SYNERGISM, MUTANTS AND HYBRIDS OF TISSUE-TYPE PLASMINOGEN ACTIVATO(t-PA) AND SINGLE CHAIN UROKINASE-TYPE PLASMINOGEN ACTIVATOR(scu-PA):POTENTIAL FORTHROMBOLYTIC THERAPY

Author(s):  
D Collen

With the development and clinical investigation of the fibrin-specific thrombolytic agents t-PA and scu-PA, many questions relating to their optimal use remain to be answered. It is, however, becoming apparent that these agents, in addition to several advantages,suffer some shortcomings, e.g. their therapeutic dose is large and their fibrin-specificity is limited.Therefore,the quest for better thrombolytic agents remains open.We will report results of four main lines of research which we are pursuing to obtain better agents or regimens for fibrin-specific thrombolytic therapy.1. Synergism between t-PA and scu-PA. t—PA and scu—PA in molar ratios between 4:1 and 1:4 show no synergism for thrombolysis of a plasma clot immersed in plasma in vitro(Thromb. Haemost. 56, 35, 1986) but display significant in vivo synergismin a rabbit model (Circulation 74, 838, 19867and in man (Am. Heart J. 112, 1083,1986).Recently we have confirmed synergism for thrombolysis between t-PA and scu-PA in a coronary thrombosis model in the dog(Zuskind et al., unpubl.) and in the baboon (Collen et al., unpubl.). Sequential infusion of t-PA followed by scu-PA butnot of scu-PA followed by t-PA is syneristic(Collen et al., this meeting).2. Mutants of t-PA. In collaboration with Larssen et al.,deletion mutants of t-PA, obtained by in vitro mutagenesis are characterized with respect to pharmacokinetics and thrombolytic properties.Mutants lacking the finger—like domain and/or the growth factor domainand/or one or all of the glycosylation sites have a much slower clearance (Larssenet al., this meeting) but unaltered specific thrombolytic properties and fibrin-specificity (Collen et al., this meeting).3. Mutants of scu-PA. A truncated form of scu-PA, lacking the 143 NH2~terminal amino acids was shown to be pharmacologically and thrombolytically indistinguishable from intact scu—PA (Stump et al.).Mutants of scu—PA in which Lys 158 is replaced,whereby they can no longer be converted to urokinase, still haveintrinsic plasminogen activating properties and act synergistically with t-PA on thrombolysis in vivo (Nelles et al., this meeting).4. Hybrids of t-PA and scu-PA. In collaboration with Pierard et al. (this meeting) hybrids of NH2~terminal regions of t-PA and COOH-terminal regions of u-PA were constructed which, after translation in transient expression systems, showed apparent specific activities comparable to that of natural two-chain u-PA. One hybrid, composed of the finger domain of t-PA and the B-chain of u-PA, was scaled up, purified and characterized (Gheysen et al., this meeting). This hybrid had theenzymatic properties typical of single chain u-PA, but had not acquired the fibrinaffinity of t-PA.Based on the finding that the isolated A-chain of t-PA retains the intact fibrin-affinity of the native molecule (Holvoet et al.,Eur.J. Biochem. 158, 173, 1986) andthat a low Mr form of scu-PA retains the functional properties of the intact moleule (J. Biol. Chem. 261, 17120, 1986), we have constructed and expressed a hybrid consisting of the NH -terminal region of t-PA (amino acids 1 to 263) and the COOH-terminal region of scu-PA (amino acids 144to 411) (Lijnen et al., this meeting). This hybrid has both fibrin affinity of t-PA (although less pronounced) and the enzymatic properties of scu-PA. The activation of plasminogen by the hybrid is apparently stimulated by fibrin.We believe that continued research along these lines will yield thrombolytic agents or therapeutic schemes, which may be superior to t-PA and/or scu-PA in terms of specific thrombolytic activity and fibrin-specificity.

