+ All Categories
Home > Documents > 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered...

98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered...

Date post: 05-Oct-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
38
Product Monograph
Transcript
Page 1: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Product Monograph

Page 2: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

INTRODUCTION

The availability of RETAVASE® (Reteplase) marks an important advance in thethrombolytic treatment of acute myocardial infarction (AMI). RETAVASE possesses a unique molecular design and provides demonstrated reduction of mortality, adocumented safety profile, and has been angiographically proven to achieve high rates of complete perfusion and patency. The correlation between patency and patientoutcome has not been established. In addition, RETAVASE offers the convenience of asimple double-bolus dosing regimen.

This monograph is provided as a comprehensive informational reference toRETAVASE. It includes a complete review of preclinical and clinical pharmacologyfindings as well as clinical efficacy and safety data. The information presented has been designed to aid in the evaluation of RETAVASE for use in clinical practice.

RETAVASE is manufactured and distributed by Centocor, Inc.

3

Page 3: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

CONTENTS

I. Brief overview of RETAVASE® (Reteplase):a novel thrombolytic agent . . . . . . . . . . . . . . . . . . . . . 7

II. Thrombolysis in the management of acute myocardial infarction. . . . . . . . . . . . . . . . . . . . . . 9

III. The open-artery hypothesis in thrombolysis . . . . . . . . . 13

IV. RETAVASE preclinical pharmacology . . . . . . . . . . . . . . . 17

V. RETAVASE clinical pharmacology. . . . . . . . . . . . . . . . . . 23

VI. RETAVASE clinical efficacy and activity . . . . . . . . . . . . . 27

VII. RETAVASE safety profile . . . . . . . . . . . . . . . . . . . . . . . . . 35

VIII. RETAVASE angiographic trials . . . . . . . . . . . . . . . . . . . . 37

IX. Summary of benefits of RETAVASE . . . . . . . . . . . . . . . . 41

X. References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43

RETAVASE full prescribing information. . . . . . . . . . . . . 45

5

Please see full prescribing information on the last pages of this monograph.

Page 4: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

R ETAVASE is a modification of the tissue-type plasminogen activator (t-PA) proteinthat maintains the kringle-2 and protease

domains of t-PA, but lacks its kringle-1, finger, andgrowth-factor domains. In contrast to t-PA, theRETAVASE molecule is nonglycosylated. Thesemodifications result in a molecule with a longerhalf-life (13 to 16 minutes). RETAVASE lacks thehigh-affinity fibrin binding characteristic of t-PA,and is produced by recombinant DNA technologyin Escherichia coli for use as a thrombolytic agent in the treatment of AMI. RETAVASE is a single-chain molecule consisting of 355 amino acids and can be converted to the two-chain form during fibrinolysis.

RETAVASE represents an important addition to the therapeutic options to treat patients withAMI. RETAVASE provides demonstrated reductionof mortality, a documented safety profile, andproven preservation of left ventricular function.Additionally, RETAVASE has been angiographicallyproven to achieve high rates of complete perfusionand patency, and offers a fast, convenient double-bolus dosing regimen. The correlation betweenpatency and patient outcome has not beenestablished.

Pursuing the ideal thrombolytic agentIt was observed in 1992 that none of the

thrombolytic agents then available could beconsidered ideal. Each had limitations. Therefore,it is appropriate to define the characteristics of an ideal thrombolytic drug to help guide thedirection of future research and development.1

Patient survival is the ultimate goal of thrombolytictherapy. An agent should produce rapidrecanalization in patients with acute coronary

thrombosis. Additional characteristics of an idealagent are listed in Table 1.

It is noteworthy that RETAVASE possesses some of the attributes of the ideal agent:administration as an intravenous bolus, relativespecificity for recent thrombi, and restoration ofcomplete flow in a high percentage of patients.RETAVASE also has no effect on circulatoryhemodynamics, is not antigenic, and has no known interactions with adjunctive therapies.

SECTION IBrief overview ofRETAVASE® (Reteplase): a novel thrombolytic agent

Table 1.—Selected Characteristics ofan Ideal Thrombolytic Agent

• Rapid recanalization after administration• Approaches 100% efficacy for recanalization• Can be given as rapid IV bolus• Specific for recent thrombi• Assists in preventing reocclusion

– Should result in sustained patency within the first 24 hours

– Appropriate half-life if fibrin-specific• Targets thrombus induced by plaque rupture• No effect on circulatory hemodynamics• No negative interactions with adjunctive therapies• No antigenicity• No significant side effects

– Adapted from Rapaport.1

7

Page 5: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Pharmacokinetic propertiesDeletion of the three N-terminal domains and

the absence of glycosylation present in native t-PAimproves the pharmacokinetic properties ofRETAVASE® (Reteplase) without sacrificing itseffectiveness as a plasminogen activator. Thesemodifications to the molecule result in a plasmahalf-life of 13 to 16 minutes, which represents anincrease over the 3- to 6-minute half-life of native t-PA.2 The longer half-life of RETAVASE permits fast,convenient double-bolus intravenous dosing, withthe second dose given 30 minutes after the first.

AdministrationThe ease of administration afforded by bolus

dosing may permit an earlier start of life-savingtherapy in the hospital emergency department orintensive care unit. Bolus dosing may also eliminatethe need for two intravenous lines duringcoadministration with heparin, since the heparininfusion can be interrupted during the injection ofRETAVASE. The intravenous line should be flushedbefore and after administration of RETAVASE.

RETAVASE is supplied in glass vials as a sterile,preservative-free, lyophilized powder. The contentsof the vial are reconstituted with Sterile Water forInjection, USP (without preservatives), andadministered as an intravenous injection (10 U)over 2 minutes. The second bolus (10 U) is given 30minutes after initiation of the first bolus injection.The dose of RETAVASE need not be adjusted forpatient weight.

Clinical studiesThe clinical benefits and safety profile of

RETAVASE have been documented in well-controlled clinical trials in patients with AMI.The International Joint Efficacy Comparison ofThrombolytics (INJECT) trial was a randomized,double-blind, multicenter comparison ofRETAVASE with Streptokinase in 6,010 AMIpatients that was conducted in Europe.3 The study was powered to confirm that RETAVASE was safe and effective in reducing mortality in AMI patients.

Results demonstrated that RETAVASE iseffective in reducing mortality in patients withAMI. Patients treated with RETAVASE had asignificantly lower incidence of atrial fibrillation,asystole, cardiogenic shock, congestive heart failure(CHF), hypotension, and allergic reactions. Overallbleeding and stroke rates were similar for bothagents. There was an increase in the incidence of in-hospital intracranial hemorrhage in patientstreated with RETAVASE (0.77%) compared to thosein the Streptokinase group (0.37%, P=0.04). TheINJECT investigators concluded that RETAVASE is an effective agent for the treatment of AMI and has an acceptable safety profile.

In the Reteplase Angiographic Phase IIInternational Dose-finding (RAPID 1) Study,infarct-related arteries were evaluatedangiographically in 606 patients after therapy withRETAVASE administered by bolus injection (threedifferent regimens) or Alteplase administered as a3-hour infusion.4 In the Reteplase vs AlteplasePatency Investigation During Acute MyocardialInfarction (RAPID 2) Study, 324 patients receivedeither RETAVASE or an accelerated-dose (90-minute) Alteplase infusion.5 Patients in both trialsalso received aspirin and heparin. In bothangiographic trials, RETAVASE achieved high rates of complete perfusion and patency.4,5 Thecorrelation between patency and patient outcomehas not been established. These studies alsodemonstrated the efficacy of RETAVASE inpreserving left ventricular function following AMI.

Low risk of antigenicityRETAVASE is synthesized in transformed

E coli and is a modified endogenous protein with a low risk for antigenic activity. Antibodies to RETAVASE were not observed in any ofapproximately 2,400 patients tested for antibody formation.

The low risk of antigenicity with RETAVASEcontrasts with that of Streptokinase and itsderivatives. Since most patients have previouslybeen exposed to hemolytic streptococcal infection,they may produce antibodies to Streptokinase, aprotein derived from Lancefield group C strains ofß-hemolytic streptococci.

8

Page 6: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Rationale for use of thrombolysis

C ardiovascular disease accounts for a largeproportion of all deaths in both men andwomen in the United States. The age-

adjusted death rate in 1991 was 186 per 100,000individuals; for coronary heart disease, it was 99.1per 100,000.6 Patients who survive the acute stage ofa myocardial infarction are at two to nine timesgreater risk of subsequent cardiovascular morbidityand mortality than the general population.6

Three perfusion methods are currently availablefor the management of patients with AMI:thrombolysis with pharmacologic agents,percutaneous transluminal coronary angioplasty(PTCA), and revascularization by coronary arterybypass grafting (CABG). The effectiveness ofthrombolytic therapy in reducing mortality inpatients with AMI has been demonstrated in large,multinational clinical trials. PTCA is an invasiveprocedure that requires a catheterization laboratoryand expert teams for implementation. PTCA is an option preferred by some physicians wherefacilities are available, especially in patients withcontraindications to thrombolytic therapy or withinfarction complicated by cardiogenic shock.

The era of thrombolysisThe nature of the infarction process was not

well understood until comparatively recent years. In1980, DeWood and colleagues provided definitiveproof that thrombosis occurs early in the infarctionprocess.7 These investigators angiographicallyvisualized total occlusion of the infarct-relatedcoronary artery within 4 hours of onset ofsymptoms in 110 of 126 patients (87%) with acutetransmural (Q-wave) infarction. In addition, theyfound that thrombi were present in 88% of patientsundergoing CABG. Since then, it has frequentlybeen observed that the arterial lumen isconsiderably narrowed by dynamically changingatheromatous plaque, which enables relatively smallthrombi to block antegrade flow and produceischemia and hypoxia. More than 85% ofmyocardial infarctions result from formation of anacute thrombus obstructing an atherosclerotic,stenotic coronary artery.8 Thus, the development ofselective coronary angiography proved to be a vitallink in the development and acceptance ofthrombolytic therapy.

SECTION II

Thrombolysis in the management of acute myocardial infarction

9

Page 7: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Mechanisms of thrombolysisLike native tissue plasminogen activators, all

thrombolytic agents activate plasminogen intoplasmin, which splits the fibrin in the clot intosoluble fibrin degradation products; fibrinogen, aprecursor of fibrin, is also degraded by plasmin.Plasminogen, which has high affinity for fibrin, isactivated much more efficiently when bound thanunbound because fibrin forms surface receptors thatallow plasminogen and plasminogen activator tocome into optimal contact (Fig 1). Thrombolyticagents differ in their affinity for fibrin; this isimportant because specificity for fibrin may result inlysis of hemostatic plugs not only in coronarythrombi but also elsewhere in the body, thuspromoting bleeding.

Early history of thrombolytic therapyThe discovery in 1933 by Tillett and Garner that

the streptococcal protein Streptokinase lyses bloodclots did not translate into a tool for the cardiologistuntil decades later.9 The first clinical indications forStreptokinase were management of proximal deep-vein thrombosis and severe pulmonary embolism.The earliest attempt to treat AMI with coronaryfibrinolysis was a pilot study in 1959 by Fletcher andcolleagues, who infused Streptokinase intravenouslyfor 30 hours, starting 6 to 72 hours after onset of

symptoms.10 However, Streptokinase available at thetime was pyrogenic, and treatment was sometimesdelayed too long to salvage myocardium. Efficacycould not be adequately documented by coronaryarteriography, left ventricular function (LVF) couldnot be assessed with noninvasive modalities, andeven the correlation between infarct size andprognosis was not apparent.

Despite these obstacles, several investigatorspersisted with Streptokinase thrombolysis in the1960s and 1970s. By 1983, Schröder and colleagueshad shown that intravenous administration ofStreptokinase could restore coronary blood flow in patients with AMI.11

Early (exogenous) thrombolytic agentsStreptokinase. Rather than enzymatically

cleaving plasminogen (the mechanism of action of t-PA, Alteplase, and RETAVASE® (Reteplase),Streptokinase forms a complex that produces aconformational change in the plasminogenmolecule. The plasminogen complex then convertsnoncomplexed plasminogen molecules to plasmin.The half-life of Streptokinase is approximately 30minutes, with much of the circulating Streptokinasecatalyzed before it can bind to polymerized fibrin.12

10

Fig 1.—Binding of plasminogen to endogenous tissue plasminogen activator (t-PA)and fibrin.

Fibrinolysis

Fibrin Fibrin

Plasmin

t-PA

t-PA

Plasminogen

Page 8: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Streptokinase indiscriminately binds to bothcirculating and thrombus-associated plasminogen;this may result in plasminemia.13 While some excessplasmin is inactivated by a2-antiplasmin, the restremains active. Thus, Streptokinase generates asystemic lytic state by depleting concentrations ofcirculating fibrinogen, plasminogen, procoagulantproteins, and a2-antiplasmin while inducing highplasma concentrations of fibrin degradationproducts.

Equilibrium is normally reached between freeand bound plasminogen. By converting circulatingplasminogen into plasmin, Streptokinase maycreate a systemic plasminogen deficit that drawsfibrin-associated plasminogen out of the clot.13

The hypothesis is that this “plasminogen steal”may attenuate plasmin formation at the site ofthe thrombus and hinder the reopening ofobstructed coronary arteries.14 The clinicalsignificance of plasminogen steal has not been established.

Streptokinase use is associated with side effects, allergic reactions, and circulatingantistreptokinase antibodies that have beenassociated with patient reactions with repeatadministration of this agent.15,16

Anistreplase. Anistreplase is an anisoylatedderivative complex prepared from plasminogen andpurified Streptokinase.12 The anisoyl group blocksactivity until released by acylation in aqueoussolution (a first-order nonenzymatic reaction), afterwhich the complex forms. Deacylated Anistreplasecleaves plasminogen with greater catalytic efficiencywithin thrombi than in the circulation. Anistreplaseis indicated for management of AMI. Afterreconstitution with sterile water, it is administeredas a single intravenous dose of 30 mg (eitherdirectly into a vein or through an intravenous line)over 2 to 5 minutes. Antibody formation against thestreptococcal protein may occur with Anistreplase.

