Progress VOL XXXIV,

in

Cardiovascular

Diseases

NO 2

SEPTEMBER/OCTOBER

Thrombosis,

Fibrinoiysis, and Thrombolytic A Perspective

1991

Therapy:

Sol Sherry and Victor J. Marder

T

HROMBOSIS as a pathological entity is second to none in importance as an acute cause of serious disability and death. Furthermore, this importance will continue to increase as the average age of the population increases. Nevertheless, although a most serious public health problem, thrombosis does not receive the attention it deserves at the clinical Ievel. This is because the clinically oriented specialist views it as an independent finding within his/her domain of interest, rather than an entity that can complicate the vascular system at any site. Although there are attempts to develop a specialty of vascular medicine that could help view thrombosis as a holistic entity, it is likely that such vascular disease specialists will find it difficult to compete with the organ-oriented practitioners for the care of patients, except perhaps for peripheral vascular problems, or to significantly enhance basic research on thrombosis. The growth and development of thrombosis research in recent years has been remarkable and this has led to a balance between research and clinical practice of hemostasis and thrombosis by skilled investigators and physicians that exceeds any that existed before. Thrombosis is a very complex phenomenon; not only can thrombi differ in composition and be initiated by different mechanisms, but the process involves an extraordinary number of interrelationships between the vessel wall, platelets, the coagulation mechanism, various activators and inhibitors, the dynamics of blood flow, and the involvement of the plasminogen-plasmin enzyme system. There is no orderly process; rather, there are multiple actions going on simultaneously with modulating systems, in turn, influenced by various factors that determine Progress

in Cardiovascular

Diseases,

Vol XXXIV,

No 2 (September/October),

their contributions to the overall process. Thus, the speed, extent, and disposition of the thrombotic process is likely to differ from case to case and from organ to organ. If this complicated situation is to be understood, it must be separated into appropriate test models, each of which could be individually understood, then woven back into the fabric of the fully reconstituted, complex entity that is the hemostatic/ thrombotic system. One such part of the whole is the plasminogenplasmin enzyme system. Recent advances in thrombolytic therapy have stimulated basic research, which in turn has led to further improvements in therapeutics. The current status of physiological and clinical aspects in this field constitutes the focus for this symposium, the plasminogen-plasmin enzyme system now being recognized as a major modulator of the thrombotic process. Indeed, the thrombus is best viewed as being in a dynamic state between two opposing forces: continuing thrombus formation and thrombus resolution. It is the balance between these forces that determines the fate of the thrombus and the clinical outcome of the patient.

From the Department of Medicine, Temple University School of Medicine, Philadelphia, PA; and the Hematology Unit, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, Nk: Supported in purt by Grant No. HL-30616, National Heart, Lung and Blood Institute, National Instifu&es of Health, Bethesda, MD. Address reprint requests to Sol Sherry, MD, Distinguished Professor of ,Medicine, 4th Floor SFC, Temple Universiry School of Medicine, 3400 N Broad St, Philade?phia, PA 19140. Copyright o 1991 by W.B. Saunders Company 0033-0620/91/3402-0001$5.00/O 1991:

pp 89-100

89

90

SHERRY

The purpose of this symposium is to review the biology of the human plasminogen-plasmin enzyme system within the vascular tree and to summarize clinical results of its pharmacological activation in a variety of thrombotic disorders. Although usualIy referred to as the fibrinolytic enzyme system, its action under physiological circumstances is not restricted to the lysis of fibrin nor is it restricted to the vasculature. Plasminogen is ubiquitous in distribution, binding to many surfaces possessing specific binding sites. Local activation to plasmin can result in proteolysis of several important native substrates, for example, collagen and platelet membrane receptors, among others. In truth, the plasminogen-plasmin enzyme system represents a major endogenous proteolytic system used by the body for a number of regulatory functions, one of which is concerned with thrombus dissolution. THE BACKGROUND

PERIOD (1945-1959)

Knowledge of the “instability” of blood clots has existed for over 150 years’ but it was Dastre, a half century later, who attributed this to fibrinolysis.* There was very slow progress in understanding this phenomenon, although this was taken advantage of in Russia by Yudin for providing incoagulable blood for transfusion purposes.3 Three observations were responsibIe for opening up this field of investigation and for initiating the explosion of knowledge that continues to this day. First, Tillett and Garner discovered that hemolytic streptococci secrete a substance that mediates the rapid lysis of human blood or plasma clots.4 Originally named streptococcal fibrinolysin, but subsequently renamed streptokinase, this substance was the first to have a demonstrated capacity to dissolve fibrin rapidly. Second, Milstone noted that streptococcal fibrinolysin did not dissolve a clot made with purified fibrinogen and thrombin but did dissolve a clot formed in the presence of human protein found in plasma “euglobulin.“5 The required plasma protein was named “plasma lysing factor.” Third, Christensen and Kaplan7 independently showed that the “plasma lysing factor” is the inactive precursor (plasminogen) of the proteolytic enzyme (plasmin) and that this enzyme. is activated in the presence of streptokinase. Christensen’s work was the more

AND

MARDER

extensive and involved characterization of the various components, including the plasma inhibitor, antiplasmin.‘*’ The terminology for this system, proposed in 1945 by Christensen and MacLoedT has remained in effect ever since. The ability to rapidIy and strikingly induce fibrinolysis by activation of this native enzyme system spurred further experimentation that followed several directions. The physiological approach was initiated by Macfarlane and Biggs’ and subsequently pursued by Kwaan and McFadzean”; the study of tissue distribution and disposition of native tissue activators was initiated by Astrup” and subsequentIy by Todd”; pharmacologic studies were begun by von Kaulla13; and therapeutic application was pioneered by Tillett and Sherry and their associates.‘4-L6 Although phenomenology dominated general coagulation research for decades, much of the early work on the plasminogen-plasmin system used biochemical techniques, which were ideally suited for the study of enzymes. This approach fostered knowledge on the isolation, purification and characterization of components,‘7-L9 kinetics of the reactions? assay of components, including methods using synthetic substrates,*’ and the basis for which plasminogen activators rather than plasmin serve best for therapeutic purposes?* In 1959, a major review of this early work provided a conceptual basis for the action of this system under physiological conditions and for its manipulation for therapeutic purposes.23 THE PERIOD OF EXPANDING RESEARCH AND THERAPEUTIC DEVELOPMENT (1960-1979)

The introduction of a potentially powerful new treatment for thrombotic and embolic disorders, namely rapid dissolution of the offending vascular obstruction by administration of a plasminogen activator such as streptokinase, now made it possible for improved management of all forms of thrombo-occlusive disease. In turn, this therapeutic opportunity excited new interest in the basic aspects of the plasminogen-plasmin enzyme system and in its therapeutic evaluation and application. This also was a period of major technical development in the laboratory (chromatography for protein purification and characterization, peptide analyzers, ultracentrifugation, tissue culture, conventional

