinical drug responses. Br
EMEA. Metalyse® (tenecteplase): expert report on the tox- J Clin Pharmacol 1998; 48: 229-39
ico-pharmacological documentation. Biberach: Boehringer 52. Hoylaerts M, Rijken DC, Lijnen HR, et al. Kinetics of the acti-
Ingelheim, 1999. (Data on file) vation of plasminogen by human plasminogen activator: role
34. Keyt BA, Berleau LT, Nguyen HV, et al. Radioiodination of of fibrin. J Biol Chem 1982; 257: 2912-9
the active site of tissue plasminogen activator: a method for 53. Lee LV, Ewald GA, McKenzie CR, et al. The relationship of
radiolabeling serine proteases with tyrosylprolyarginyl soluble fibrin and cross-linked fibrin degradation products to
chloromethyl ketone. Anal Biochem 1992; 206: 73-83 the clinical course of myocardial infarction. Arterioscler
35. Bakhit C, Lewis D, Busch U, et al. Biodisposition and catabo- Thromb Vasc Biol 1997; 17: 628-33
lism of tissue-type plasminogen activator in rats and rabbits. 54. Seifried E, Tanswell P, Rijken DC, et al. Fibrin degradation
Fibrinolysis 1988; 2: 31-6 products are not specific markers for thrombolysis in myo-
36. Lucore CL, Sobel BE. Interactions of tissue-type plasminogen cardial infarction. Lancet 1987; II: 333-4
activator with plasma inhibitors and their pharmacologic im- 55. Walker JB, Nesheim ME. The molecular weights, mass distri-
plications. Circulation 1988; 77: 660-9 bution, chain composition, and structure of soluble fibrin deg-
37. Tanswell P, Seifried E, Stang E, et al. Pharmacokinetics and radation products released from a fibrin clot perfused with
hepatic catabolism of tissue-type plasminogen activator. plasmin. J Biol Chem 1999; 274: 5201-12
Arzneimittel Forschung 1991; 41: 1310-9 56. Binbrek A, Rao N, Absher M, et al. The relative rapidity of
38. Krause J. Hepatic catabolism of tissue-type plasminogen acti- recanalization induced by recombinant tissue-type plasmino-
vator (t-PA), its variants, mutants and hybrids. Fibrinolysis gen activator (r-tPA) and TNK-tPA, assessed with enzymatic
1988; 2: 133-42 methods. Coron Artery Dis 2000; 11 (5): 429-35
39. Bu G, Williams S, Strickland DK, et al. Low density lipoprotein 57. Ohman EM, Armstrong PW, White HD, et al. GUSTO-III In-
receptor-related protein/alpha(2)-macroglobulin receptor is vestigators. Risk stratification with a point-of-care cardiac
an hepatic receptor for tissue-type plasminogen activator. troponin T test in acute myocardial infarction. Am J Cardiol
Proc Natl Acad Sci U S A 1992; 89: 7427-31 1999; 84: 1281-6
40. Smedsrod B, Einarsson M. Clearance of tissue plasminogen 58. Antman EM, Tanasijevic MJ, Thompson B, et al. Cardiac spe-
activator by mannose and galactose receptors in the liver. cific troponin I levels to predict the risk of mortality in pa-
Thromb Haemost 1990; 63: 60-6 tients with acute coronary syndromes. N Engl J Med 1996;
41. Tanswell P, Heinzel G, Greischel A, et al. Nonlinear pharma- 335: 1342-9
cokinetics of tissue-type plasminogen activator in three ani- 59. The TIMI study group. The thrombolysis in myocardial infarc-
mal species and isolated perfused rat liver. J Pharmacol Exp tion (TIMI) trial. N Engl J Med 1985; 312: 932-6
Ther 1990; 255: 318-24 60. Gibson CM, Cannon CP, Daley WL, et al. TIMI Frame Count:
42. Cannon CP, Gibson CM, McCabe CH, et al. TIMI 10B Inves- a quantitative method of assessing coronary artery flow. Cir-
tigators. TNK tissue plasminogen activator compared with culation 1996; 93: 879-88
front-loaded alteplase in acute myocardial infarction: results 61. Mehta SR, Eikeboom JW, Yusuf S. Risk of intracranial haem-
of the TIMI 10B trial. Circulation 1998; 98: 2805-14 orrhage with bolus vs infusion thrombolytic therapy: a meta-
43. Van de Werf F, Cannon CP, Luyten A, et al. Safety assessment analysis. Lancet 2000; 356: 449-54
of single-bolus administration of TNK tissue-plasminogen 62. Liao WC, Beierle FA, Stouffer BC, et al. Single bolus regimen
activator in acute myocardial infarction: the ASSENT-1 trial. of lanoteplase (nPA) in acute myocardial infarction: pharma-
The ASSENT-1 Investigators. Am Heart J 1999; 137: 786-91 cokinetic evaluation from the InTIME-1 study. Circulation
44. Tanswell P, Seifried E, Su CAPF, et al. Pharmacokinetics and 1997; 96 Suppl. I: 1260-1
systemic effects of tissue-type plasminogen activator in nor- 63. Gibson CM, Cannon CP, Murphy SA, et al. Weight-adjusted
mal subjects. Clin Pharmacol Ther 1989; 46: 155-62 dosing of TNK-tissue plasminogen activator and its relation
45. Cannon CP, McCabe CH, Gibson CM, et al. TNK-tissue plas- to angiographic outcomes in the thrombolysis in myocardial
minogen activator in acute myocardial infarction: results of infarction 10 B trial. Am J Cardiol 1999; 84: 976-80
the Thrombolysis In Myocardial Infarction (TIMI) 10A dose- 64. Hammann GF, Okada Y, del Zoppo GJ. Hemorrhagic transfor-
ranging trial. Circulation 1997; 95: 351-6 mation and microvascular integrity during focal cerebral isch-
46. Van de Werf F, ASSENT-3 investigators. Efficacy and safety emia/reperfusion. J Cereb Blood Flow Metab 1996; 16:
of tenecteplase in combination with enoxaparin, abciximab, 1373-8
or unfractionated heparin: the ASSENT–3 randomised trial
in acute myocardial infarction. Lancet 2001; 358: 605-13
47. Sulikowski T, Patson PA. The inhibition of TNK-tPA by C1-
inhibitor. Blood Coagul Fibrinolysis 2001; 12: 75-7 Correspondence and offprints: Dr Paul Tanswell, Depart-
48. Van Griensven JMT, Koster RW, Burggraaf J, et al. Effects of
ment of Pharmacokinetics and Metabolism, Boehringer
liver blood flow on the pharmacokinetics of tissue-type plas-
minogen activator (alteplase) during thrombolysis in patients Ingelheim Pharma KG, Birkendorfer Strasse 65, 88397
with acute myocardial infarction. Clin Pharmacol Ther 1998; Biberach, Germany.
63: 39-47 E-mail: paul.tanswell@bc.boehringer-ingelheim.com

© Adis International Limited. All rights reserved. Clin Pharmacokinet 2002; 41 (15)

You might also like