Endogenous thrombolytic agents andsynthetic analogues

Alteplase. Alteplase, a mixture of one-chainand two-chain t-PA, is produced by means ofrecombinant technology. Alteplase is activated inthe presence of fibrin. Therefore, it may elicit littleplasminogen from the clot to replenish plasmalevels. Moreover, plasmin formed in the thrombusmay be less accessible to inhibitors such as a2-antiplasmin. Alteplase reduces plasmafibrinogen levels only slightly. It was demonstratedangiographically and clinically in the GlobalUtilization of Streptokinase and TissuePlasminogen Activator for Occluded CoronaryArteries (GUSTO-I) study that Alteplase recanalizesarteries more frequently than Streptokinase.17,18

Effective thrombolysis requires that Alteplasedoses result in plasma levels of unbound drugcapable of saturating the endogenous inhibitors.Alteplase is cleared rapidly by the liver, resulting in a half-life of 3 to 6 minutes.2 The rapid clearance of Alteplase from plasma is mediated by two domains: the epidermal growth factor and kringle-1 domains. Binding of these domainsto liver receptors promotes elimination of theagent.19,20 Therefore, infusion is usually utilized to sustain therapeutic plasma concentrations of Alteplase.

11

Page 9: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Other issues and considerations forcurrent thrombolytic therapy

Adjunctive anticoagulation. The activity ofthrombolytic agents can be potentiated by theconcurrent administration of aspirin for itsantiplatelet effect and heparin for its anticoagulanteffect. Aspirin has been recommended forcoadministration with thrombolytic therapy sincethe Second International Study of Infarct Survival(ISIS-2), which showed benefit for aspirin whenadministered alone, as well as concurrently with Streptokinase.21 Without heparin, ongoingcoagulation may outpace thrombolysis and result in a lower patency rate.

Heparin in conventional doses has a narrowtherapeutic range. Whether used as the soleantithrombotic agent, the initiating agent, or anadjunctive anticoagulant, it is associated with a risk of serious bleeding in 1% to 8% of patients.22

This risk is compounded by coadministration ofantiplatelet agents.

The Heparin-Aspirin Reperfusion Trial(HART), which compared early intravenousheparin with low-dose oral aspirin as adjunctivetreatment with recombinant tissue plasminogenactivator during AMI, found that t-PA and heparintreatment resulted in a higher rate of arterialpatency (82%) after 7 to 24 hours than t-PA andaspirin treatment (52%). These study resultssuggest that early systemic heparin treatmentshould be administered during thrombolytictherapy with t-PA.23

The addition of intravenous heparin toStreptokinase in GUSTO-I produced results nobetter than those with subcutaneous heparin.In combination with either intravenous orsubcutaneous heparin, Streptokinase produced a lower incidence of stroke without decreasingmortality compared to accelerated-dose Alteplasedosing with intravenous heparin.17 Aspirin, on theother hand, is a valuable addition to Streptokinase,as seen in ISIS-2.21 In ISIS-3, the addition ofheparin to aspirin was associated with an excess oftransfused or other major noncerebral bleeds andof definite or probable cerebral hemorrhage.24

Cost-effectiveness of thrombolysis. Anyanalysis of the cost-effectiveness of thrombolysismust consider not only the price of the agent usedbut also the total cost of care and the gain insurvival. Aggressive intervention, even with themost expensive thrombolytic agents, may reducecosts in the long run by lessening both short- andlong-term risks of CHF and other cardiaccomplications.

12

Page 10: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Early reperfusion and reduction in mortality

A ccording to the open-artery hypothesis,reperfusion of ischemic areas as earlyas possible after the appearance of

symptoms of AMI (ie, within 3 hours) is crucial for limiting infarct size, preserving left ventricularfunction, and improving the odds of survival.25

The rationale for rapid reopening is based onanimal experiments: a smaller area of infarction was observed in animals in which transientlyinduced coronary occlusion was relieved by early reperfusion than in animals subjected topermanent occlusion.26

As a corollary to the open-artery hypothesis,early thrombolysis (with adjunctive treatment suchas aspirin, anticoagulants, or coronary angioplasty)may recanalize obstructed coronary arteries andsalvage jeopardized myocardium, while reducingprogression to chronic CHF.

The open-artery hypothesis suggests that theuse of thrombolytic agents that reopen occludedcoronary arteries earlier and more completely inpatients with AMI may translate into lowermortality. However, the correlation betweenpatency and patient outcome has not beenestablished.

Early clinical trialsTIMI-1. The Thrombolysis in Myocardial

Infarction (TIMI-1) Study 27 in 1985 was the firsttrial to demonstrate that Alteplase is more effectivethan Streptokinase in reopening occluded coronaryarteries. In this randomized trial, 90-minuteangiography in 290 patients demonstrated thatAlteplase achieved significantly higher rates of

patency and complete perfusion thanStreptokinase.27

ECSG-1. The European Cooperative StudyGroup (ECSG-1) observed a patency rate of 70% in64 patients randomized to Alteplase and 55% in 65patients randomized to Streptokinase (P=0.054).28

These data suggested that perfusion achieved lateafter the onset of the symptoms of AMI may notpreserve ischemic myocardium, since prolongedischemia damages not only the muscle but also theassociated microvasculature.

Large-scale trialsGISSI-1. The Gruppo Italiano per lo Studio

della Streptochinasi nell’Infarto Miocardico (GISSI-1) was a milestone study that established the value of thrombolytic therapy for patients withAMI. GISSI-1 was the first large-scale (N=11,806)study to demonstrate a significant reduction inmortality achieved with Streptokinase thrombolysiscompared to placebo.29

ISIS-2. In ISIS-2, another large-scale(N=17,187) trial, a combination regimen ofStreptokinase and aspirin was shown to reducecumulative vascular mortality (deaths from cardiac,cerebral, hemorrhagic, or other known vasculardisease) in the first 35 days more effectively thaneither aspirin or Streptokinase monotherapy orplacebo.21

SECTION III

The open-artery hypothesis in thrombolysis

13

Page 11: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Clinical megatrials: implications for theopen-artery hypothesis

GISSI-2. The Gruppo Italiano per lo Studio dellaSopravvivenza nell’Infarto Miocardico (GISSI-2)study was the first large-scale trial to assess thecomparative effects of Streptokinase 1.5 MU infusedintravenously over 30 to 60 minutes and Alteplase

100 mg infused intravenously over 3 hours.30 In thismulticenter, randomized, open trial in 12,490patients with AMI admitted to coronary care unitswithin 6 hours from onset of symptoms, half thepatients were also randomized to receive 12,500 Uof heparin subcutaneously twice daily, starting 12hours after beginning thrombolytic therapy.

14

Fig 2.—Cumulative mortality (%) in days 0 to 35 in ISIS-3 and GISSI-2.

– Adapted from ISIS-3.24

Death in ISIS-3SK: 1,455/13,780 (10.6%)APSAC: 1,448/13,773 (10.5%)

SK, APSAC

P=NS% %

(A)

0

0 7 14 21Days from Randomization Days from Randomization

28 35

5

10

Death in ISIS-3SK: 1,455/13,780 (10.6%)rt-PA: 1,418/13,746 (10.3%)

SK

P=NS

(B)

0

0 7 14 21 28 35

5

10 Duteplase

Alteplase

SK = Streptokinase; APSAC = Anistreplase; rt-PA = Alteplase/Duteplase.

Death in GISSI-2 SK: 958/10,396 (9.2%) rt-PA: 993/10,372 (9.6%)

SK

P=NS% %

(C)

0

0 7 14 21Days from Randomization Days from Randomization

28 35

5

10

Death in ISIS-3 and GISSI-2SK: 2,413/24,176 (10.0%)rt-PA: 2,411/24,118 (10.0%)

SK, rt-PA

P=NS

(D)

0

0 7 14 21 28 35

5

10

(A) All patients allocated SK vs all allocated APSAC in ISIS-3 only; (B, C, D) all patients allocated SK vs all allocated rt-PA in (B) ISIS-3; (C) GISSI-2; (D) ISIS-3 and GISSI-2 combined.

Page 12: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Atenolol (45.3%) and aspirin (87%) wereadministered to patients without contraindications.There was no significant difference in overallmortality in the Alteplase-treated patients (9%) andthe Streptokinase group (8.6%). There was also nosignificant difference in the combined end point ofdeath plus clinical CHF or extensive left ventriculardamage in the absence of CHF.30

International Study Group. The InternationalStudy Group combined mortality data from the12,490 patients in the GISSI-2 trial30 with data from8,401 patients recruited in other countries. Again,no significant differences in hospital mortality werefound between Alteplase (8.9%) or Streptokinase(8.5%) or between subcutaneous heparin (8.5%)and no heparin (8.9%) treatment.31

ISIS-3. In the Third International Study ofInfarct Survival (ISIS-3), 41,299 patients entering914 hospitals up to 24 hours after the onset ofsuspected AMI were randomized to Streptokinase,1.5 MU infused over 1 hour; Duteplase (a double-chain t-PA), 0.04 MU/kg bolus over 1 minute,0.36 MU/kg during the remainder of the first hour,and 0.067 MU/kg/h over the next 3 hours; orAnistreplase (30 U over 3 minutes).24 All patientsreceived aspirin, and half also received 12,500 IU of calcium heparin, starting 4 hours afterrandomization and given subcutaneously twice daily for 7 days. Cumulative deaths during days 0 to 35 are shown in Fig 2. There were no significant differences in mortality between any two groups (10.6% Streptokinase, 10.5%Anistreplase, 10.3% Duteplase). There were also nosignificant differences in 35-day mortality betweenthe aspirin only (10.6%) and aspirin plus heparin(10.3%) treated patients.24

GUSTO-I. The GUSTO-I study,17 the largestmegatrial comparing the effect of Alteplase andStreptokinase on mortality (41,021 patients withconfirmed AMI treated at 1,081 hospitals in 15countries), differed in several important respectsfrom the ISIS and GISSI trials.

In the GUSTO-I study, the accelerated-dose or“front-loaded” Alteplase regimen was given as aninitial bolus, followed by a 90-minute infusion,with two thirds of the dose given in the first 30minutes. This dosing regimen was designed toimprove perfusion rates. In another departure from previous megatrials, heparin was given bybolus followed by intravenous infusion, rather than subcutaneously, and was continued for at least48 hours to limit fibrin formation that mightproduce reocclusion. Similar to GISSI-2, patients in GUSTO-I were admitted within 6 hours after the onset of symptoms. In ISIS-2 and ISIS-3,however, patients were included up to 24 hours after onset of symptoms.

Patients in the GUSTO-I trial were randomizedto one of four treatment regimens:

• Alteplase (bolus dose of 15 mg, followed by aninfusion of 0.75 mg/kg over 30 minutes, not toexceed 50 mg, and 0.5 mg/kg, up to 35 mg,over the next 60 minutes) plus intravenousheparin

• Streptokinase and intravenous heparin• Streptokinase and subcutaneous heparin• Both thrombolytics together plus intravenous

heparinThe primary clinical end point of 30-day

mortality was 6.3% in patients receiving accelerated-dose Alteplase plus intravenous heparin, comparedto 7.2% and 7.4% for the two Streptokinase groups(Table 2 and Fig 3).17 There was a significantreduction in mortality with accelerated-doseAlteplase compared with the two Streptokinaseregimens (10 lives saved per 1,000 patients treated;risk reduction, 14%). The GUSTO-I trial adds to a growing body of evidence in support of theopen-artery hypothesis.

In the GUSTO-I Angiographic Investigatorssubstudy, 2,431 of the patients randomized to thefour treatment regimens were assigned to undergocardiac angiography at 90 minutes, 180 minutes,24 hours, or 5 to 7 days after the initiation ofthrombolytic therapy; angiography was repeatedafter 5 to 7 days in the 90-minute group.18 The rate

15

Page 13: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

of patency of the infarct-related artery at 90 minuteswas highest in the Alteplase plus heparin group(81%), compared with the Streptokinase andsubcutaneous heparin group (54%, P<0.001), theStreptokinase and intravenous heparin group (60%,P<0.001), and the combination therapy group(73%, P=0.032). By 180 minutes, the four groupshad similar patency rates and reocclusion rates. Leftventricular function, which paralleled patency rate at90 minutes, was best in the Alteplase group and inpatients with normal flow, regardless of treatment.Mortality at 30 days was lowest (4.4%) among

patients with complete coronary flow at 90 minutesand highest (8.9%) among patients with no flow in the infarct-related artery (P=0.009). Theinvestigators suggested that the mechanism by which accelerated-dose Alteplase therapy produced a superior outcome in the GUSTO-I trial was itsmore complete restoration of flow through theinfarct-related artery than Streptokinase, which inturn resulted in improved ventricular performanceand lower mortality due to AMI.18

16

Fig 3.—Mortality (30 days) in the four treatment groups of GUSTO-I.