THROMBOSIS

AND

FIBRINOLYSIS

PERSPECTIVE

and scanning electron microscopy, gel electrophoresis, immunologic identification) and in the clinic (angiography, venography, intravascular catheterization, etc), which made it possible to expand the information pool qualitatively and quantitatively. During this period there was further clarification of the details of the fundamental mechanisms of thrombolysis,24 striking advances in the biochemistry of plasminogen and its activation,Z-34 description of an intrinsic system for plasminogen activation that complemented the more obvious extrinsic system,3s3’ elucidation of the role of streptokinase in mediating the activation of plasminogen,29334 purification and characterization of tissue plasminogen activator,3*-40 isolation and characterization of native inhibitors to plasmin,41-44 and elucidation of fibrinogen structure, its fragmentation by plasmin and the effect of these products on blood coagulation4’-” The detailed biochemical and molecular data of this period provided support and further insights for the broad conceptual outlines that were formulated in a major review published in 1959.23 At that time, there was recognition of the local orientation of the plasminogenplasmin enzyme system within the vascular bed. According to this understanding, fibrin deposition, anoxia, or vasoactive substances stimulated endothelial cell release of activator into the involved area. Because the concentration of endothelial cells per unit of vascular volume in small vessels was much greater than in large vessels, fibrinolytic activity in the capillary bed was more intense than in the larger vessels. This explained why thrombus formation was more common in arteries and veins, because the same thrombogenic stimulus would easily outbalance the fibrinolytic response. In contrast, fibrin formed within the capillary bed due to local vascular injury or filtered out by the microcirculation after formation in the blood is subjected to high concentrations of plasminogen activator, resulting in prompt lysis of the fibrin and reperfusion of the capillaries. It was apparent that the total plasmin inhibitory activity of plasma far exceeds the plasmin concentration that could be produced by such local activation of plasminogen. Thus the presumed role for the antiplasmin(s) was to prevent the escape of plasmin into the systemic circulation. A similar

91

role probably exists for the inhibitors of endogenously released plasminogen activators (PAIs). This mechanism helps to circumscribe the action of the plasminogen-plasmin system to sites of vascular injury or vascular involvement, even while potent fibrinolytic activity is induced locally. As for the ability of plasma to dissolve fibrin (fibrinolysis) or a thrombus (thrombolysis), this is due to the activator concentration of plasma not to its plasmin contem2’ This special selectivity of the plasminogen-plasmin system toward fibrin dissolution without inducing fibrinogen proteolysis by plasmin is due to the binding of plasminogen to fibrin. The amount of plasmaplasminogen bound to fibrin during clot formation is relatively small (only about 4% of the available plasma plasminogen), but when converted to plasmin directly on the fibrin surface, there are no other substrates available to compete for its action and no effective neutralization of its action by plasmin inhibitors. Thus the notable feature of this enzyme system in vivo is not a selective action of plasmin but rather a selective binding of plasminogen to fibrin. Under physiological conditions this provides a mechanism by which plasmin restricts its effects to one substrate (fibrin) and in a relatively inhibitor-free environment. However, activity of this enzyme system in vivo is determined by the local concentration of plasminogen activator released by endothelial cells in response to local stimulatory events. Today we understand more clearly the molecular basis’* for the selective action of this nonspecific proteolytic enzyme system, a process that is finely tuned for enhancement on a surface, such as fibrin, but ineffective with regards to activation of plasminogen in plasma. The clinical advances kept pace with the biochemistry, physiology, and pharmacology, with notable contributions including development of the first nonantigenic thrombolytic agent (urokinase) for clinical medicine,53”5 introduction of antifibrinolytic agents for the treatment of various bleeding disorders,5b-64discovery of prourokinase,“.” extensive controlled trials with streptokinase and urokinase for acute myocardial infarction, pulmonary embolism and deep vein thrombosis,6”73 and formal approval of plasminogen activators for general clinical use.74 These important studies laid the founda-

92

SHERRY

tion for the current phase of greatly expanded clinical progress, primarily the result of involvement by the cardiology community. THE COMING OF AGE FOR THROMBOSIS RESEARCH AND THE EXPLOSION OF CLINICAL TRIALS (1980-1990)

Of greatest importance in the surge of information regarding the therapeutic potential of thrombolytic agents was the rediscovery of coronary thrombosis as the responsible cause of the sustained ischemic event underlying acute myocardial infarction” and the direct angiographic evidence that either local or systemic infusion of a plasminogen activator induces early reperfusion.76.77With the evident advantage of rapid delivery of agent afforded by intravenous administration, data quickly followed that clearly demonstrated functional and clinical benefit in the form of limitation of infarct size78-80and reduced mortality.79~81-85Because acute myocardial infarction is second to none as a major public health problem in the aging population, thrombolytic therapy for this disorder has gained worldwide recognition and has led to the development of second generation thrombolytic agents.86-90Each plasminogen activator has in turn spawned a unique area of research interest in relation to its pharmacology, clinical use, and, where appropriate, human biology. Again, the extraordinary progress during this period was made possible by new scientific techniques, the most striking of which was in the discipline of molecular biology involving recombinant DNA technology and immunology. New studies allowed for definition of PAI’s,~~-~~more accurate and specific measurements of circulating activators and inhibitors and their complexes95-101and of antifibrinolytic activities in response to physiological and abnormal stimulastructural analyses of the proteins,31’87.‘08-“2 ~ion,1me’07 relationships of plasminogen and plasminogen activator on vascular surfaces and understanding of the effects of plasmin formation at these sites,“3~‘z’ identification of hereditary molecular deficiency or dysfunctional states and their clinical effects,1u-129 improvements in drug design for second and third generation thrombolytic agents,Bp90and the preparation of large quantities of highly purified native activators for therapeutic purposes.87.13a’” Also, new drug regimens have been tested for the

AND

MARDER

treatment of pulmonary embolism135-137and peripheral arterial thrombo-occlusive events,138-142 and clinical investigations have been initiated in other coronary syndromes, such as unstable angina and non-Q wave infarctions. Insights from clinical trials have reaffirmed that aged thrombi are more resistant to lysis than fresh thrombi or emboli, as is now evident in the treatment of acute myocardial infarction,120~‘43and that bleeding is more the result of hemostatic plug dissolution,‘” less related to the extent of hypofibrinogenemia.w,7’,145S16 Other data have altered previously held concepts. For example, we now recognize that striking fibrinogenolysis may be an advantage in thrombolytic therapy rather than a disadvantage because it does not cause more severe or more frequent bleeding complications but is associated with a lower likelihood of rethrombosis.‘Z03’4”1a Furthermore, mortality reduction in myocardial infarction may not depend solely on infarct size reduction but may also depend on late reperfusion (after the infarct is completed)‘47~‘49~151and that 90minute patency rates have provided misleading information, because they do not correlate with observations on myocardial salvage or mortality reduction.‘47,‘52-‘55 There has been recognition of the importance of early rethrombosis as a significant problem in achieving lasting vascular patency and full clinical benefit. Early rethrombosis is not prevented by heparin,156 but early and continued heparin infusion and oral aspirin may both improve survival rates after acute myocardial infarction.83 None of the plasminogen activators administered via the intravenous route has documented clinical superiority in safety or efficacy over other agents for thrombotic disorders, especially for acute myocardial infarction, and each has its respective advantages and disadvantageS~0.120,152.157,158 One exception to this rule may exist, as urokinase has been claimed to be the for local perfusion of periphagent of choice’59X160 eral arterial thrombo-occlusions. Whichever agent is applied for acute myocardial infarction, the best therapy is applied early after symptom onset, because treatment initiated after the opportunity to salvage heart muscle achieves less impressive results in mortality reduction. To fulfill the potential that thrombolytic ther-