– Adapted from GUSTO Investigators.17

Streptokinase and IV heparinStreptokinase and SC heparinAlteplase and StreptokinaseAccelerated-dose Alteplase

Days After Randomization

Mortality(%)

00123456789

10

2 4 6 8 10 12 14 16 18 20 22 24 26 28 30

Table 2.—Major Clinical Outcomes in GUSTO-I Percentage of Patients

Streptokinase Both P Value,and Streptokinase Accelerated-Dose Thrombolytic Accelerated-Dose

Subcutaneous and Alteplase and Agents and Alteplase vs Both Heparin IV Heparin IV Heparin IV Heparin Streptokinase

Outcome (n = 9,796) (n = 10,377) (n = 10,344) (n = 10,328) Groups

24-hour mortality 2.8 2.9 2.3 2.8 0.005

30-day mortality 7.2 7.4 6.3 7.0 0.001Or nonfatal stroke 7.9 8.2 7.2 7.9 0.006Or nonfatal

hemorrhagic stroke 7.4 7.6 6.6 7.4 0.004Or nonfatal

disabling stroke 7.7 7.9 6.9 7.6 0.006

– From GUSTO Investigators.17

Page 14: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

RETAVASE: a novel thrombolytic agent foracute myocardial infarction

V arious thrombolytic agents are indevelopment for potential improvementsin half-life, mode of administration, fibrin

specificity, and proteolytic activity. RETAVASE isthe first available agent using selected domainsfrom the native t-PA molecule.16 The moleculeconsists of the kringle-2 and protease domains of native t-PA and shows predilection for fibrin-bound plasminogen. RETAVASE possessesimportant pharmacokinetic and mechanisticfeatures that contribute to the feasibility of adouble-bolus dosing regimen and restoration of flow in AMI patients.

DescriptionRETAVASE is a plasminogen activator whose

design was based on naturally occurring t-PA and is produced by recombinant genetic technology inE coli.19 It is prepared as a white, sterile, lyophilizedpowder for intravenous bolus injection afterreconstitution with Sterile Water for Injection, USP(without preservatives). The potency standard forRETAVASE, expressed in units (U), is notcomparable to that for other thrombolytics.RETAVASE is supplied as a 10.8 U vial to ensuresufficient drug for administration of each 10 Udose. Each vial of the lyophilized product contains18.8 mg of RETAVASE, along with arginine,phosphoric acid, and polysorbate 20 as inactiveingredients.

Biochemistry: a recombinant plasminogen activator

RETAVASE is a nonglycosylated recombinantplasminogen activator. It is a single-chain moleculethat consists of 355 amino acids corresponding to coding sequences 1 to 3 and 176 to 527 ofnative t-PA. Expression in E coli results in anonglycosylated protein, which accumulates inside cells as inactive inclusion bodies that must be refolded in vitro and purified to restore the native structure.19,32

The RETAVASE gene lacks the complementaryDNA sequence coding for the three N-terminal(finger, epidermal growth factor, and kringle-1)domains found in native t-PA but retains thekringle-2 and serine protease domains in afunctional form.32 The domain structures of theAlteplase and RETAVASE molecules are shown inFig 4 on page 18, and the structure-functionrelationships of these domains are shown in Table 3 on page 19.

Alteplase retains the kringle-1 and epidermalgrowth factor domains deleted from RETAVASE.20,32

Alteplase binds to liver receptors by means of thesedomains, thereby facilitating hepatic clearance.20

Oligosaccharide side chains retained by Alteplaseand not by RETAVASE also influence clearance.These deletions from the RETAVASE moleculeprovide a longer half-life than that of Alteplase,33

with two consequences: (1) less drug is required to maintain therapeutic levels and (2) RETAVASEcan be administered by intravenous bolus over aperiod of 2 minutes.

SECTION IV

RETAVASE® (Reteplase)preclinical pharmacology

17

Page 15: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

RETAVASE differs further from Alteplase in that it lacks the finger domain, a fibronectin-likeprojection that promotes high-affinity fibrinbinding.16 The RETAVASE molecule is also specificfor plasminogen bound to fibrin, but its affinity tofibrin is lower than that of Alteplase. RETAVASE mayactivate plasminogen at the surface as well as in theinterior of the thrombus.

In vitro catalytic activity and fibrin binding

RETAVASE has approximately 20% to 30% ofthe in vitro plasminogenolytic potency of Alteplaseas determined by a standard in vitro assay in whicheach activator is incubated with plasminogen in the presence of cyanogen bromide (CNBr) splitproducts (fragments) of fibrinogen that serve as a stimulator.32

RETAVASE, like Alteplase, is cleaved by plasminspecifically at the Arg275-Ile276 bond, indicating thatthe protease and kringle-2 domains have the samestructure as in native t-PA.32 Both RETAVASE andAlteplase lose activity in a similar manner whenincubated with increasing levels of PAI-1.

18

Fig 4.—Molecular structures of Alteplase and RETAVASE ®(Reteplase).

RETAVASE

Kringle 1

Protease

Kringle 2

EGF

Finger

Alteplase

Page 16: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Besides its lower plasminogenolytic activity in the presence of a stimulator, RETAVASE®

(Reteplase) does not bind to fibrin like Alteplase.32 Invitro fibrin binding was analyzed by addingRETAVASE or Alteplase to a newly forming clot in atest tube and measuring binding in response toincreasing amounts of fibrin. As shown in Fig 5,Alteplase is nearly 100% bound by 200 µg of fibrin,while only 20% of RETAVASE is found in the clotfraction. The amount of RETAVASE in the clotfraction increases at higher fibrin concentrations,possibly because of a nonspecific protein-proteininteraction. Thus, throughout the range of fibrinconcentration

from 0 to 1,000 µg, the binding of RETAVASEranges from about 10% to 30% of that of Alteplase.The lower catalytic activity and fibrin binding of RETAVASE relative to Alteplase may be due to the missing finger domain. This leaves only thekringle-2 lysine-binding site as the focus of theinteraction between RETAVASE and fibrin.20,32

19

Fig 5.—In vitro fibrin binding of RETAVASE®(Reteplase) and Alteplase in the presence ofincreasing fibrin concentrations.

– Data on file.34

0

20

250 500Fibrin (µg)

RelativeBinding

(%)

Alteplase

RETAVASE

750 1,000

60

80

100

40

Table 3.—Structure-Function Relationships of Native t-PA DomainsStructure Function

F domain High-affinity fibrin binding(finger, fibronectin-like)

E domain (epidermal growth factor-like domain) Receptor binding (liver)

K1 domain (kringle-1) Liver receptor binding(?) Fibrin binding(?)

K2 domain (kringle-2) Low-affinity interaction with fibrin (stimulation effect by fibrin)

P domains Protease function (plasminogen specific)

Carbohydrates Mediation of t-PA plasma clearance (half-life)

Page 17: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Fig 6 is a comparison of concentration-dependent in vitro clot lysis by RETAVASE,Alteplase, melanoma t-PA (which differs fromAlteplase in carbohydrate side chains), andUrokinase, based on the method of Collen et al.35

Lytic activity is represented by the percentage of radioactivity released from iodine 125 (125I)radiolabeled fibrinogen plasma clots after 4 hours of incubation with RETAVASE or one of the threecomparative agents.32 These data show thatRETAVASE had the same maximal lytic activity asAlteplase at equipotent concentrations, despite alower in vitro potency. RETAVASE is less active thanAlteplase in lysis of platelet-rich plasma clots andaged clots.36 This diminished activity of RETAVASEtoward old thrombi may reduce lysis of hemostatic

plugs, which are thought to be older clots that sealsmall vessel wall injuries.37

In vivo animal studies showed greaterpotency of RETAVASE than Alteplase

Jugular vein thrombolysis in rabbits. The invivo thrombolytic activity of RETAVASE was studiedin rabbits and dogs. In the rabbit model of jugularvein thrombosis, the RETAVASE effective dose for50% lysis (ED50) was 5.3-fold more potent per unit than the Alteplase ED50.

32 The investigatorsattributed the greater in vivo potency of RETAVASEto its lower clearance rate. An important conclusionof this study is that the relative in vitro potencies ofAlteplase and RETAVASE are reversed in animals.

20

The paradox: in vivo and in vitro profile reversal• The in vitro potency (specific activity) of RETAVASE as determined by plasminogenolytic assay

is lower than that of Alteplase by a factor of 2 or 3• RETAVASE is significantly more potent than Alteplase in in vivo models• The slower hepatic clearance and longer plasma half-life of RETAVASE may explain this reversal

Fig 6.—Concentration-dependent lysis of 125I-labeled human platelet-poor plasma clots byAlteplase, melanoma t-PA, RETAVASE® (Reteplase), and Urokinase.

– Adapted from Martin et al.32

Vehicle 0.1 1Concentration (nmol)

AlteplaseMelanoma t-PARETAVASEUrokinase

10 100

Clot Lysis(%)

Mean values.

100

80

60

40

20

0

Page 18: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Greater activity in coronary thrombolysis in dogs. The activity of RETAVASE in inducingcoronary reperfusion was investigated in open-chested anesthetized dogs in which electrical injuryto the intima of the left circumflex coronary arterywas used to simulate AMI.32 A double-bolusintravenous regimen of RETAVASE proved to besuperior (P<0.05) to a single bolus in stabilizingcoronary artery blood flow and reducingreocclusion, but doubling the dose of a single bolusdid not have this effect.32 The total plasma clearancerate of RETAVASE was at least three times lowerthan that of Alteplase, suggesting that thiscontributed to the greater thrombolytic potency of RETAVASE.32

In another study utilizing the same caninemodel, RETAVASE was shown to induce reperfusion

significantly more rapidly than Alteplase,Anistreplase, Streptokinase, and Urokinase whenadministered at clinically equivalent doses (Fig 7).32

Pharmacokinetics: clearance through liver andkidneys. The pharmacokinetic properties ofRETAVASE were extensively studied in rats, rabbits,dogs, and nonhuman primates.32 Half-life and totalplasma clearance parameters for RETAVASE incomparison with Alteplase in humans are shown inTable 4. Based on the mean liver uptake of therespective injected doses, hepatic clearance accountsfor much less of the total plasma clearance ofRETAVASE than for Alteplase in all species studied.RETAVASE is cleared mainly by the kidneys,although inhibitory proteins in the blood contributeto RETAVASE clearance through biochemicalinactivation.

21

Table 4.—Pharmacokinetic Properties of RETAVASE® (Reteplase) in Plasma Following Intravenous Bolus Injection

Dose N t½α (min) t½β (min) CL (mL/min)

6 U (10.4 mg) 7 13.9 ± 0.7 173 ± 33 183 ± 15

10 U (18 mg) 4 19.2 ± 1.0 375 ± 34 104 ± 11

15 U (27 mg) 10 18.8 ± 0.8 377 ± 20 139 ± 21

Mean ± SEM. – From Martin et al.32

t½ = half-life; CL = total plasma clearance.

Fig 7.—More rapid coronary reperfusion in a canine model.

– Adapted from Martin et al.32

20

0

60

40

80

100

120

RETAVASE®

(Reteplase)

Time toReperfusion

(min)

Alteplase Anistreplase

At Dose Corresponding to Clinical Practice(Alteplase by standard infusion)

Streptokinase Urokinase

** *

*

*P < 0.05 vs Reteplase.Mean ± SEM.n = 3 to 6 per group.

Page 19: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Toxicology showed no internal bleeding, evenat highest dose. The effect of RETAVASE®

(Reteplase) on platelets, bleeding times, and plasmaproteins was studied in rats, rabbits, and dogs.Pharmacologically effective doses produced a dose-dependent reduction in fibrinogen, plasminogen,and a2-antiplasmin 2 hours post-injection.32 In a14-day subchronic toxicity study in cynomolgusmonkeys, all dose levels induced unilateral focalhemorrhages that correlated with low red blood cellcounts, but internal hemorrhaging did not develop,even at the highest dose.

Mutagenicity was not detectable in Salmonellatyphimurium or in rat bone marrow erythrocytestested in vivo for 2 weeks with daily intravenousadministration of doses up to 1.4 U/kg.32 No adverseeffects on reproduction or fetal development wereseen at doses of 4.3 U/kg in rats. The minimumlethal dose of RETAVASE (from hemorrhage and/or

hypotension) in rats, rabbits, and monkeys exceeds8.4 U/kg given as a single injection; no deathsoccurred in animals receiving 4.2 U/kg.32

Summary of preclinical findings. Thepreclinical pharmacology studies demonstrate thatRETAVASE has more potent in vivo thrombolyticactivity than Alteplase on both a dose and molarbasis, which contrasts with the results of in vitrostudies. RETAVASE induces more rapid reperfusionthan other thrombolytic agents in animal models ofcoronary thrombosis. Reperfusion is significantlymore rapid after intravenous bolus injection thanafter infusion, and a double bolus is significantlymore active in stabilizing coronary artery blood flowand preventing reocclusion.

22

Page 20: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Well-tolerated hemostatic and fibrinolytic effects

The hemostatic and fibrinolytic properties ofRETAVASE were investigated in a Phase Idose-ranging study in 18 healthy male

volunteers.38 Intravenous bolus doses ranging from0.11 U to 5.5 U given over 2 minutes did not alterplasma fibrinogen levels except at the higher doses.However, a2-antiplasmin and fibrin D-dimersdecreased in a dose-dependent fashion. RETAVASEwas well tolerated, and antibodies to RETAVASE didnot appear in plasma samples taken up to 1 yearafter the study.

A single intravenous bolus dose of 6 U (10.4 mg) of RETAVASE or placebo was given over 2minutes to seven healthy male volunteers in anotherPhase I study, this time using a randomized, single-blind, placebo-controlled, crossover design.20

Fibrinogen levels were unchanged by RETAVASE.Plasminogen and a2-antiplasmin levels, however,were reduced to 83% and 64%, respectively, ofbaseline values, probably reflecting a systemicactivation of the fibrinolytic system. RETAVASE was well tolerated, and antibodies to RETAVASEwere not detected.20

Dose-response restoration of coronary flowin patients with AMI

The hemostatic and fibrinolytic effects ofRETAVASE doses were evaluated in patients withAMI. The effects of RETAVASE on fibrinogen,plasminogen,a2-antiplasmin, fibrin degradationproducts (FDP, as represented by fibrin-specific D-dimers), and fibrinogen degradation products(FgDP) in the two German RecombinantPlasminogen Activator Studies in patients with AMI (GRECO39 and GRECO-DB40) are summarizedin Table 5. At 2 hours after administration (2-hourvalues were chosen because they represent thestrongest effects), mean values of fibrinogen wereapproximately 45% of baseline for the 15 U and 10 + 5 U regimens and 60% of baseline for the 10 U regimen. The higher doses used in the GRECO studies resulted in a greater reduction inplasminogen and a2-antiplasmin and a greaterincrease in D-dimers and FgDP. The 15 U and the 10 + 5 U regimens had a greater effect on theseparameters than the 10-U regimen, suggesting dose dependency.

Administration of RETAVASE in the GRECO-DB(double bolus) study caused the median fibrinogenlevel to decrease from a baseline of 286 mg/dL to 63%of this value at 24 hours (Fig 8).40

SECTION V

RETAVASE® (Reteplase) clinical pharmacology

23

Table 5.—Hemostatic and Fibrinolytic Parameters at 2 Hours in the GRECO and GRECO-DB Studies

2–Hour Values as a Mean Percentage of Baseline

10 U RETAVASE 15 U RETAVASE 10 + 5 U RETAVASEParameter (n = 38-41) (n = 66-99) (n = 43-49)

Fibrinogen 60 44 45

Plasminogen 55 41 42

α2-antiplasmin 30 26 19

D-dimers 2,940 5,709 5,986

FgDP 10,449 19,507 42,611

FgDP = fibrinogen degradation products. – Data on file.34

Page 21: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Pharmacokinetics in healthy volunteersThe pharmacokinetics of RETAVASE were

evaluated in three studies in healthy volunteers.Drug was administered by IV injection over 2 minutes in each study. Plasma concentrations of RETAVASE antigen, ie, protein, were measuredusing a monoclonal antibody enzyme-linkedimmunosorbent assay. A plasminogenolytic activity assay based on the method of Verheijen et al41 was used to measure the more relevant active concentrations.

In one of the studies, seven subjects received a single intravenous 6.0 U bolus dose ofRETAVASE.20 The activity half-life was 11.2±0.4 minand the antigen half-life was 13.9±0.7 min (2.4 mL/min/kg), followed by a terminal antigenhalf-life of 173±33 min. Plasma clearance was371±13 mL/min (4.9 mL/min/kg) for activity inplasma (ie, effect on fibrin) and 183±5 mL/min (2.4 mL/min/kg) for antigen. The half-life andclearance of RETAVASE activity were 3.3-fold longerand 3.3-fold lower, respectively, than in volunteerswho received Alteplase in a study by Seifried et al.42

In addition, the half-life and clearance for antigenwere 4.2-fold longer and 3.9-fold lower, respectively.42

In another study, 18 volunteers receivedsequential doses of RETAVASE between 0.11 U and 5.5 U.38 Plasma AUC and Cmax showed dose-dependent linear increases. Clearance was 406±40mL/min (4 mL/min/kg), and half-life for the 5.5 Udose was 14.4±1 min.

Data from the studies in healthy subjectsdemonstrate that RETAVASE pharmacokinetics arelinear over the dose range of 0.11 to 5.5 U. Plasmaconcentrations of RETAVASE antigen persist for alonger period of time than do those of activity.

Pharmacokinetics in AMIThe pharmacokinetics of RETAVASE in patients

with AMI were evaluated using subsets of patients in two trials. In the GRECO study, doses wereadministered as single boluses of 10 U or 15 U39;in MF4292, patients received a single bolus of 15 Uor a double bolus of either 10 + 5 U or 10 + 10 U.34

The mean RETAVASE plasma concentrations(activity) for the two studies are shown in Fig 9 and Fig 10.

24

Fig 8.—Fibrinogen profile over time following a 10 + 5 U intravenous double bolus of RETAVASE® (Reteplase) in GRECO-DB.

– Adapted from Tebbe et al.40

Time (hours)

Percentageof

Baseline

00

20

40

60

80

100

2 4 8 24

Page 22: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Linear pharmacokineticsThe pharmacokinetics of RETAVASE in patients

with AMI as well as in healthy volunteers are linearover the dose range of 0.11 to 20 U and do notappear to be affected by the target disease. The

effective half-life of RETAVASE is 13 to 16 minutes.Plasma antigen concentrations persist for a longerperiod than plasma activity. This is not unexpected,since the antigen assay measures all RETAVASE-related protein, not all of which is active RETAVASE.

25

Fig 9.—Mean RETAVASE® (Reteplase) plasma concentrations (activity) afteradministration of 10 U and 15 U doses in patients with AMI in the GRECO study.

– Data on file.34

RETAVASE(U/mL)

Time (hours)

10 U15 U

500

0 0.5 1 1.5 2

0

1,000

1,500

2,000

2,500

3,000

3,500

Fig 10.—Mean RETAVASE plasma concentrations (activity) after administration of 10 + 5 U, 10 + 10 U, and 15 U doses in patients

with AMI in MF4292.

– Data on file.34

RETAVASE(U/mL)

Time (hours)

10 + 5 U10 + 10 U15 U

0 0.5 1 1.5 2

0

500

1,000

1,500

2,000

Page 23: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

RETAVASE: an effective thrombolytic agent

S ix clinical trials of significance haveidentified the appropriate dose anddemonstrated the efficacy and safety of

RETAVASE as therapy for AMI. Three were Phase IIdose-finding studies (without comparative agents)

evaluating the activity of RETAVASE in restoringblood flow through the infarct-related artery (Table 6). Three comparative studies wereperformed to compare RETAVASE withStreptokinase (INJECT) or Alteplase (RAPID 1 and RAPID 2) (Table 7). Two of these, RAPID 2 and INJECT, were Phase III studies, while RAPID 1was a Phase II study.

SECTION VI

RETAVASE® (Reteplase) clinical efficacy and activity

27

Table 6.—Non-Comparative Studies in Patients With AMIStudy Design Dose Strength Frequency, Number Primary Key Entry

Protocol Location Population and Form Duration of Patients Evaluation Criteria

GRECO39 Germany OL 10 U RP Single bolus 42 90-minute ST segment patency and elevation

AMI patients 15 U RP Single bolus 100 TIMI 3 ratesOnset ofischemic

pain within 6 hours

18-75 years old

GRECO- Germany OL 10 + 5 U RP Double bolus 51 90-minute ST segment DB40 patency and elevation

AMI patients TIMI 3 ratesOnset of

ischemic pain within 6 hours

18-75 years old

MF429234 Germany R, OL 15 U RP Single bolus 9 90-minute ST segment patency and elevation

AMI patients 10 + 5 U RP Double bolus 8 TIMI 3 rates,pharmacokinetics, Onset of

10 + 10 U RP Double bolus 8 hemostasis ischemic painparameters within 6 hours

18-75 years old

OL = open label; R = randomized; RP = Reteplase.

Page 24: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

The six studies evaluated four different dosage regimens of RETAVASE™ (Reteplase):

• Single 10 U bolus• Single 15 U bolus• 10 U bolus followed by a 5 U bolus

30 minutes later (10 + 5 U)• 10 U bolus followed by a 10 U bolus

30 minutes later (10 + 10 U).Each bolus was administered as a slow

intravenous injection over an interval not exceeding 2 minutes. Of the 3,805 patientstreated with RETAVASE, 3,292 received the10 + 10 U regimen.

Dose-finding trials determined patency ofinfarct-related arteries

GRECO. This was a sequential rising-dose studythat enrolled 142 patients with AMI who presentedless than 6 hours after the onset of symptoms.39

RETAVASE was given as a single 10 U bolus to thefirst 42 patients and as a 15 U bolus to the next100 patients. Patients also received an intravenousbolus of heparin immediately after the bolusdose of RETAVASE, and aspirin was given dailythroughout the study. Nitroglycerin wasadministered intravenously unlesshemodynamically contraindicated.

28

Table 7.—Comparative Studies in Patients With AMIStudy Design Dose Strength Frequency, Number Primary Key Entry

Protocol Location Population and Form Duration of Patients Evaluation Criteria

RAPID 1 United States R, OL 15 U RP Single bolus 146 90-minute ST segment (Phase II)4 Germany (angiograms patency elevation

United Kingdom read 10 + 5 U RP Double bolus 152Austria blinded) Onset of

10 + 10 U RP Double bolus 154 ischemic pain AMI patients within 6 hours

100 mg AP Bolus + 154(standard dose) 3-hour

infusion 18-75 years old

RAPID 25 United States R, OL 10 + 10 U RP Double bolus 169 90-minute ST segment Germany (angiograms TIMI grade elevation or

read blinded) 100 mg AP Bolus + 155 bundle branch (accelerated 1.5-hour block

dose) infusionAMI patients Onset of

ischemic pain within 12 hours

≥18 years old

INJECT3 United Kingdom R, DB 10 + 10 U RP Double bolus 3,004* 35-day ST segment Germany mortality elevation or

Poland AMI patients 1.5 MU SK 1-hour infusion 3,006* bundle branch Sweden block

HungaryFinland Onset ofSpain ischemic pain

Lithuania within 12 hoursAustria

≥18 years old

*74 patients were randomized but were not treated. Overall, 2,965 RETAVASE® (Reteplase) patients and 2,971 Streptokinase patients received treatment. AP =Alteplase; DB = double-blind; INJECT= International Joint Efficacy Comparison of Thrombolytics; OL = open label; R = randomized;RAPID 1 = Reteplase Angiographic Phase II International Dose-finding Study; RAPID 2 = Reteplase vs Alteplase Patency Investigation During Acute MyocardialInfarction Study; RP = RETAVASE; SK = Streptokinase.

Page 25: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Activity was evaluated by means ofangiographically determined patency of infarct-related arteries. Coronary arteriography of thepresumed infarct-related artery was conducted30 and 90 minutes after the bolus injection ofRETAVASE™ (Reteplase, recombinant) and repeatedat 24 to 48 hours and prior to discharge at 14 to 21days. Patency was assessed by TIMI flow grade andhad to be rated as grade 2 or 3 by two independent,experienced investigators.

The patency rate (TIMI 2 + 3 flow) at 90minutes for the single-bolus 10 U dose group was66%.39 Since the 66% was below the predetermined70% lower limit of activity for Alteplase in the TIMItrial,43 the next 100 patients received a single bolus of15 U.39 The 90-minute patency rate in these patients

was increased to 75% and the TIMI 3 rate alone was69% (Fig 11), a result similar to that associated withAlteplase thrombolysis.39

GRECO-DB. A second bolus of RETAVASEmaintained patency of coronary arteries. While theGRECO study demonstrated that single-bolusadministration of RETAVASE could lead to highpatency rates in patients with AMI, fluctuations wereobserved in the early angiograms.39 In some patients,infarct-related arteries that were patent at 30 or 60minutes became reoccluded at 90 minutes.

In GRECO-DB, patients received an initial 10 U bolus of RETAVASE followed by 5 U ofRETAVASE 30 minutes later.40 The purpose of thisdouble bolus was to optimize perfusion of theinfarct-related artery and maintain patency during

29

The TIMI flow gradesSince the TIMI trial, coronary flow after reperfusion therapy has been classified by TIMIangiographic grade.27

• TIMI 0: no penetration of contrast medium beyond the thrombus• TIMI 1: minimal penetration; no significant flow• TIMI 2: restored but sluggish flow• TIMI 3: brisk flow

Fig 11.—Patency of infarct-related arteries in the GRECO study.

– Adapted from Neuhaus et al.39

0Dose

20

40

60Patients

(%)

80

100

Angiogramat 30 min

10 U(n = 32)

15 U(n = 62)

Angiogramat 60 min

10 U(n = 41)

15 U(n = 99)

Angiogramat 90 min

10 U(n = 42)

15 U(n = 99)

Angiogramat 24 to 48 hr

10 U(n = 42)

15 U(n = 95)

Angiogramat 14 to 21 days

10 U(n = 38)

15 U(n = 85)

TIMI 3TIMI 2

65 6673 74

66

75

92 9289

46

38

48

5852

69

88 85 86 85

90

Page 26: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

the early phase of thrombolysis by sustaining the RETAVASE™ (Reteplase, recombinant) plasma level.40 Otherwise, the protocol was similar to GRECO.

Fifty-one patients were treated with the 10 + 5 U RETAVASE regimen, and 50 underwentangiography (Table 8).40 The 90-minute patency rate(TIMI 2 + 3 flow) of 78% was slightly higher thanthe 75% observed with the 10 + 5 U regimen inGRECO; the TIMI 3 rate was 58%, compared with69% in GRECO.

Comparative clinical trialsThe safety and efficacy of RETAVASE were

evaluated in three controlled clinical trials in whichRETAVASE was compared to other thrombolyticagents. INJECT compared RETAVASE toStreptokinase to assess the relative effects on 35-daymortality rates. RAPID 1 and RAPID 2 were bothangiographic trials comparing the effect on coronarypatency of RETAVASE and Alteplase (see Section VIIIbeginning on page 37).

INJECT establishes efficacy ofRETAVASE™ (Reteplase, recombinant) in reducing mortality

INJECT was a randomized, double-blind,Phase III multicenter study designed to confirm that RETAVASE was effective in reducing mortalityin AMI. In INJECT, RETAVASE was compared toStreptokinase, the standard thrombolytic agent in Europe.3 The trial was not powered, however,to statistically detect modest differences between the two agents. The goal of the study was todemonstrate the efficacy and safety of RETAVASE.The 35-day mortality rate of RETAVASE wascompared to the standard Streptokinase regimen.

A total of 5,936 patients in nine Europeancountries received thrombolytic therapy no laterthan 12 hours after the onset of symptoms of AMI.3

There was no upper age limit. A total of 2,965patients randomized to RETAVASE 10 + 10 U (thesecond injection given 30 minutes after the first) and2,971 patients randomized to a 1.5 MU infusion ofStreptokinase over 60 minutes were assessable. Allpatients received intravenous heparin for at least 24hours. Patients were given 250 mg to 350 mgaspirin, then 75 mg to 150 mg daily.