THROMBOSIS

AND

FIBRINOLYSIS

PERSPECTIVE

apy offers will require mechanisms for instituting therapy as quickly as possible. THE NEXT

DECADE

(1991-2000)

Impressive progress has been made in the last 45 years to realize the potential of plasminogen activators as valid therapeutic agents for all manner of thrombotic disease.12’ Yet, issues remain to be settled, therapy is far from ideal, and even accepted and proven indications are not fully applied to the patient population. There still is vigorous debate as to whether one plasminogen activator is preferable to another, despite the lack of clinical data to justify such a choice. Thus, a major challenge for the clinician wilI be to choose between three, four, or more excellent plasminogen activators and a multitude of regimens using activators with various anticoagulants, as all therapeutic options are not likely to have been compared with any single study. The action of plasminogen activators on the thrombus, blood, and hemostatic plug is shown in Fig 1, which schematically compares results obtained with the more “fibrinogen-sparing” (“fibrin-selective”) agents, such as rt-PA and prourokinase (scu-PA) with those that induce a marked degradation of plasma fibrinogen, such as streptokinase, urokinase, and anisoylated plasminogen-streptokinase activator complex (APSAC).“” For patients treated 3 or more hours after symptom onset, the more fibrinogensparing (fibrin-selective) agents induce reperfusion more rapidly than the agents that induce a potent lytic state. However, along with the lesser degree of lytic state associated with rt-PA therapy, there is also a greater tendency for rethrombosis and reocclusion and the cumulative patency rates of all of the agents at 3 hours14’ or at 24 hours’“’ after treatment induction are the same. Thus, the greatest need for improving the efficacy of streptokinase, urokinase, and APSAC is to achieve more rapid thrombolysis and reperfusion, whereas that for rt-PA is to achieve more lasting patency, free of the greater tendency for reocclusion. Although single-chain urokinase (scu-PA) similar to single-chain rt-PA has a fast action in vivo for lysing thrombi, it differs from rt-PA when administered intravenously in large amounts, for example 80 mg is equivalent to

93

12.4 million urokinase units. With such large doses, a portion of the activator is rapidly converted to urokinase in the circulation. The result is a systemic lytic state that is much more extensive than observed with rt-PA and a low reocclusion rate.162.‘63Whether amelioration of efficacy will result from better adjunct anticoagulant treatment, from a new regimen of plasminogen activator, from combined use of two plasminogen activators,‘64.‘” or from a new plasminogen activator altogether, awaits further study. New plasminogen activators include unique proteins that combine a catalytic site with an antifibrin antibody,“’ mutant deletions of rt-PA that have a prolonged half-life in blood,‘67 and a recombinant bat salivary gland plasminogen activator with an exceptionally high fibrinogen-sparing tendency.‘68 However, in vitro biochemical advantage and even superiority in animals or in human vascular reperfusion studies do not necessarily ensure functional or clinical superiority of such innovative agents over existing activators. An especially recalcitrant negative aspect of treatment is the increased risk of hemorrhage over and above that which attends heparin treatment.74.‘69”70 None of the agents thus far used in human disease lacks this bleeding propensity. Although an agent with exceptionally high fibrinogen-sparing capacity has not yet been available for treatment and theoretically could be safer, it seems unlikely that hemostatic plugs will resist their action and protect against bleeding from sites of vascular injury.‘58 The most dreaded complication is intracranial hemorrhage, an event that has been noted in patients with a prior history of a cerebrovascular accident, especially of hemorrhagic nature. Perhaps the last frontier for potential use of thrombolytic treatment is in patients who have fresh thrombotic occlusions of the carotid cerebral, basilar, or vertebral arteries. Long a subject of clinical research interest,“‘.“’ a renewed effort in patients with this potential indication concentrates on very early treatment and on ruling out a nascent hemorrhagic component with computerized tomographic scanning.‘73,‘74 If the early encouraging data hold,“’ then perhaps a subset of patients with thrombotic cerebrovascular accidents can be identified and saved from disabling or life-threatening disease.

94

SHERRY

AND

MARDER

A

B

C ‘,

.:.. :.

;.:

,::.:.. . . . .

Fig 1. Relative effects of plasminogen activators in a vessel occluded with thrombus and in which a site of prior trauma is sealed with a hemostatic plug. Two groups of plasminogen activator are compared. those with the attribute of greater “fibrinogen-sparing” (“fibrin selectivity“) (t-PA and scu-PA) and those with more potent effects on the blood coagulation and flbrinolytic proteins (SK, UK, APSAC). The latter group causes a greater decrease in plasma fibrinogen, as indicated schematically by the lower density of dots, but both types of plasminogen activator induce bleeding at the same rate and to the same degree by virtue of hemostatic plug dissolution. Assuming the occluded vessel is a coronary artery in a patient with acute myocardial infarction, reperfusion follows at approximately equal rates and speed for “fresh” thrombi of 2 hours duration, but more rapid lysis is achieved with the more fibrinogen-sparing (fibrin-selective) agents when thrombi are older than 3 hours. Thus, at phase A (approximately 90 minutes after initiation of treatment), t-PA and scu-PA will have achieved a higher rate of vascular reperfusion than SK, UK, and APSAC, whereas with t-PA the blood fibrinogen concentration will not have decreased as much. However, this does not occur with scu-PA, because it is partially converted to UK in the circulation, resulting in a potent lytic state. By 3 to 24 hours after treatment initiation (phases Band C), thrombus exposed to the nonfibrinogen-sparing agents also dissolves, producing a stable condition of a striking lytic state and persistent vascular patency. With t-PA, there is a higher tendency for vascular reocclusion (shown in phase C), perhaps related to the lesser degree of fibrinogen degradation and blood hypocoagulability. However, scu-PA therapy is associated with blood hypocoagulability and a low incidence of rethrombosis. The final patency status (70% to 80%) resulting from slower, progressive and stable reperfusion induced by SK, UK, and APSAC is equivalent to that which follows more rapid reperfusion counterbalanced by increased rethrombosis associated with &PA or scu-PA.