30

Table 8.—Patency of Infarct-Related Arteries in GRECO-DBPatency Rate

Angiogram at 30 minutes (n = 48)TIMI grade 2 or 3 50%TIMI grade 3 31%

Angiogram at 60 minutes (n = 50)TIMI grade 2 or 3 72%TIMI grade 3 50%

Angiogram at 90 minutes (n = 50)TIMI grade 2 or 3 78%TIMI grade 3 58%

Angiogram at 24 to 48 hours (n = 50)TIMI grade 2 or 3 94%TIMI grade 3 84%

Angiogram at 14 to 21 days (n = 49)TIMI grade 2 or 3 90%TIMI grade 3 82%

TIMI = Thrombolysis in Myocardial Infarction trial. – From Tebbe et al.40

Page 27: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

ResultsThe 35-day study mortality rates are shown in

Fig 12, and the Kaplan-Meier survival curves overthe 35-day interval are depicted in Fig 13.3 Mortalityat day 35 in patients who received study medicationwas 8.90% in the RETAVASE group and 9.43% inthe Streptokinase group, a difference of 0.53%.

These data provide confirmation that RETAVASE is effective in reducing mortality after AMI.

At 6 months, mortality rates (Kaplan-Meierestimate) were 11.02% for RETAVASE and 12.05%for Streptokinase, a difference of 1.03%.3 Theseresults provide further evidence that RETAVASE iseffective in reducing mortality after AMI.3

31

Fig 12.—35-day mortality rates in the INJECT study.

– Adapted from INJECT.3

Patients(%)

0RETAVASETM

(Reteplase, recombinant)10 U + 10 U(n = 2,965)

8.90%

2

4

6

8

10

Streptokinase1.5 MU/60 min

(n = 2,971)

9.43%

Difference in mortality: RETAVASE – Streptokinase = –0.53% (90% CI: –1.76% to 0.71%).

P=NS

Fig 13.—Kaplan-Meier survival curves from day 0 to 35 in INJECT.

– Adapted from INJECT.3

0

2

6

8

10

4

0 7 14 21 28 35

Days in Study

Mortality (%)

Streptokinase

RETAVASE

Page 28: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

INJECT substudy demonstrates earlyresolution of ST segment elevation

INJECT investigators conducted an exploratorysubstudy among the 1,909 patients in Germany toevaluate the prognostic power of early resolution of ST segment elevation.44

ST segment elevation was classified as achievingcomplete (≥70%), partial (30% to 70%), or no (0%to 30%) resolution after 3 hours of thrombolytictherapy.44

Results irrespective of therapy showed that the 35-day mortality rate in 1,398 patients withinfarct age ≤6 hours for complete, partial, or no ST resolution was 2.5%, 4.3%, and 17.5%,respectively (P<0.0001).44 As can be seen in Table 9,RETAVASE™ (Reteplase, recombinant) achievedcomplete or partial ST segment resolution in 82% of patients at 3 hours.44 Although the authorsconcluded that resolution of ST segment elevationmay correlate with mortality, the correlation withmortality and other patient outcomes was notestablished.

32

Table 9.—Percentages of RETAVASE® (Reteplase) Patients in INJECT With Complete, Partial, or No ST Segment Elevation Resolution

All Patients Anterior MI Inferior MI

RETAVASE RETAVASE RETAVASE(n = 692) (n = 318) (n = 374)

ST resolution (%)

Complete 51 36 65

Partial 31 43 22

No 17 22 14

MI = myocardial infarction. – From Schröder et al.44

Page 29: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

GUSTO-III: a large-scale, mortality trialversus Alteplase

The accelerated-dose Alteplase regimenprovided a 14% relative reduction in mortality fromAMI over Streptokinase in the GUSTO (now knownas GUSTO-I) trial in 1993.17 The most plausibleexplanation is that Alteplase achieved a higher TIMI3 flow rate in infarct-related coronary arteries, thussupporting the open-artery hypothesis.18 Impressiveas these results may seem, however, analysis ofangiograms taken 90 minutes after the start oftherapy reveals that only 54% of the Alteplase-treated patients actually attained TIMI 3 flow.This suggests that further increases in 30-daysurvival may be achieved if greater reopening of occluded arteries is also achieved. Moreover,once achieved, the patency should be sustained,since the initial benefit may be attenuated bysubsequent reocclusion.45

As discussed in Section VIII beginning on page37, the RAPID 2 trial showed differences in patencybetween RETAVASE™ (Reteplase, recombinant) andAlteplase.5 Direct comparison of these two agents fortheir effects on mortality was the logical next step.

In recognition of the need for additional study,the GUSTO-III (Global Use of Strategies to OpenOccluded Coronary Arteries) trial was initiated inOctober 1995 with plans of enrolling approximately15,000 patients of any age who present within 6hours of the onset of symptoms of an AMI. Patientswere randomized in a 2:1 ratio, 10,000 to thedouble-bolus RETAVASE regimen and 5,000 to theaccelerated-dose Alteplase regimen (Fig 14).

33

Fig 14.—GUSTO-III protocol algorithm.

– Data on file.34

Identify eligible patient

Aspirin 150-325 mg2:1 Randomization

10 U bolus 3230 min apart

5,000 U bolus For patients ≥ 80 kg: 1,000 U/hr For patients < 80 kg: 800 U/hrIV heparin 3 ≥ 24 hours

15 mg bolus0.75 mg/kg over 30 min not to exceed 50 mg0.50 mg/kg over 60 min not to exceed 35 mgTotal dose ≤100 mg

Primary end point: 30-day mortality

IV AlteplaseHeparinIV Reteplase

N = 15,000

2 1

Page 30: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Table 11.—In-Hospital CerebrovascularEvents (%) in INJECT Trial 3

RETAVASE StreptokinaseEvent (n = 2,965) (n = 2,971)

All in-hospital strokes 1.21 1.01*

Intracranial hemorrhage 0.77 0.37†

Nonhemorrhagic stroke 0.30 0.30

Unknown etiology 0.13 0.34

*P = 0.45; †P = 0.04.

Double-bolus safety profile established

T he safety of RETAVASE was evaluated in3,805 patients with AMI and 45 healthyvolunteers. Most of the patients (3,296)

participating in the clinical trials received the 10 + 10 U double-bolus regimen. These studiesdemonstrated that the safety profile of RETAVASE issimilar to that of other available thrombolytic agentswith respect to

• Cardiac events• Allergic events• Other adverse events

Bleeding Bleeding is the most common complication

encountered during the use of thrombolytics.Internal bleeding can occur at intracranial orretroperitoneal sites or in the gastrointestinal,genitourinary, or respiratory tracts. Superficialbleeding occurs mainly at sites of invasion ordisturbance (eg, venous cutdowns, arterialpunctures, or recent surgical incisions). Intracranialbleeding is the most important safety concernassociated with the use of thrombolytic agents.The overall incidence for RETAVASE was 0.71%.

The incidence of bleeding varied widely amongthe clinical trials and was influenced by the use ofarterial catheterization and other highly invasiveprocedures, and whether the study was performed in Europe or the United States. The overall incidenceof bleeding in patients who received the RETAVASE 10 + 10 U double-bolus regimen in INJECT,3

RAPID 1,4 and RAPID 25 was 21.1%. This percentagewas similar to that of Streptokinase and Alteplase.The rates of bleeding in the INJECT trial aresummarized in Table 10.

Strokes In-hospital stroke rates for the 10 + 10 U

RETAVASE regimen in the INJECT trial issummarized in Table 11.

There were no significant differences in overallstroke rates between RETAVASE and Streptokinasein the INJECT study.3 Overall in-hospital strokerates for the RETAVASE and Streptokinase treatmentgroups were 1.23% and 1.00%, respectively. Therewas an increase in the incidence of in-hospitalintracranial hemorrhage for RETAVASE (0.77%)versus Streptokinase (0.37%, P=0.04). Thepercentage of patients requiring transfusion wassimilar in the two groups (0.7% for RETAVASE and1.0% for Streptokinase).

SECTION VII

RETAVASE® (Reteplase) safety profile

35

Table 10.—Bleeding Incidence (%) inINJECT Trial

RETAVASEBleeding Site (n = 2,965)

Injection site 4.6

Gastrointestinal 2.5

Genitourinary 1.6

Anemia, site unknown 2.6

Page 31: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

No dosing adjustment is required based onpatient’s weight

In low-weight (<65 kg) and higher-weight (>65 kg) RETAVASE® (Reteplase)-treated patients,the rates of cerebrovascular events, total strokes, andhemorrhagic strokes were similar. In the INJECTstudy,3 hemorrhagic stroke rates were 0.7% for bothsubgroups.

For hemorrhages of any type, combined datafrom INJECT3 and the RAPID4,5 studies showed that 25.8% of low-weight patients and 21.0% ofhigher-weight patients experienced at least onehemorrhage. For hemorrhages requiring atransfusion, the low-weight and higher-weightgroups were more similar (3.8% vs 3.1%,respectively).3,4

Because of these results, and particularly because the incidence of hemorrhagic strokes wassimilar in both low-weight and higher-weightpatients, it was concluded that dose adjustmentbased on a patient’s weight is not necessary forRETAVASE use.

RETAVASE, therefore, can be administeredrapidly in a convenient double-bolus dosingregimen, with no dosage adjustment necessaryregardless of weight.

Congestive heart failure and cardiogenicshock in INJECT

As can be seen in Table 12, significantly fewerpatients treated with RETAVASE in the INJECTtrial had new or worsening CHF (P=0.004) andcardiogenic shock (P=0.03) than patients treatedwith Streptokinase. These results demonstrate thatthe rate of CHF reduced by Streptokinase can befurther reduced by RETAVASE.

Other cardiovascular events in INJECTThe incidence of hypotension, pulmonary

edema, atrial fibrillation or flutter, and asystole wereall significantly lower for patients treated withRETAVASE.3 While a variety of additionalcardiovascular events had similar rates for bothagents, there were no cardiac complications forwhich the incidence rate was significantly lower in the Streptokinase group. Overall, 60.3% ofRETAVASE-treated patients and 63.0% ofStreptokinase-treated patients experienced at leastone cardiovascular adverse event, while 20.0% and 22.2%, respectively, had at least one seriouscardiovascular adverse event.

Allergic eventsThe incidence of allergic events in the INJECT

study was lower in the RETAVASE group (1.1%)than in the Streptokinase group (2.0%). Seriousevents occurred more often in patients receivingStreptokinase (0.5%) than in those receivingRETAVASE (0.1%).

Across all studies, 42 of 3,288 RETAVASE-treated patients (1.3%) experienced an allergicevent, compared to 59 of 2,971 Streptokinase-treatedpatients (2.0%).

Other adverse eventsThere were no major differences between

RETAVASE and the control agents in the incidenceof other events. The higher incidences of minorbleeding and other events in RAPID 1 and 2 than in INJECT are due to angiographic intervention and are consistent with rates in other angiographictrials. There was also a more detailed reportingscheme in the RAPID trials, since INJECT focusedon major clinical end points (mortality, stroke,cardiovascular events, allergic reactions) appropriatefor a mortality trial.

36

Table 12.—Patients (%) Reported With CHF and Cardiogenic Shock in INJECTRETAVASE Streptokinase(n = 2,965) (n = 2,971) P Value

Congestive heart failure 24.8 28.1 0.004

Cardiogenic shock 4.6 5.8 0.03

Page 32: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

37

SECTION VIII

RETAVASE® (Reteplase) angiographic trials

RAPID 1 and RAPID 2 clinical trialsestablish activity of bolus dosing

Both the RAPID 14 and RAPID 25 clinical trials were angiographic studies designed to test the hypothesis that bolus dosing of RETAVASEwas superior to administration of Alteplase in achieving complete perfusion (TIMI 3 flow) andpatency (TIMI 2 + 3 flow) of the infarct-relatedartery 90 minutes after initiation of thrombolytictherapy in patients with AMI.

Secondary end points in RAPID 1 and RAPID 2were patency at 60 minutes after the initiation oftherapy, patency prior to hospital discharge (in 5 to14 days), and LVF prior to discharge.4,5

The correlation between patency and patientoutcome has not been established. The angiographic

trials were not designed or powered to compareRETAVASE and Alteplase with respect to theoutcomes of mortality or stroke, consequentlycomparisons of mortality or stroke cannot be made.

RAPID 1. The RAPID 1 study compared threedoses of RETAVASE to Alteplase in patients withAMI.4 A total of 606 patients treated within 6 hoursof the onset of symptoms of AMI were randomizedto one of four treatment groups:

• 146 had a 15 U single RETAVASE bolus• 152 had a 10 + 5 U RETAVASE

double-bolus dose• 154 had a 10 + 10 U RETAVASE

double-bolus dose• 154 had a 3-hour infusion of Alteplase

(100 mg)

Fig 15.—Summary of infarct-related artery patency rates in RAPID 1.

– Adapted from Smalling et al.4

TIMI 3

77.2

62.8*66.7†

85.2

*P <0.01 compared to Alteplase.†P <0.05 compared to Alteplase.‡P <0.001 compared to Alteplase.The P-values represent a comparison of the multiple RETAVASE groups to the single Alteplase group without adjustment for multiple comparisons.

TIMI 2

0

40

20

60

80

100

RETAVASE15 U

RETAVASE10 + 5 U

RETAVASE10 + 10 U

Patients(%)

(n = 137)

Alteplase3 hr

(n = 145) (n = 138) (n = 142)

87.8 85.580.5

95.1†

0

40

20

60

80

100

RETAVASE15 U

RETAVASE10 + 5 U

RETAVASE10 + 10 U

Patients(%)

(n = 124)

Alteplase3 hr

(n = 123) (n = 123) (n = 123)

49.0

40.945.7

62.7†

70.7 71.0 73.2

87.8‡

Patency at 90 minutes Patency at hospital discharge

Page 33: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Aspirin was given at a dose of 200 to 325 mg immediately before administration ofeach thrombolytic agent and continued daily until hospital discharge. Heparin was started as an intravenous bolus just before the start ofthrombolytic therapy and then infused at the rate of 1,000 U/hr for at least 24 hours.

Coronary arteriography was performed at 30 and 60 minutes after initiation of thrombolytictherapy (when possible) and at 90 minutes. Leftventriculography was also performed after coronaryarteriography, and TIMI grade was estimated at 90 minutes. The complete angiographic procedurewas repeated between 5 days after admission anddischarge from the hospital.

Results of RAPID 1. Patency data in theRAPID 1 study are summarized in Fig 15 on page 37.4 The 10 + 10 U RETAVASE™ (Reteplase,recombinant) group achieved patency (TIMI 2 + 3flow) in 85% of patients at 90 minutes compared to 77% of patients treated with Alteplase. Thisdifference was not statistically significant. At 5 to 14days after treatment, however, RETAVASE achievedsignificantly higher patency rates than the Alteplasegroup (95% vs 88%, P<0.05). Bolus administrationof 10 + 10 U of RETAVASE resulted in improvedcomplete flow, both at 90 minutes and beforehospital discharge, compared with standard-doseAlteplase. These findings were associated withimproved global and regional function at hospitaldischarge. Patients in the RETAVASE 10 + 10 U andAlteplase groups exhibited similar global ventricularfunction in the acute stage of treatment. There was,however, a significant difference in favor ofRETAVASE at hospital discharge in left ventricularejection fraction and regional wall motion.4 Thebleeding risk of RETAVASE was similar to thatassociated with standard-dose Alteplase.

RAPID 1 provided dose-finding data thatsupported the use of the 10 + 10 U double-bolusregimen as the standard for subsequent studies.The correlation between patency and patient

outcome has not been established. This trial was not designed or powered to make comparisons ofmortality or stroke.

The need for coronary interventions within 6 hours of thrombolysis was also evaluated inRAPID 1. There were no significant differences inthe incidence of early rescue PTCAs (14.9% versus22.1%),4 or the overall in-hospital incidences ofPTCAs (RETAVASE 46.8%, Alteplase 50.6%).34

RAPID 2. In the RAPID 2 study, 324 patientstreated within 12 hours of the onset of symptoms of AMI were randomized to receive either a 10 + 10 U double bolus of RETAVASE 30 minutesapart (n=169) or the accelerated (front-loaded) 90-minute Alteplase regimen (n=155). Patients also received aspirin and heparin.5 Angiographicprocedures were conducted as in RAPID 1.

Results of RAPID 2. Patency (TIMI 2 + 3 flow)and complete perfusion (TIMI 3 flow) rates weresignificantly higher for the 10 + 10 U RETAVASEregimen than for Alteplase at 60 minutes and 90minutes5 (Fig 16). At the primary time point of 90minutes, the patency rate in the RETAVASE groupwas 10% higher and the complete perfusion rate(TIMI 3) was 15% higher (P=0.011) than in theAlteplase group.

In RAPID 2, patients who received treatmentwithin 6 hours after onset of symptoms had betteroverall patency and complete perfusion in bothgroups than did patients who presented between 6 and 12 hours. Notably, patients treated withRETAVASE had better patency in all time-to-treatment categories. RETAVASE achieved 90-minute patency (TIMI 2 + 3 flow) in 86.5% and complete perfusion (TIMI 3 flow) in 62.4% ofpatients treated within 6 hours. The correspondingrates for Alteplase were 77.2% and 47.2%.5

The correlation between patency and patientoutcome has not been established. This trial was not designed or powered to make comparisons ofmortality or stroke.

38

Page 34: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

The need for coronary interventions within 6 hours of thrombolysis was also evaluated inRAPID 2. There were significantly fewer early rescue PTCAs for patients treated with RETAVASE

compared to accelerated-dose Alteplase. There wereno differences in the overall in-hospital incidences of PTCAs (Fig 17).5

39

Fig 16.—Patency profile at 60, 90 minutes and 5 to 14 days in RAPID 2.

– Adapted from Bode et al.5

0

20

60

80

100

40

Patients(%)

60 min 90 min 5-14 days

90 89

73

83†

66

82*

Alteplase(n = 115)

RETAVASETM

(Reteplase,recombinant)

(n = 121)

Alteplase(n = 146)

RETAVASETM

(Reteplase,recombinant)

(n = 157)

Alteplase(n = 113)

RETAVASETM

(Reteplase,recombinant)

(n = 128)

77 75

45

60†

37

51†

*P <0.01 RETAVASE vs Alteplase.†P <0.05 RETAVASE vs Alteplase.

The correlation between patency and patient outcome has not been established. Due to different numbers of patients in the 60- and 90-minute groups and nonrandomization to these time points, no comparison can be made between 60- and 90-minute data.

TIMI 3 TIMI 2

Fig 17.—Incidence of PTCA interventions in RAPID 2.

– Adapted from Bode et al.5

Patients(%)

0

10

20

30

40

50

60

23.9

Alteplase1.5 hr

(n = 155)

RETAVASE10 + 10 U (n = 169)

12.4*

57.4

Alteplase1.5 hr

(n = 155)

RETAVASE10 + 10 U (n = 169)

52.7†

*P < 0.01 vs Alteplase.†P =NS vs Alteplase.

Within 6 hours Overall

Page 35: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

Demonstrated mortality reduction/documented safety profile

Adouble-blind mortality trial hasdemonstrated that RETAVASE is effectivein reducing mortality following an AMI.3

Furthermore, patients treated with RETAVASE hada significantly lower incidence of atrial fibrillation,asystole, cardiogenic shock, CHF, and hypotension,and significantly fewer allergic reactions thanpatients treated with Streptokinase.3

With the exception of intracranial hemorrhagein which the incidence with RETAVASE was higher,the incidence of serious complications, such ascerebrovascular and cardiovascular events, wassimilar to that of control agents.

Restoration of coronary flow in more patients

RETAVASE has been angiographically proven to achieve complete perfusion and patency insignificantly more patients than the accelerated-dose Alteplase regimen.5 The correlation betweenpatency and patient outcome has not beenestablished. In addition, RETAVASE is proven to preserve left ventricular function.4,5

The GUSTO-III trial with 15,000 patients wasdesigned to study 30-day mortality with RETAVASEcompared to Alteplase. Enrollment was completedin January 1997. Secondary end points included in-hospital rates of reinfarction, CHF, stroke, andintracranial hemorrhage.

Simple, convenient administrationThe unique molecular structure of RETAVASE

and its long therapeutic half-life permit aconvenient double-bolus dosing regimen.Administration of RETAVASE as a double-bolusregimen—intravenous injections 30 minutesapart—may permit an earlier start of life-savingtherapy in the hospital emergency department orintensive care unit, and the dose does not have to beadjusted for the patient’s weight. The greater ease ofRETAVASE administration may reduce time andstaff required for administration and may alsoobviate the need for two intravenous lines, sinceheparinization can be interrupted duringRETAVASE bolus injections. Intravenous lines,however, should be flushed between administrationof RETAVASE and other agents.

SECTION IX

Summary of benefits ofRETAVASE® (Reteplase)

41

Page 36: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

SECTION X

References

1. Rapaport E. The ideal thrombolytic agent. In: Sleight P, Tavazzi L, eds. TheMajor Clinical Trials on Thrombolysis for Acute Myocardial Infarction. NewYork, NY: Raven Press; 1992:1-5.

2. Garabedian HD, Gold HK, Leinbach RC, et al. Comparative properties oftwo clinical preparations of recombinant human tissue-type plasminogenactivator in patients with acute myocardial infarction. J Am Coll Cardiol.1987;9:599-607.

3. International Joint Efficacy Comparison of Thrombolytics. Randomised,double-blind comparison of reteplase double-bolus administration withstreptokinase in acute myocardial infarction (INJECT): trial to investigateequivalence. Lancet. 1995;346:329-336.

4. Smalling RW, Bode C, Kalbfleisch J, et al. More rapid, complete, and stablecoronary thrombolysis with bolus administration of reteplase comparedwith alteplase infusion in acute myocardial infarction. Circulation.1995;91:2725-2732.

5. Bode C, Smalling RW, Berg G, et al. Randomized comparison of coronarythrombolysis achieved with double-bolus reteplase (recombinantplasminogen activator) and front-loaded, accelerated alteplase(recombinant tissue plasminogen activator) in patients with acutemyocardial infarction. Circulation. 1996;94:891-898.

6. American Heart Association. Heart and Stroke Facts: 1995 StatisticalSupplement. Dallas, Tex: American Heart Association; 1995:1,6,8.

7. DeWood MA, Spores J, Notske R, et al. Prevalence of total coronaryocclusion during the early hours of transmural myocardial infarction.N Engl J Med. 1980;303:897-902.

8. Fletcher JG. Acute myocardial infarction. In: Lilly LS, ed: Pathophysiology ofHeart Disease. Philadelphia, Pa: Lea & Febiger; 1993:113-129.

9. Tillett WS, Garner RL. The fibrinolytic activity of hemolytic streptococci.J Exp Med. 1933;58:485-502.

10. Fletcher AP, Sherry S, Alkjaersig N, et al. The maintenance of a sustainedthrombolytic state in man: II. clinical observations on patients withmyocardial infarction and other thromboembolic disorders. J Clin Invest.1959;38:1111-1119.

11. Schröder R, Biamino G, v. Leitner E-R, et al. Intravenous short-terminfusion of streptokinase in acute myocardial infarction. Circulation.1983;67:536-548.

12. Handin RI, Loscalzo J. Hemostasis, thrombosis, fibrinolysis, andcardiovascular disease. In: Braunwald E, ed. Heart Disease: A Textbook ofCardiovascular Medicine. 4th ed. Philadelphia, Pa: WB Saunders Co;1992:1767-1789.

13. Sobel BE, Hirsh J. Principles and practice of coronary thrombolysis andconjunctive treatment. Am J Cardiol. 1991;68:382-388.

14. Sobel BE, Nachowiak DA, Fry ETA, et al. Paradoxical attenuation offibrinolysis attributable to “plasminogen steal” and its implications forcoronary thrombolysis. Coron Artery Dis. 1990;1:111-119.

15. Pasternak RC, Braunwald E, Sobel BE. Acute myocardial infarction. In:Braunwald E, ed. Heart Disease: A Textbook of Cardiovascular Medicine. 4thed. Philadelphia, Pa: WB Saunders Co; 1992: 1200-1291.

16. Agnelli G. The pharmacological basis of thrombolytic therapy. In:Agnelli G, ed. Thrombolysis Yearbook 1995. Amsterdam: Excerpta Medica;1995:31-61.

17. The GUSTO Investigators. An international randomized trial comparingfour thrombolytic strategies for acute myocardial infarction. N Engl J Med.1993;329:673-682.

18. The GUSTO Angiographic Investigators. The effects of tissue plasminogenactivator, streptokinase, or both on coronary-artery patency, ventricularfunction, and survival after acute myocardial infarction.N Engl J Med. 1993;329:1615-1622.

19. Kohnert U, Rudolph R, Verheijen JH, et al. Biochemical properties of thekringle 2 and protease domains are maintained in the refolded t-PAdeletion variant BM 06.022. Protein Eng. 1992;5:93-100.

20. Martin U, von Möllendorff E, Akpan W, et al. Pharmacokinetic andhemostatic properties of the recombinant plasminogen activator BM06.022 in healthy volunteers. Thromb Haemost. 1991;66:569-574.

21. ISIS-2 (Second International Study of Infarct Survival) CollaborativeGroup. Randomised trial of intravenous streptokinase, oral aspirin, both,or neither among 17 187 cases of suspected acute myocardial infarction:ISIS-2. Lancet. 1988;2:349-360.

22. Jandl JH. Thrombotic and fibrinolytic disorders. In: Blood: Textbook ofHematology. Boston, Mass: Little, Brown and Co; 1987:1141-1175.

23. Hsia J, Hamilton WP, Kleiman N, et al, for the Heparin-AspirinReperfusion Trial (HART) Investigators. A comparison between heparinand low-dose aspirin as adjunctive therapy with tissue plasminogenactivator for acute myocardial infarction. N Engl J Med. 1990;323:1433-1437.

24. ISIS-3 (Third International Study of Infarct Survival) Collaborative Group.ISIS-3: a randomised comparison of streptokinase vs tissue plasminogenactivator vs anistreplase and of aspirin plus heparin vs aspirin alone among41 299 cases of suspected acute myocardial infarction. Lancet.1992;339:753-770.

25. Braunwald E. The open-artery theory is alive and well—again. N Engl JMed. 1993;329:1650-1652.

26. Braunwald E. The path to myocardial salvage by thrombolytic therapy.Circulation. 1987;76(suppl 2):II-2—II-7.

27. The TIMI Study Group. Special Report: The Thrombolysis in MyocardialInfarction (TIMI) trial: phase I findings. N Engl J Med. 1985;312:932-936.

28. Verstraete M, Bernard R, Bory M, et al. Randomised trial of intravenousrecombinant tissue-type plasminogen activator versus intravenousstreptokinase in acute myocardial infarction: report from the EuropeanCooperative Study Group for Recombinant Tissue-Type PlasminogenActivator. Lancet. 1985;1:842-847.

29. Gruppo Italiano per lo Studio della Streptochinasi nell’Infarto Miocardico(GISSI). Effectiveness of intravenous thrombolytic treatment in acutemyocardial infarction. Lancet. 1986;1:397-402.

30. Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto Miocardico.GISSI-2: a factorial randomised trial of alteplase versus streptokinase andheparin versus no heparin among 12 490 patients with acute myocardialinfarction. Lancet. 1990;336:65-71.

31. The International Study Group. In-hospital mortality and clinical course of20 891 patients with suspected acute myocardial infarction randomisedbetween alteplase and streptokinase with or without heparin. Lancet.1990;336:71-75.

32. Martin U, Bader R, Böhm E, et al. BM 06.022: a novel recombinantplasminogen activator. Cardiovasc Drug Rev. 1993;11:299-311.

33. Tanswell P, Seifried E, Su PCAF, et al. Pharmacokinetics and systemiceffects of tissue-type plasminogen activator in normal subjects. ClinPharmacol Ther. 1989;46:155-162.

34. Data on file, Boehringer Mannheim Corporation–Therapeutics Division.

35. Collen D, De Cock F, Demarsin E, et al. Absence of synergism betweentissue-type plasminogen activator (t-PA), single-chain urokinase-typeplasminogen activator (scu-PA) and urokinase on clot lysis in a plasmamilieu in vitro. Thromb Haemost. 1986;56:35-39.

36. Martin U, Sponer G, Strein K. Differential fibrinolytic properties of therecombinant plasminogen activator BM 06.022 in human plasma andblood clot systems in vitro. Blood Coagul Fibrinolysis. 1993;4:235-242.

37. Kanamasa K, Watanabe I, Cercek B, et al. Selective decrease in lysis of oldthrombi after rapid administration of tissue-type plasminogen activator.J Am Coll Cardiol. 1989;14:1359-1364.

38. Martin U, von Möllendorff E, Akpan W, et al. Dose-ranging study of thenovel recombinant plasminogen activator BM 06.022 in healthy volunteers.Clin Pharmacol Ther. 1991;50:429-436.

43

Page 37: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

was significantly higher with Retavase® than with Alteplase at 90 minutes after the initiation of therapy. In bothclinical trials the reocclusion rates were similar for Retavase® and Alteplase. The relationship between coronaryartery patency and clinical efficacy has not been established.

*p values represent one of multiple dose comparisons. Approximately 70% (RAPID 1) and 78% (RAPID 2) of the patients in the arteriographic studies under-

went optional arteriography at 60 minutes following the administration of the study agents. In both trials thepercentage of patients with complete flow at 60 minutes was significantly higher with Retavase® than withAlteplase. Neither RAPID clinical trial was designed nor powered to compare the efficacy or safety of Retavase® and Alteplase with respect to the outcomes of mortality and stroke. INDICATIONS AND USAGERetavase® (Reteplase) is indicated for use in the management of acute myocardial infarction (AMI) in adults for the improvement of ventricular function following AMI, the reduction of the incidence of congestive heartfailure and the reduction of mortality associated with AMI. Treatment should be initiated as soon as possibleafter the onset of AMI symptoms (see CLINICAL PHARMACOLOGY).CONTRAINDICATIONSBecause thrombolytic therapy increases the risk of bleeding, Retavase® is contraindicated in the followingsituations:

• Active internal bleeding• History of cerebrovascular accident• Recent intracranial or intraspinal surgery or trauma (see WARNINGS)• Intracranial neoplasm, arteriovenous malformation, or aneurysm• Known bleeding diathesis• Severe uncontrolled hypertension

WARNINGSBleeding

The most common complication encountered during Retavase® therapy is bleeding. The sites of bleed-ing include both internal bleeding sites (intracranial, retroperitoneal, gastrointestinal, genitourinary, or respira-tory) and superficial bleeding sites (venous cutdowns, arterial punctures, sites of recent surgical intervention).The concomitant use of heparin anticoagulation may contribute to bleeding. In clinical trials some of the hem-orrhage episodes occurred one or more days after the effects of Retavase® had dissipated, but while heparintherapy was continuing.

As fibrin is lysed during Retavase® therapy, bleeding from recent puncture sites may occur. Therefore,thrombolytic therapy requires careful attention to all potential bleeding sites (including catheter insertion sites,arterial and venous puncture sites, cutdown sites, and needle puncture sites). Noncompressible arterial punc-ture must be avoided and internal jugular and subclavian venous punctures should be avoided to minimizebleeding from noncompressible sites.

Should an arterial puncture be necessary during the administration of Retavase®, it is preferable touse an upper extremity vessel that is accessible to manual compression. Pressure should be applied for atleast 30 minutes, a pressure dressing applied, and the puncture site checked frequently for evidence of bleeding.

Intramuscular injections and nonessential handling of the patient should be avoided during treatmentwith Retavase®. Venipunctures should be performed carefully and only as required.

Should serious bleeding (not controllable by local pressure) occur, concomitant anticoagulant therapyshould be terminated immediately. In addition, the second bolus of Retavase® should not be given if seriousbleeding occurs before it is administered.

Each patient being considered for therapy with Retavase® should be carefully evaluated and anticipat-ed benefits weighed against the potential risks associated with therapy. In the following conditions, the risksof Retavase® therapy may be increased and should be weighed against the anticipated benefits:

• Recent major surgery, e.g., coronary artery bypass graft, obstetrical delivery, organ biopsy • Previous puncture of noncompressible vessels• Cerebrovascular disease• Recent gastrointestinal or genitourinary bleeding• Recent trauma• Hypertension: systolic BP ≥180 mm Hg and/or diastolic BP ≥110 mm Hg• High likelihood of left heart thrombus, e.g., mitral stenosis with atrial fibrillation• Acute pericarditis• Subacute bacterial endocarditis• Hemostatic defects including those secondary to severe hepatic or renal disease• Severe hepatic or renal dysfunction• Pregnancy• Diabetic hemorrhagic retinopathy or other hemorrhagic ophthalmic conditions• Septic thrombophlebitis or occluded AV cannula at a seriously infected site• Advanced age• Patients currently receiving oral anticoagulants, e.g., warfarin sodium• Any other condition in which bleeding constitutes a significant hazard or would be particularly

difficult to manage because of its location

Cholesterol EmbolizationCholesterol embolism has been reported rarely in patients treated with thrombolytic agents; the true incidenceis unknown. This serious condition, which can be lethal, is also associated with invasive vascular procedures(e.g., cardiac catheterization, angiography, vascular surgery) and/or anticoagulant therapy. Clinical features ofcholesterol embolism may include livedo reticularis, “purple toe” syndrome, acute renal failure, gangrenousdigits, hypertension, pancreatitis, myocardial infarction, cerebral infarction, spinal cord infarction, retinalartery occlusion, bowel infarction, and rhabdomyolysis. ArrhythmiasCoronary thrombolysis may result in arrhythmias associated with reperfusion. These arrhythmias (such assinus bradycardia, accelerated idioventricular rhythm, ventricular premature depolarizations, ventricular tachy-cardia) are not different from those often seen in the ordinary course of acute myocardial infarction andshould be managed with standard antiarrhythmic measures. It is recommended that antiarrhythmic therapyfor bradycardia and/or ventricular irritability be available when Retavase® is administered.

DESCRIPTIONRetavase® (Reteplase) is a non-glycosylated deletion mutein of tissue plasminogen activator (tPA), containingthe kringle 2 and the protease domains of human tPA. Retavase® contains 355 of the 527 amino acids of native tPA (amino acids 1-3 and 176-527). Retavase® is produced by recombinant DNA technology in E. coli. The protein is isolated as inactive inclusion bodies from E. coli, converted into its active form by anin vitro folding process and purified by chromatographic separation. The molecular weight of Reteplase is39,571 daltons.

Potency is expressed in units (U) using a reference standard which is specific for Retavase® and is notcomparable with units used for other thrombolytic agents.

Retavase® is a sterile, white, lyophilized powder for intravenous bolus injection after reconstitution withSterile Water for Injection, USP (without preservatives) provided as part of a kit. Following reconstitution, thepH is 6.0 ± 0.3. Retavase® is supplied as a 10.4 U vial to ensure sufficient drug for administration of each 10 U dose. Each single-use vial contains:

10.4 U (18.1 mg) VialReteplase 18.1 mgTranexamic Acid 8.32 mgDipotassium Hydrogen Phosphate 136.24 mgPhosphoric Acid 51.27 mg

CLINICAL PHARMACOLOGYGeneralRetavase® is a recombinant plasminogen activator which catalyzes the cleavage of endogenous plasminogen togenerate plasmin. Plasmin in turn degrades the fibrin matrix of the thrombus, thereby exerting its thrombolyticaction.1,2 In a controlled trial, 36 of 56 patients treated for an acute myocardial infarction (AMI) had a decreasein fibrinogen levels to below 100 mg/dL by 2 hours following the administration of Retavase® as a double-bolus intravenous injection (10 + 10 U) in which 10 U (17.4 mg) was followed 30 minutes later by a secondbolus of 10 U (17.4 mg).

3The mean fibrinogen level returned to the baseline value by 48 hours.

PharmacokineticsBased on the measurement of thrombolytic activity, Retavase® is cleared from plasma at a rate of 250-450mL/min, with an effective half-life of 13-16 minutes. Retavase® is cleared primarily by the liver and kidney. Clinical StudiesThe safety and efficacy of Retavase® were evaluated in three controlled clinical trials in which Retavase® wascompared to other thrombolytic agents. The INJECT study was designed to assess the relative effects ofRetavase® or the Streptase® brand of Streptokinase upon mortality rates at 35 days following an AMI. Theother studies (RAPID 1 and RAPID 2) were arteriographic studies which compared the effect on coronarypatency of Retavase® to two regimens of Alteplase (a tissue plasminogen activator; Activase® in the USA andActilyse® in Europe) in patients with an AMI. In all three studies, patients were treated with aspirin (initialdoses of 160 mg to 350 mg and subsequent doses of 75 mg to 350 mg) and heparin (a 5,000 U IV bolusprior to the administration of Retavase®, followed by a 1000 U/hour continuous IV infusion for at least 24 hours).

3,4,5The safety and efficacy of Retavase® have not been evaluated using antithrombotic or

antiplatelet regimens other than those described above. Retavase® (10 + 10 U) was compared to Streptokinase (1.5 million units over 60 minutes) in a double-

blind, randomized, European study (INJECT), which studied 6,010 patients treated within 12 hours of theonset of symptoms of AMI. To be eligible for enrollment, patients had to have chest pain consistent withcoronary ischemia and ST segment elevation, or a bundle branch block pattern on the EKG. Patients withknown cerebrovascular or other bleeding risks or those with a systolic blood pressure >200 mm Hg or a diastolic blood pressure >100 mm Hg were excluded from enrollment. The results of the primary endpoint(mortality at 35 days), six month mortality and selected other 35 day endpoints are shown in Table 1 forpatients receiving study medications.

*p value for the exploratory analysis comparing Retavase® versus Streptokinase.†Kaplan-Meier estimates.

For mortality, stroke and the combined outcome of mortality or stroke, the 95% confidence intervals inTable 1 reflect the range within which the true difference in outcomes probably lies and includes the possibilityof no difference. The incidences of congestive heart failure and of cardiogenic shock were significantly loweramong patients treated with Retavase®.

The total incidence of stroke was similar between the groups. However, more patients treated withRetavase® experienced hemorrhagic strokes than patients treated with Streptokinase. An exploratory analysisindicated that the incidence of intracranial hemorrhage was higher among older patients or those with elevatedblood pressure. The incidence of intracranial hemorrhage among the 698 patients treated with Retavase® whowere older than 70 years was 2.2%. Intracranial hemorrhage occurred in 8 of the 332 (2.4%) patients treatedwith Retavase® who had an initial systolic blood pressure >160 mm Hg and in 15 of the 2,629 (0.6%)Retavase® patients who had an initial systolic blood pressure <160 mm Hg.

Two arteriographic studies (RAPID 1 and RAPID 2) were performed utilizing open-label administrationof the study agents and a blinded review of the arteriograms. In RAPID 1, patients were treated within 6 hoursof the onset of symptoms, and in RAPID 2, patients were treated within 12 hours of the onset of symptoms.Both studies evaluated coronary artery perfusion through the infarct-related artery 90 minutes after the initia-tion of therapy as the primary endpoint. Some patients in each study also had perfusion through the infarct-related artery evaluated at 60 minutes after the initiation of therapy. In RAPID 1, Retavase® (in doses of 10 + 10 U, 15 U, or 10 + 5 U) was compared to a 3 hour regimen of Alteplase (100 mg administered over 3 hrs). InRAPID 2, Retavase® (10 + 10 U) was compared to an accelerated regimen of Alteplase (100 mg administeredover 1.5 hrs). The percentages of patients with partial or complete flow (TIMI grades 2 or 3) and completeflow (TIMI grade 3), are shown along with ventricular function assessments in Table 2. The follow-up arteriogram was performed at a median of 8 (RAPID 1) and 5 (RAPID 2) days following the administration ofthe thrombolytics. In RAPID 1 the best patency results were obtained with the 10 + 10 U dose. In RAPID 2,the percentage of patients with partial or complete flow and the percentage of patients with complete flow

Table 1INJECT TRIAL

Incidence of Selected Outcomes

Endpoint Retavase® Streptokinase Retavase®-Streptokinase pn = 2,965 n = 2,971 difference (95% CI) Value

35 Day mortality 8.9% 9.4% -0.5 (-2.0, 0.9) 0.49*

6 Month mortality† 11.0% 12.1% -1.1 (-2.7, 0.6) 0.22

Combined outcome of 9.6% 10.2% -0.6 (-2.1, 1.0) 0.4735 day mortality or nonfatal stroke within 35 days

Heart failure 24.8% 28.1% -3.3 (-5.6, -1.1) 0.004

Cardiogenic shock 4.6% 5.8% -1.2 (-2.4, -0.1) 0.03

Any stroke 1.4% 1.1% 0.3 (-0.3, 0.8) 0.34

Intracranial hemorrhage 0.8% 0.4% 0.4 (0.0, 0.8) 0.04

Table 2RAPID 1 and RAPID 2 TRIALS

Arteriographic Results

Outcome RAPID 2 RAPID 1*

Retavase® Alteplase p Retavase® Alteplase p(10 +10 U) (Accelerated (10 +10 U) (Standard

regimen) regimen)