THROMBOSIS

AND

FIBRINOLYSIS

95

PERSPECTIVE

The greatest potential for expanding clinical benefit to many more patients does not require an innovative plasminogen activator, a new biochemical insight, or a large-scale clinical trial. At present, less than 15% of patients with acute myocardial infarction receive thrombolytic treatment with any agent,‘76 but an expanded program of thrombolytic treatment may be possible, for example, using early administration by ambulance personnel.“’ In addition, if

increased use in the at-risk population could be achieved within 2 or 3 hours after symptom onset, then significantly more patients could be spared short (1 month) or long-term (1 year) lethal outcomes from acute myocardial infarction. Such an accomplishment would represent the full fruition of research on the plasminogenplasmin enzyme system, initiated nearly 60 years ago with the identification of streptococcal fibrinolysin.4

REFERENCES 1. Macfarlane RG, Biggs R: Fibrinolysis. Its mechanism and significance. Blood 3:1167-1187, 1948 2. Dastre A: Fibrinolyse dans le sang. Arch Norm Pathol 5661-663, 1893 3. Yudin SS: Transfusion of cadaver blood. JAMA 106: 997-999,1936 4. Tillett WS, Garner RL: The fibrinolytic activity of hemolytic streptococci. J Exp Med 58485502,1933 5. Milstone H: A factor in normal human blood which participates in streptococcal fibrinolysis. J Immunol42:109116,194l 6. Christensen LR: Streptococcal fibrinolysis: A proteolytic reaction due to a serum enzyme activated by streptococcal fibrinolysin. J Gen Physiol28:363-383, 1945 7. Kaplan MH: Nature and role of the lytic factor in hemolytic streptococcal fibrinolysis. Proc Sot Exp Biol Med 57140-43.1944 8. Christensen LR: The activation of plasminogen by chloroform. J Gen Physiol28:559-583,1945 9. Christensen LR, MacLoed CM: A proteolytic enzyme of serum. characterization, activation, and reaction with inhibitors. J Gen Physiol28:559-83. 1945 10. Kwaan HC, McFadzean AJS: Plasma fibrinolytic activity induced by ischaemia. Clin Sci 15:245-257, 1956 11. Astrup T: Fibrinolysis in the organism. Blood 11:781806,1956 12. Todd AS: Fibrinolysis autographs. Nature 181:495496,1958 13. vonKaulla KN: Intravenous protein-free pyrogen. A powerful fibrinolytic agent in man. Circulation 17:187-198, 1958 14. Tillett WS, Sherry S: The effect in patients of streptococcal fibrinolysis (streptokinase) and streptococcal desoxyribonuclease on fibrinous, purulent and sanguinous pleural exudations. J Clin Invest 28:173-190,1949 15. Johnson AJ, McCarty WR: The lysis of artificially induced intravascular clots in many by intravenous infusions of streptokinase. J Clin Invest 38:1627-1643, 1959 16. Fletcher AP, Alkjaersig N, Sherry S: The maintenance of a sustained thrombolytic state in man. I. Induction and effects. J Clin Invest 38:1096-1110,1959 16a. Fletcher AP, Sherry S, Alkjaersig N, et al: The maintenance of a sustained thrombolytic state in man. II. Clinical observations on patients with myocardial infarction and other thromboembolic disorders. J Clin Invest 38:11111119,1959

17. Kline DL: Purification and crystallization of plasminogen (profibrinolysin). J Biol Chem 204:949-955,1953 18. Alkjaersig N, Fletcher AP, Sherry S: Activation of human plasminogen. 1. Spontaneous activation in glycerol. J Biol Chem 233:81-85,1958 19. Shulman S, Alkjaersig N, Sherry S: Physicochemical studies on human plasminogen (profibrinolysin) and plasmin (fibrinolysin). J Biol Chem 233:91-97,1958 20. Alkjaersig N, Fletcher AP, Sherry S: Activation of human plasminogen. 2. A kinetic study of activation with trypsin, urokinase and streptokinase. J Biol Chem 233:8690,1958 21. Troll W, Sherry S, Wachman J: Action of plasmin on synthetic substrates. J Biol Chem 208:85-93,1954 22. Alkjaersig N, Fletcher AP, Sherry S: The mechanism of clot dissolution by plasmin. J Clin Invest 38:1086-1095, 1959 23. Sherry S, Fletcher AP, Alkjaersig N: Fibrinolysis and fibrinolytic activity in man. Physiol Rev 39:343-382, 1959 24. Wiman B, Collen D: Molecular mechanisms of physiological fibrinolysis. Nature 272:549-550,1978 25. Summaria L, Hsieh B, Robbins KC: The specific mechanism of activation of human plasminogen to plasmin. J Biol Chem 242:4279-4283,1967 26. DeRenzo EC, Siiteria PK, Hutchings BL, et al: Preparation and properties of highly purified streptokinase. J Biol Chem 242:533-542, 1967 27. Deutsch DG, Mertz ET: Plasminogen: Purification from human plasma by affinity chromatography. Science 170:1095-1096,197O 28. Rickli EE, Cuendet PA: Isolation of plasmin-free human plasminogen with N-terminal glutamic acid. Biochim Biophys Acta 250:447-451, 1971 29. Castellino FJ, Soditz JM, Brockway WJ, et al: Streptokinase. Methods Enzymol45:244-257, 1976 30. Robbins KC: The human plasma fibrinolytic system: Regulation and control. Mol Cell Biochem 20:149-157, 1978 31. Sottrup-Jensen L, Claeys H, Zajdel M, et al: The primary structure of human plasminogen: Isolation of two lysine-binding fragments and one “mini”-plasminogen (MW 38,000) by elastase-catalyzed specific limited proteolysis, in Davidson JF, Rowan RM, Samama MM, et al (eds): Progress in Chemical Fibrinolysis and Thrombolysis, vol 3. New York, NY, Raven Press, 1978, p 191 32. Dayhoff MO: Atlas of Protein Sequence and Structure, vol 5. Silver Spring, MD, National Biomedical Research Foundation, 1978, p 91 (suppl3)