90 minute patency rates n = 157 n = 146 n = 142 n = 145

TIMI 2 or 3 83% 73% 0.03 85% 77% 0.08TIMI 3 60% 45% 0.01 63% 49% 0.02

Follow-up patency rates n = 128 n = 113 n = 123 n = 123

TIMI 2 or 3 89% 90% 0.76 95% 88% 0.040TIMI 3 75% 77% 0.72 88% 71% 0.001

Follow-up ejection fraction n = 89 n = 77 n = 91 n = 84

mean % 52% 54% 0.25 53% 49% 0.03

Follow-up regional wall motion n = 87 n = 72 n = 84 n = 80

Standard deviation from -2.3 -2.3 0.96 -2.2 -2.6 0.02mean normal value

10.4 U (18.1 mg) VialReteplase 18.1 mgTranexamic Acid 8.32 mgDipotassium Hydrogen Phosphate 136.24 mgPhosphoric Acid 51.27 mgSucrose 364.0 mgPolysorbate 80 5.20 mg

Page 38: 98CNTP572.v4 content [mono] (Page 1) · thrombolytic agents then available could be considered ideal. Each had limitations. Therefore, it is appropriate to define the characteristics

PRECAUTIONSGeneralStandard management of myocardial infarction should be implemented concomitantly with Retavase®

treatment. Arterial and venous punctures should be minimized (see WARNINGS). In addition, the secondbolus of Retavase® should not be given if the serious bleeding occurs before it is administered. In the eventof serious bleeding, any concomitant heparin should be terminated immediately. Heparin effects can bereversed by protamine. ReadministrationThere is no experience with patients receiving repeat courses of therapy with Retavase®. Retavase® did notinduce the formation of Retavase® specific antibodies in any of the approximately 2,400 patients who weretested for antibody formation in clinical trials. If an anaphylactoid reaction occurs, the second bolus ofRetavase® should not be given, and appropriate therapy should be initiated.Drug InteractionsThe interaction of Retavase® with other cardioactive drugs has not been studied. In addition to bleeding associated with heparin and vitamin K antagonists, drugs that alter platelet function (such as aspirin, dipyri-damole, and abciximab) may increase the risk of bleeding if administered prior to or after Retavase® therapy. Drug/Laboratory Test InteractionsAdministration of Retavase® may cause decreases in plasminogen and fibrinogen. During Retavase® therapy,if coagulation tests and/or measurements of fibrinolytic activity are performed, the results may be unreliableunless specific precautions are taken to prevent in vitro artifacts. Retavase® is an enzyme that when present in blood in pharmacologic concentrations remains active under in vitro conditions. This can lead to degradation of fibrinogen in blood samples removed for analysis. Collection of blood samples in the presenceof PPACK (chloromethylketone) at 2 µM concentrations was used in clinical trials to prevent in vitrofibrinolytic artifacts.6

Use of AntithromboticsHeparin and aspirin have been administered concomitantly with and following the administration of Retavase® in the management of acute myocardial infarction. Because heparin, aspirin, or Retavase® maycause bleeding complications, careful monitoring for bleeding is advised, especially at arterial puncture sites.Carcinogenesis, Mutagenesis, Impairment of FertilityLong-term studies in animals have not been performed to evaluate the carcinogenic potential of Retavase®.Studies to determine mutagenicity, chromosomal aberrations, gene mutations, and micronuclei inductionwere negative at all concentrations tested. Reproductive toxicity studies in rats revealed no effects on fertilityat doses up to 15 times the human dose (4.31 U/kg). Pregnancy Category CReteplase has been shown to have an abortifacient effect in rabbits when given in doses 3 times the humandose (0.86 U/kg). Reproduction studies performed in rats at doses up to 15 times the human dose (4.31U/kg) revealed no evidence of fetal anomalies; however, Reteplase administered to pregnant rabbits resultedin hemorrhaging in the genital tract, leading to abortions in mid-gestation. There are no adequate and well-controlled studies in pregnant women. The most common complication of thrombolytic therapy is bleedingand certain conditions, including pregnancy, can increase this risk. Reteplase should be used during pregnancy only if the potential benefit justifies the potential risk to the fetus. Nursing MothersIt is not known whether Retavase® is excreted in human milk. Because many drugs are excreted in humanmilk, caution should be exercised when Retavase® is administered to a nursing woman.Pediatric UseSafety and effectiveness of Retavase® in pediatric patients have not been established.ADVERSE REACTIONSBleedingThe most frequent adverse reaction associated with Retavase® is bleeding (see WARNINGS). The types ofbleeding events associated with thrombolytic therapy may be broadly categorized as either intracranial hemor-rhage or other types of hemorrhage. • Intracranial hemorrhage (see CLINICAL PHARMACOLOGY)

In the INJECT clinical trial the rate of in-hospital, intracranial hemorrhage among all patientstreated with Retavase® was 0.8% (23 of 2,965 patients). As seen with Retavase® and other throm-bolytic agents, the risk for intracranial hemorrhage is increased in patients with advanced age or withelevated blood pressure.

• Other types of hemorrhageThe incidence of other types of bleeding events in clinical studies of Retavase® varied depending upon the use of arterial catheterization or other invasive procedures and whether the study was per-formed in Europe or the USA. The overall incidence of any bleeding event in patients treated withRetavase® in clinical studies (n = 3,805) was 21.1%. The rates for bleeding events, regardless ofseverity, for the 10 + 10 U Retavase® regimen from controlled clinical studies are summarizedin Table 3.

*includes the arterial catheterization site (all patients in the RAPID studies underwent arterial catheterization).

In these studies the severity and sites of bleeding events were comparable for Retavase® and thecomparison thrombolytic agents.

Should serious bleeding in a critical location (intracranial, gastrointestinal, retroperitoneal, pericardial)occur, any concomitant heparin should be terminated immediately. In addition, the second bolus of Retavase® should not be given if the serious bleeding occurs before it is administered. Death and permanentdisability are not uncommonly reported in patients who have experienced stroke (including intracranial bleed-ing) and other serious bleeding episodes.

Fibrin which is part of the hemostatic plug formed at needle puncture sites will be lysed duringRetavase® therapy. Therefore, Retavase® therapy requires careful attention to potential bleeding sites (e.g.,catheter insertion sites, arterial puncture sites). Allergic ReactionsAmong the 2,965 patients receiving Retavase® in the INJECT trial, serious allergic reactions were noted in 3patients, with one patient experiencing dyspnea and hypotension. No anaphylactoid reactions were observedamong the 3,856 patients treated with Retavase® in initial clinical trials. In an ongoing clinical trial two ana-phylactoid reactions have been reported among approximately 2,500 patients receiving Retavase®.Other Adverse ReactionsPatients administered Retavase® as treatment for myocardial infarction have experienced many events whichare frequent sequelae of myocardial infarction and may or may not be attributable to Retavase® therapy.These events include cardiogenic shock, arrhythmias (e.g., sinus bradycardia, accelerated idioventricular

rhythm, ventricular premature depolarizations, supraventricular tachycardia, ventricular tachycardia, ventricu-lar fibrillation), AV block, pulmonary edema, heart failure, cardiac arrest, recurrent ischemia, reinfarction,myocardial rupture, mitral regurgitation, pericardial effusion, pericarditis, cardiac tamponade, venous throm-bosis and embolism, and electromechanical dissociation. These events can be life-threatening and may leadto death. Other adverse events have been reported, including nausea and/or vomiting, hypotension, and fever.DOSAGE AND ADMINISTRATIONRetavase® (Reteplase) is for intravenous administration only. Retavase® is administered as a 10 +10 U double-bolus injection. Each bolus is administered as an intravenous injection over 2 minutes. The secondbolus is given 30 minutes after initiation of the first bolus injection. Each bolus injection should be given via anintravenous line in which no other medication is being simultaneously injected or infused. No other medicationshould be added to the injection solution containing Retavase®. There is no experience with patients receivingrepeat courses of therapy with Retavase®. Heparin and Retavase® are incompatible when combined in solution. Do not administer heparin andRetavase® simultaneously in the same intravenous line. If Retavase® is to be injected through an intravenousline containing heparin, a normal saline or 5% dextrose (D5W) solution should be flushed through the lineprior to and following the Retavase® injection.

Although the value of anticoagulants and antiplatelet drugs during and following administration ofRetavase® has not been studied, heparin has been administered concomitantly in more than 99% of patients.Aspirin has been given either during and/or following heparin treatment. Studies assessing the safety and effi-cacy of Retavase® without adjunctive therapy with heparin and aspirin have not been performed.ReconstitutionReconstitution should be carried out using the diluent, syringe, needle and dispensing pin provided withRetavase®. It is important that Retavase® be reconstituted only with Sterile Water for Injection, USP (withoutpreservatives). The reconstituted preparation results in a colorless solution containing Retavase® 1 U/mL.Slight foaming upon reconstitution is not unusual; allowing the vial to stand undisturbed for several minutesis usually sufficient to allow dissipation of any large bubbles.

Because Retavase® contains no antibacterial preservatives, it should be reconstituted immediatelybefore use. When reconstituted as directed, the solution may be used within 4 hours when stored at 2-30˚C(36-86˚F). Prior to administration, the product should be visually inspected for particulate matter anddiscoloration. Reconstitution InstructionsUse aseptic technique throughout.Step 1: Remove the protective flip-cap from one vial of Sterile Water for Injection, USP (SWFI).

Open the package containing the 10 cc syringe with attached needle. Remove the protective cap from the needle and withdraw 10 mL of SWFI from the vial.

Step 2: Open the package containing the dispensing pin. Remove the needle from the syringe, discard the needle. Remove the protective cap from the luer lock port of the dispensing pin and connect the syringe to the dispensing pin.Remove the protective flip-cap from one vial of Retavase®.

Step 3: Remove the protective cap from the spike end of the dispensing pin, and insert the spike into the vialof Retavase® until the security clips lock onto the vial. Transfer the 10 mL of SWFI through the dispensing pin into the vial of Retavase®.

Step 4: With the dispensing pin and syringe still attached to the vial, swirl the vial gently to dissolve theRetavase®. DO NOT SHAKE.

Step 5: Withdraw 10 mL of Retavase® reconstituted solution back into the syringe. A small amount of solutionwill remain in the vial due to overfill.

Step 6: Detach the syringe from the dispensing pin, and attach the sterile 20 gauge needle provided. Step 7: The 10 mL bolus dose is now ready for administration.

Safely discard all used reconstitution components and the empty Retavase® vial according to institutional procedures. HOW SUPPLIEDRetavase®, is supplied as a sterile, preservative-free, lyophilized powder in 10.4 U (18.1 mg) vials without avacuum, in a kit with components for reconstitution. Each kit contains a package insert, 2 single-useRetavase® vials 10.4 U (18.1 mg), 2 single-use diluent vials for reconstitution (10 mL Sterile Water forInjection, USP), 2 sterile 10 cc syringes with 20 G needle attached, 2 sterile dispensing pins, 2 sterile 20 Gneedles for dose administration, and 2 alcohol swabs. NDC 57894-040-01. StorageStore the kit containing Retavase® at 2-25˚C (36-77˚F). Kit should remain sealed until use to protect thelyophilisate from exposure to light. Do not use beyond expiration date printed on the kit. References1. Martin U, Sponer G, Strein K. Evaluation of thrombolytic and systemic effects of the novel recombi-

nant plasminogen activator BM 06.022 compared with alteplase, anistreplase, streptokinase andurokinase in a canine model of coronary artery thrombosis. JACC. 1992;19:433-440.

2. Kohnert U, Rudolph R, Verheijen JH. Biochemical properties of the kringle 2 and protease domains aremaintained in the refolded t-PA deletion variant BM 06.022. Protein Engineering. 1992;5:93-100.

3. Smalling R, Bode C, Kalbfleisch J, et al. More rapid, complete, and stable coronary thrombolysis withbolus administration of reteplase compared with alteplase infusion in acute myocardial infarction.Circulation. 1995;91:2725-2732.

4. Bode C, Smalling R, Gunther B, et al. Randomized comparison of coronary thrombolysis achievedwith double bolus reteplase (recombinant plasminogen activator) and front-loaded, acceleratedalteplase (recombinant tissue plasminogen activator) in patients with acute myocardial infarction.Circulation. 1996;94:891-898.

5. INJECT Study Group. Randomised, double-blind comparison of reteplase double-bolus administrationwith streptokinase in acute myocardial infarction (INJECT): trial to investigate equivalence. Lancet.1995;346:329-336.

6. Martin U, Gärtner D, Markl HJ, et al. D-PHE-PRO-ARGCHLOROMETHYLKETONE prevents in vitro fib-rinogen reduction by the novel recombinant plasminogen activator BM 06.022. Ann Hematol. 1992;64(suppl)A47.

Retavase®,Reteplase,recombinantManufactured by:Boehringer Mannheim GmbHNonnenwald 2D-82377 PenzbergGermany

For:Centocor, Inc.Malvern, PA 19355U.S. License Number 1242

Table 3Retavase® Hemorrhage Rates

Bleeding Site INJECT RAPID 1 and RAPID 2

Europe USA Europen = 2,965 n = 210 n =113

Injection Site* 4.6% 48.6% 19.5%

Gastrointestinal 2.5% 9.0% 1.8%

Genitourinary 1.6% 9.5% 0.9%

Anemia, site unknown 2.6% 1.4% 0.9%

© 1999 Centocor, Inc. Printed in U.S.A. January 1999 RP-98041

January 1999 RP-98041


Recommended