SHERRY

33. Castellino FJ, Violand BN: The fibrinolytic systembasic considerations. Prog Cardiovasc Dis 21:241-254, 1979 34. Castellino FJ: A unique enzyme-protein substrate modifier reaction. Plasministreptokinase interaction. Trends Biocbem Sci 41-51979 35. Kluft C, Trumpi-Kalshoven MM, Jie AFH, et al: Factor XII-dependent fibrinolysis. A double function of plasma kallikrein and the occurrence of a previously undescribed factor XII and kallikrein dependent plasminogen proactivator. Thromb Haemost 41:756-773,1979 36. Colman RW: Activation of plasminogen by human plasma kallikrein. Biochim Biophys Acta 35:273-279,1969 37. Mandle R Jr, Kaplan AP: Hageman factor substrates. Human plasma prekallikrein: Mechanism of activation by Hageman factor and participation in Hageman factordependent fibrinolysis. J Biol Chem 252:6097-6104,1977 38. Binder BR, Spragg J, Austen KF: Purification and characterization of human vascular plasminogen activator derived from blood vessel perfusates. J Biol Chem 254:19982003,1979 39. Cole ER, Bachmann F: Purification and properties of a plasminogen activator from pig heart. J Biol Chem 25213729-3737, 1977 40. Rijken DC, Wijngaards G, Zaal DeJong M, et al: Purification and partial characterization of plasminogen activator from human uterine tissue. Biochim Biophys Acta 580:140-153,1979 41. Mullertz S, Clemmensen I: The primary inhibitor of plasmin in human plasma. Biochem J 159:545-553, 1976 42. Wiman B, Cohen D: Purification and characterization of human antiplasmin, the fast-acting plasmin inhibitor in plasma. Eur J Biochem 78:19-26,1977 43. Wiman B, Collen D: On the mechanism of the reaction between human a,-antiplasmin and plasmin. J Biol Chem 254:9291-9297,1979 44. Moroi M, Aoki N: Isolation and characterization of a,-plasmin inhibitor from human plasma: A novel proteinase inhibitor which inhibits activator-induced clot lysis. J Biol Chem 251:5956-5965,1976 45. Fletcher AP, Alkjaersig N, Sherry S: Pathogenesis of the coagulation defect developing during pathological plasma proteolytic (“fibrinolytic”) states. I. The significance of fibrinogen proteolysis and circulating fibrinogen breakdown products. II. The significance, mechanism and consequences of defective fibrin polymerization. III. Demonstration of abnormal clot structure by electron microscopy. J Clin Invest 41:896-948,1962 46. Latallo ZS, Fletcher AP, Alkjaersig N, et al: Inhibition of fibrin polymerization by fibrinogen proteolysis products. Am J Physiol202:681-686,1962 47. Triantaphyllopoulus DC: Anticoagulant effect of incubated fibrinogen on blood coagulation. Can J Biochem Physiol36:249-259,1958 48. Nussenzweig V, Seligmann M, Grabar P: Les produits de degradation du fibrinogene human par la plasmine. II. Etude immunologique: Mise en evidence d’anticorps anti-fibrinogene natif posstdant de specifites differentes. Ann Inst Pasteur 100:490-508,1961 49. Marder VJ, Shulman NR, Carroll WR: High molecular weight derivatives of human fibrinogen produced by plasmin. I. Physicochemical and immunological characteriza-

AND

MARDER

tion and II. Mechanisms of their anticoagulant activity. J Biol Chem 244:2111-2124,1969 50. Hall CE, Slayter HS: The fibrinogen molecule: Its size, shape, and mode of polymerization. J Biophys Biothem Cytol5:11-16,1959 51. Gardlund-B, Hessel B, Marguerie G, et al: Primary structure of human fibrinogen. Characterization of disulfidecontaining cyanogenbromide fragments. Eur J Biochem 77:595-610,1977 52. Cohen D: On the regulation and control of fibrinolysis. Thromb Haemost 43:77-89,198O 53. Lesuk A, Terminiello L, Traver JH: Crystalline human urokinase: Some properties. Science 147:880-882,1965 54. White WF, Barlow GH, Mozen MM: The isolation and characterization of plasminogen activators (urokinase) from human urine. Biochemistry 5:2160-2169,1966 55. Fletcher AP, AIkjaersig N, Sherry S, et al: Development of urokinase as a thrombolytic agent. Maintenance of a sustained thrombolytic state in man by its intravenous infusion. J Lab Clin Med 65:713-731,1965 56. Sherry S, Fletcher AP, Alkjaersig N, et al: e-aminocaproic acid, “a potent anti-fibrinolytic agent.” Trans Assoc Am Physicians 72:62-70,1959 57. Okamoto S: British patent specification no. 770,693. London, England, Her Majesty’s Stationary Office, 1957 58. Brockway WJ, Castellino FJ: The mechanism of the inhibition of plasmin by e-aminocaproic acid. J Biol Chem 246:4641-4647,197l 59. Andersson L, Nilsson IM, Nilehn J-E, et al: Experimental and clinical studies on AMCA, the anti-fibrinolytic active isomer of p-aminomethylcyclohexame carboxylic acid. Stand J Haematol2:230-247,1965 60. Okamoto S, Oshiba S, Mihara H, et al: Synthetic inhibitors of fibrinolysis. In vitro and in vivo mode of action. Ann NY Acad Sci 146:414-429,1968 61. Wiman B: On the reaction of plasmin or plasmin streptokinase complex with aprotinin or a-anti-plasmin. Thromb Res 17:143-152,198O 62. Nilsson IM, Andersson L, Bjorkman SE: Epsilonamino caproic acid (E-ACA) as a therapeutic agent: Based on 5 years’ clinical experience. Acta Med Stand 448:1-46, 1966 (suppl) 63. Prentice CRM: Indications for antifibrinolysis therapy. Thromb Diath Haemorrh 34:634-643,1975 64. McNicol GP: Disordered fibrinolytic activity and its control. Scott Med J 7:266-276,1962 65. Bernik MA, Oller EP: Plasminogen activator and proactivator (urokinase precursor) in lung cultures. J Am Med Worn Assoc 31:465-472,1976 66. Nolan C, Hall LS, Barlow GH, et al: Plasminogen activator from human embryonic kidney cell culturesevidence for a proactivator. Biochim Biophys Acta 496:384400,1977 67. Hussain SS, Lipinski B, Gurewich V: US patent no. 431346 filed Nov 13,1979 68. Stampfer MJ, Goldhaber SZ, Yusuf S, et al: Effect of intravenous streptokinase on acute myocardial infarction: Pooled results from randomized trials. N Engl J Med 307:1180-1182,1982 69. Urokinase-Pulmonary Embolism Trial Study Group:

THROMBOSIS

AND

FIBRINOLYSIS

PERSPECTIVE

Urokinase-pulmonary embolism trial: A national cooperative study. Circulation 47:IIl-108, 1973 (suppl2) 70. Urokinase-streptokinase pulmonary embolism trial: Phase 2 results. A national cooperative study. JAMA 229:1606-1613, 1974 71. Marder VJ, Soulen RL, Atichartakarn V, et al: Quantitative venographic assessment of deep vein thrombosis in the evaluation of streptokinase and heparin therapy. J Lab Clin Med 89:1018-1029, 1977 72. Arnesen H, Heilo A, Jakobsen E, et al: A prospective study of streptokinase and heparin in the treatment of deep vein thrombosis. Acta Med Stand 203:457-463, 1978 73. Elliot MS, Immelman EJ, Jeffery P, et al: A comparative randomized trial of heparin versus streptokinase in the treatment of acute proximal venous thrombosis: An interim report of a prospective trial. Br J Surg 66:838-843, 1979 74. Sherry S, Bell WR, Duckert FH, et al: Thrombolytic therapy in thrombosis: A National Institutes of Health Consensus Development Conference. Ann Intern Med 93:141-144,198O 75. DeWood MA, Spores J, Notske R, et al: Prevalence of total coronary occlusion during the early hours of transmural myocardial infarction. N Engl J Med 303:897902,198O 76. Rentrop P, Blanke H, Kostering K, et al: Acute myocardial infarction: Intracoronaty application of nitroglycerin and streptokinase in combination with transluminal recanalization. Clin Cardiol2:354-363, 1979 77. Schroder R, Biamino G. Enz-Rudiger L, et al: Intravenous short-term infusion of streptokinase in acute myocardial infarction. Circulation 63:536-548, 1983 78. Markis JE, Malagold M, Parker JA, et al: Myocardial salvage after intracoronary thrombolysis with streptokinase in acute myocardial infarction: Assessment by intracoronary thallium-20. N Engl J Med 305:777-782,198l 79. Simoons ML, Serruys PW. van den Brand M, et al: Early thrombolysis in acute myocardial infarction: Limitation of infarct size and improved survival. J Am Coll Cardiol 71717-728, 1986 80. Sherry S: Overview of current thrombolytic agents, in Sleight P, Chamberlain DA (eds): Thrombolysis: The Dawn of a New Era? London, England, IBC Technical Services, 1990, p 1 81. Kennedy JW, Ritchie JL, Davis KB, et al: Western Washington randomized trial of intracoronary streptokinase in acute myocardial infarction. N Engl J Med 309:14771482,1983 82. Gruppo Italian0 per lo Studio della Streptokinasi nell’Infarto Miocardico (GISSI): Effectiveness of intravenous thrombolytic treatment in acute myocardial infarction. Lancet 1:397-402, 1986 83. ISIS-2 (Second International Study of Infarct Survival) Collaborative Group: Randomized trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. Lancet 2:349-360,1988 84. AIMS Trial Study Group: Effect of intravenous APSAC on mortality after acute myocardial infarction: Preliminary report of a placebo-controlled clinical trial. Lancet 1:545-549,1988 85. Wilcox RG. von der Lippe GV, Olsson CG, et al:

97

Trial of tissue plasminogen activator for mortality reduction in acute myocardial infarction. Anglo Scandinavian Study of Early Thrombolytic Therapy (ASSET). Lancet 2:525-530. 1988 86. Rijken DC, Collen D: Purification and characterization of the plasminogen activator secreted by human melanoma cells in culture. J Biol Chem 256:7035-7041, 1981 87. Pennica D, Holmes LWE, Kohr WJ, et al: Cloning and expression of human tissue-type plasminogen activator cDNA in E cd. Nature 301:214-221, 1983 88. Smith RAG, Dupe RJ, English PD, et al: Fibrinolysis with acyl enzymes: A new approach to thrombolytic therapy. Nature 290:505-508,198l 89. Hussain SS, Gurewich V, Lipinski B: Purification and partial characterization of a single-chain, high-molecularweight form of urokinase from human urine. Arch Biochem Biophys 220:31-38,1983 90. Bode C, Matsueda GR, Hui KY, et al: Antibody directed urokinase: A specific fibrinolytic agent. Science 229:765-767,1985 91. Chmielewska J, Ranby M, Wiman B: Evidence for a rapid inhibitor to tissue plasminogen activator in plasma. Thromb Res 31:427-436.1983 92. Verheijen JH, Chang GTG, Kluft C: Evidence for the occurrence of a fast-acting inhibitor for tissue-type plasminogen activator in human plasma. Thromb Haemost 51:392-395, 1984 93. Juhan-Vague I, Moerman B, DeCock F, et al: Plasma levels of a specific inhibitor of tissue-type plasminogen activator (and urokinase) in normal and pathological conditions. Thromb Res 33:523-530,1984 94. Aoki N, Harpel PC: Inhibitors of the fibrinolytic enzyme system. Semin Thromb Hemost 10:24-41, 1984 95. Spengers ED, Kluft C: Plasminogen activator inhibitors. Blood 69:381-387, 1987 96. Korninger C, Wagner 0, Binder BR: Tissue plasminogen activator inhibitor in human plasma. Development of a functional assaysystem and demonstration of a correlating M, = 50,000 antiactivator. J Lab Clin Med 105:718-724, 1985 97. Mahmoud M, Gaffney PJ: Bioimmunoassay (BIA) of tissue plasminogen activator (t-PA) and its specific inhibitor (t-PA/Inh). Thromb Haemost 53:356-359,1985 98. Sprengers ED: A sensitive assay,specific for endothelial cell type plasminogen activator inhibitor in blood plasma. Thromb Haemost 55:74-77,1986 99. Harpel PC: Alpha,-plasmin inhibitor and alpha,macroglobulin-plasmin complexes in plasma. Quantification by an enzyme-linked differential antibody immunosorbent assay.J Clin Invest 68:46-55, 1981 100. Wiman B: Human q-antiplasmin. Methods Enzymol80:395-408, 1981 101. Gurewich V, Pannell R, Louie S, et al: Effective and fibrin specific lysis by a zymogen precursor form of urokinase (pro-urokinase). A study in vitro and in vivo two animal species. J Clin Invest 73:1731-1739, 1984 102. Booth NA, Walker E, Maughan R, et al: Plasminogen activator in normal subjects after exercise and venous occlusion: t-PA circulations as complexes with C-l inhibitor and PAI-1. Blood 69:1600-1604,1987 103. Smith D, Gilbert M, Owen WG: Tissue plasminogen

98

activator release in vivo in response to vasoactive agents. Blood 66:835-839,1985 104. Bennett B, Croll A, Ferguson K, et al: Complexing of tissue plasminogen activator with PAI-1, u,-macroglobulin and Cl-inhibitor: Studies in patients with defibrination and a fibrinolytic state after electroshock or complicated labor. Blood 75:671-676,199O 105. Levin RI, Harpel PC, Harpel JG, et al: Inhibition of tissue plasminogen activator activity by aspirin in vivo and its relationship to levels of tissue plasminogen activator antigen, plasminogen activator inhibitor, and their complexes. Blood 74:1635-1643,1989 106. Suffredini AF, Harpel PC, Parrillo JE: Promotion and subsequent inhibition of plasminogen activation after administration of intravenous endotoxin to normal subjects. N Engl J Med 320:1165-1172,1989 107. Munkvad S, Jespersen J, Gram J, et al: Interrelationship between coagulant activity and tissue-type plasminogen activator (t-PA) system in acute ischaemic heart disease. Possible role of the endothelium. J Intern Med 228:361-366, 1990 108. Ranby M, Bergsdorf N, Pohl G, et al: Purification and characterization of two variants of native one-chain tissue plasminogen activator. FEBS Lett 146:289-292, 1982 109. Kasai S, Arimura H, Nishida M, et al: Primary structure of single-chain pro-urokinase. J Biol Chem 260: 12382-12389,1985 110. Gunzler W, Steffens G, Otting F, et al: The primary structure of high molecular mass human urokinase from human urine. The complete amino acid sequence of the A chain. Hoppe Seyler’s 2 Physiol Chem 363:1155-1165, 1982 111. Holmes WE, Nelles L, Lijnen HR, et al: Primary structure of human alpha-2-antiplasmin, a serine protease inhibitor (serpin). J Biol Chem 262:1659-1664,1987 112. Ny T, Sawdey M, Lawrence DA, et al: Cloning and sequence of a cDNA coding for the human P-migrating endothelial cell-type plasminogen activator inhibitor. Proc Nat1 Acad Sci USA 83:6776-6780,1986 113. Silverstein RL, Leung LLK, Harpel PC et al: Complex formation of platelet thrombospondin with plasminogen: Modulation of activation by tissue activator. J Clin Invest 74:1625-1633,1984 113a. Silverstein RL, Leung LLK, Harpel PC, et al: Platelet thrombospondin forms a trimolecular complex with plasminogen and histidine-rich glycoprotein. J Clin Invest 75:2065-2073,1985 114. Loscalzo J, Vaughan DE: Tissue plasminogen activator promotes platelet disaggregation. J Clin Invest 79:17491755,1987 115. Hajjar KA, Harpel PC, Jaffe EA, et al: Binding of plasminogen to cultured human endothelial cells. J Biol Chem 261:11656-11662,1986 116. Hajjar KA, Hamel NM, Harpel PC, et al: Binding of tissue plasminogen activator to cultured human endothelial cells. J Clin Invest 80:1712-1719,1987 117. Knudsen BS, Silverstein RL, Leung LLK, et al: Binding of plasminogen to extracellular matrix. J Biol Chem 261:10765-10771,1986 118. Miles LA, Plow EF: Binding and activation of plasminogen on the platelet surface. J Biol Chem 260:43034311,1985

SHERRY

AND

MARDER

119. Stricker RB, Wond D, Shiu DT, et al: Activation of plasminogen by tissue plasminogen activator on normal and thrombasthenic platelets: Effects on surface proteins and platelet aggregation. Blood 68:1280-1284,1986 120. Marder VJ, Sherry S: Thrombolytic therapy: Current status. N Engl J Med 318:1512-1520,1585-1595,1988 121. Caller B: Platelets and thrombolytic therapy. N Engl J Med 322:33-42,199O 122. Aoki N: Genetic abnormalities of the fibrinolytic system. Semin Thromb Hemost l&42-50,1984 123. Aoki N, Saito H, Kamiya T, et al: Congenital deficiency of q-plasmin inhibitor associated with severe hemorrhagic tendency. J Clin Invest 63:877-884,1979 124. Kazama M, Tahara C, Suzuki Z, et al: Abnormal plasminogen, a case of recurrent thrombosis. Thromb Res 21:517-522,198l 125. Schleef RR, Higgins DL, Pilleiner E, et al: Bleeding diathesis due to decreased functional activity of type 1 plasminogen activator inhibitor. J Clin Invest 83:1747-1752, 1989 126. Dieval J, Nguyen G, Gross S, et al: A lifelong bleeding disorder associated with a deficiency of plasminogen activator inhibitor type 1. Blood 77:528-532,199l 127. Nilsson IM, Tengborg L: A family with thrombosis associated with high level of tissue plasminogen activator inhibitor. Haemostasis 14:24,1984 (abstr) 128. Stead NW, Bauer KA, Kinney TR, et al: Venous thrombosis in a family with defective release of vascular plasminogen activator and elevated plasma factor VIII/van Willebrand’s factor. Am J Med 74:33-39,1983 129. Jorgensen M, Mortensen JZ, Madsen AG, et al: A family with reduced plasminogen activator activity in blood associated with recurrent venous thrombosis. Stand J Haematol29:217-223, 1982 130. Holmes WE, Pennica D, Blaber M, et al: Cloning and expression of the gene for pro-urokinase in Escherichia coli. Biotechnology 3:923-929,1985 131. Nolli ML, Sarubbi E, Corti A, et al: Production and characterization of human recombinant single chain urokinase-type plasminogen activator from mouse cells. Fibrinolysis 3:101-106, 1989 132. Heyneker HL, Holmes WE, Vehar GA: Preparation of functional human urokinase proteins. European patent application no. 83103629.8, publication no. 0092182A2. European patent office, Munich, Federal Republic of Germany, 1983 133. Collen D, Stassen JM, Marafino BJ Jr, et al: Biological properties of human tissue-type plasminogen activator obtained by expression of recombinant DNA in mammalian cells. J Pharmacol Exp Ther 231:146-152,1984 134. Cohen D: Human tissue-type plasminogen activators: From the laboratory to the bedside (editorial). Circulation 72:18-20,1985 135. Petitpretz P, Simmoneau G, Cerrina J, et al: Effect of a single bolus of urokinase in patients with lifethreatening pulmonary emboli: A descriptive trial. Circulation 70:861-866,1984 136. Ozbek C, Sen S, Frank S, et al: Rapid high dose streptokinase in severe pulmonary embolism. Lancet 2:229230,1989 137. Goldhaber SZ, Kessler CM, Heit J, et al: Acute

THROMBOSIS

AND

FIBRINOLYSIS

PERSPECTIVE

pulmonary embolism treated with tissue plasminogen activator. Lancet 1:886-889, 1986 138. Comerota AJ, Rubin RN, Tyson RR, et al: Intraarterial thrombolytic therapy in peripheral vascular disease. Surg Gynecol Obstet 1651~81987 139. McNamara TO, Fischer JR: Thrombolysis of peripheral arterial and graft occlusions. Improved results using high-dose urokinase. Am J Roentgen01 144:769-775,1985 140. McNamara TO, Bowberger RA: Factors affecting initial and 6-month patency rates after intraarterial thrombolysis with high dose urokinase. Am J Surg 152:709-712, 1986 141. Katzen BT: Technique and results of “low dose infusion.” Cardiovasc Intervent Radio1 11541-547, 1988 (SUPPI) 142. Groar RA, Risius B, Young R, et al: Peripheral artery and bypass graft thrombolysis with recombinant tissue-type plasminogen activator. J Vast Surg 3:115-124, 1986 143. Chesebro JH, Knatterud G, Roberts R, et al: Thrombolysis in Myocardial Infarction (TIMI) Trial, Phase I: A comparison between intravenous tissue plasminogen activator and intravenous streptokinase. Circulation 76:142-154, 1987 144. Marder VJ: The use of thrombolytic agents: Choice of patient, drug administration, laboratory monitoring. Ann Intern Med 90:802-808,1979 145. Rao AK, Pratt C, Berke A, et al: Thrombolysis in Myocardial Infarction (TIMI) Trial, Phase I: Hemorrhagic manifestations and changes in plasma fibrinogen and fibrinolytic system in patients treated with recombinant tissue plasminogen activator and streptokinase. J Am Coil Cardiol 11:1-l& 1988 146. Stump DC, Califf RM, Top01 EJ, et al: Pharmacodynamics of thrombolysis with recombinant tissue-type plasminogen activator. Correlation with characteristics of and clinical outcomes in patients with acute myocardial infarction. Circulation 80:1222-1230,1989 147. Sherry S, Marder VJ: Streptokinase and tissue plasminogen activator therapy are of equivalent value for survival after acute myocardial infarction: 90-minute patency rates did not predict these results. Ann Intern Med 114:417-423, 1991 148. Sherry S: Thrombolytic therapy for acute myocardial infarction: Lessons to be learned. Tex Heart Inst J 18:103-109,199l 149. Schroder R: Mechanisms by which thrombolysis reduces mortality: Limitation of infarct size-what else, in Sleight P, Chamberlain DA (eds): Thrombolysis: The Dawn of a New Era? London, England, IBC Technical Services Ltd. 1990, p 46 150. Braunwald E: Myocardial reperfusion, limitation of infarct size, reduction of left ventricular dysfunction, and improved survival. Should the paradigm be expanded? Circulation 13:1130-1132,1989 151. Califf RM: Myocardial reperfusion: Is it ever too late? J Am Coll Cardiol13:1130-1132,1989 152. Gruppo Italian0 Per Lo Studio Della Soprawivenza Nell’Infarto Miocardico: GISSI-2: A factorial randomized trial of alteplase versus streptokinase and heparin versus no

99

heparin among 12,490 patients with acute myocardial infarction. Lancet 336:65-71, 1990 153. The International Study Group: In-hospital mortality and clinical course of 20,891 patients with suspected acute myocardial infarction randomized between alteplase and streptokinase with or without heparin. Lancet 336:7175,199o 154. White HD, Rivers JT, Maslowski AH, et al: Effect of intravenous streptokinase as compared to that of tissue plasminogen activator on left ventricular function after first myocardial infarction. N Engl J Med 320:817-&X21, 1989 155. Dalen JE, Gore JM, Braunwald E, et al: Six- and twelve-month follow-up of the Phase I Thrombolysis in Myocardial Infarction (TIMI) Trial. Am J Cardiol 62:179185,1988 156. Johns JA, Gold HK, Leinbach RC, et al: Prevention of coronary artery reocclusion and reduction in late coronary artery stenosis after thrombolytic therapy in patients with acute myocardial infarction. A randomized study of maintenance infusion of recombinant human tissue-type plasminogen activator. Circulation 78:546-556,1988 157. Sherry S: An appraisal of various thrombolytic agents in the treatment of acute myocardial infarction. Am J Med 83:31-46, 1987 (suppl2A) 158. Marder VJ: Thrombolytic agents: Balancing cost, efficacy, and side effects. Clin Cardiol13:IV37-41. 1990 159. van Breda A, Katzen BT, Deutsch AS: Urokinase versus streptokinase in local thrombolysis. Radiology 165: 109-111.1987 160. Belkin M, Belkin B, Buckman CA, et al: Intraarterial fibrinolytic therapy-efficacy of streptokinase versus urokinase. Arch Surg 121:769-773, 1986 161. White HD: GISSI-2 and the heparin controversy. Lancet 336:297-298,199O 162. Vanhove P and the Belgian Saruplase Aheplase Trial Group: Effects of saruplase and of alteplase on hemostasis in patients with acute myocardial infarction. A single-blind randomized trial. J Am Coll Cardiol 17: 186A, 1991 (abstr) 163. Diefenbach C, Bar F, Meyer J, on behalf of the PRIM1 Study Group: Early patency and reocclusion rates in acute myocardial infarction. Circulation 82:111-537. 1990 (abstr) (suppl3) 164. Grines CL, Nissen SE, Booth DC, et al: A new thrombolytic regimen for acute myocardial infarction using combination half dose tissue type plasminogen activator with full dose streptokinase: A pilot study. J Am Co11 Cardiol 14:573-580, 1989 165. Neuhaus K-L. Tebbe U, Gottwick M, et al: Intravenous recombinant tissue plasminogen activator (rt-PA) and urokinase in acute myocardial infarction: Results of the German Activator Urokinase Study (GAUS). J Am Co11 Cardiol 12581-587, 1988 166. Runge MS, Bode C, Matsueda GR, et al: Antibody enhanced thrombolysis: Targeting of tissue plasminogen activator in vivo. Proc Nat1 Acad Sci USA 84:7659-7662. 1987 167. Larsen GR, Metzger M, Henson K. et al: Pharmacokinetic and distribution analysis of variant forms of tissuetype plasminogen activator with prolonged clearance in rat. Blood 73:1842-1850, 1989

100

168. Garde11 SJ, Duong LT, Diehl RE, et al: Isolation, characterization and cDNA cloning of a vampire bat salivary plasminogen activator. J Biol Chem 26417947-17952, 1989 169. Marder VJ, Bell WR: Fibrinolytic therapy, in Colman RW, Hirsh J, Marder VJ, et al (eds): Hemostasis and Thrombosis: Basic Principles and Clinical Practice (ed 2). Philadelphia, PA, Lippincott, 1987, pp 1395-1437 170. Goldhaber SZ: Thrombolysis in venous thromboembolism. An international perspective. Chest 97:176S-181S, 1990 171. Fletcher AP, Alkjaersig N, Lewis M, et al: A pilot study of urokinase therapy in cerebral infarction. Stroke 7:135-142,1976 172. Sloan MA: Thrombolysis and stroke. Past and future. Arch Neural 44:748-768,1987

SHERRY

AND

MARDER

173. Del Zoppo GJ, Zeumer H, Harker LA: Thrombolytic therapy in stroke: Possibilities and hazards. Stroke 17:595-607,1986 174. Brott T, Haley EC, Levy DE, et al: The investigational use of tPA for stroke. Ann Emerg Med 17:1202-1205, 1988 175. Hacke W, Zeumer H, Ferbert A, et al: Intra-arterial thrombolytic therapy improves outcome in patients with acute vertebrobasilar occlusive disease. Stroke 19:12161222,1988 176. Grines CL, DeMaria AN: Optimal utilization of thrombolytic therapy for acute myocardial infarction: Concepts and controversies. J Am Co11Cardiol16:223-231,199O 177. Castaigne AD, HervC C, Duval-Moulin A-M, et al: Prehospital use of APSAC: Results of a placebo-controlled study. Am J Cardiol64:30A-33A, 1989

Thrombosis, fibrinolysis, and thrombolytic therapy: a perspective.

Progress VOL XXXIV, in Cardiovascular Diseases NO 2 SEPTEMBER/OCTOBER Thrombosis, Fibrinoiysis, and Thrombolytic A Perspective 1991 Therapy:...
1MB Sizes 0 Downloads 0 Views