1987 ◽  
Author(s):  
J M Stassen ◽  
D Collen

t-PA and scu-PA, in molar ratios between 1:4 and 4:1 do not act synergically in vitro (Thromb. Haemost. 56,35,1986) but display marked synergism in a rabbit model (Circulation 74, 838, 1986) and in man (Am. Heart J. 112, 1083, 1986). To investigate the mechanism of in vivo synergism in the rabbit model (J. Clin. Invest. 71, 368, 1983), t-PA and scu-PA were infused 1) simultaneously over 4 hrs, 2) t-PA over 1 hr, then 15 min later scu-PA over 2 hrs and 3) scu-PA over 1 hr, then 15 min later t-PA over 2 hrs.Significant synergism on thrombolysis is observed when t-PA and scu-PA are infused simultaneously or when t-PA is followed by scu-PA but not when scu-PA is followed by t-PA. These results suggest that low dose t-PA induces some plasminogen activation, sufficient to partially degrade fibrin, exposing COOH-terminal lysines with high affinity for plasminogen (Eur. J. Biochem. 140, 513, 1984). scu-PA might then activate surface-bound Glu-pla-minogen more efficiently.Sequential therapy with t-PA (or any other agent which "predigests" the thrombus), followed by scu-PA might constitute an alternative to simultaneous infusion of synergistic thrombolytic agents.


1987 ◽  
Author(s):  
R S Rappaport ◽  
M R Blume ◽  
R L Vogel ◽  
M H Levner ◽  
P P Hung

There is mounting evidence from animal models and the clinic that combination thrombolytic therapy with tissue-type plasminogen activator (tPA) and single chain urokinase (scuPA) is synergistic. Yet, efforts to demonstrate synergism between these two plasminogen activators in vitro have met with discordant results. Collen et al (Thromb. Haemostasis, 56:35, 1986) reported an absence of synergism between these two agents on clot lysis in an in vitro plasma milieu when they were evaluated at molar ratios of 1:4 (tPA:scuPA and vice versa). Gurewich and Pannell (Thromb. Res., 44:217, 1986), however, reported a synergistic effect on fibrin-specific clot lysis in vitro when the agents were combined in concentrations exceeding molar ratios of 1:4 (tPA:scuPA). Here, we present evidence that synergism between tPA and scuPA may be demonstrated in vitro provided that the molar ratio of tPA to scuPA exceeds 1:4 and that the concentration of clot bound or unbound tPA is minimized. In order to achieve this experimental condition, the standard in vitro plasma clot lysis assay was modified. Human plasma clots were incubated first for a short time in plasma containing varying amounts of tPA. After incubation, the clots were washed thoroughly and reimmersed in plasma alone or in plasma containing varying amounts of scuPA or tPA. Under these conditions, lysis proceeded at a greater rate and to a greater extent when tPA clots were immersed in plasma containing an appropriate amount of scuPA than when they were immersed in plasma alone or in plasma containing appropriate amounts of tPA. Lysis of untreated clots or clots exposed first to scuPA and then to plasma containing varying amounts of scuPA proceeded far less efficiently with a characteristic lag. The enhanced lysis produced by tPA and scuPA obeyed the classical definition of synergy: the same biological effect can be obtained with two drugs together at algebraic fractional combinations of less than 1 (Berenbaum, M.C., Clin. Exp. Immunol., 28:1-18, 1977). Thus, conditions that more closely mimic the in vivo situation resulting from a bolus injection of tPA followed by infusion with scuPA, may provide a system for duplication of in vivo synergism in. vi tro and investigation of the mechanism thereof.


2000 ◽  
Vol 167 (1) ◽  
pp. 117-124 ◽  
Author(s):  
C Galet ◽  
M Chopineau ◽  
N Martinat ◽  
Y Combarnous ◽  
F Guillou

The C-terminal region of the beta subunit of the human chorionic gonadotrophin (hCG) is implied in heterodimer stability (beta26-110 disulphide bridge), in vitro LH bioactivity (region beta102-110) and in in vivo LH bioactivity (beta CTP). Like the hCG beta, the equine eLH and eCG beta subunits, also possess a C-terminal extension (CTP). But, in contrast to hCG, eLH and eCG bind to both LH and FSH receptors in species other than the horse. This allows investigation of the roles of the beta subunit C-terminal region of a eLH/CG recombinant molecule on both LH and FSH activities. To do so, the CTP was deleted and/or the beta26-110 disulphide bond was mutated and the resulting mutated beta subunits were transiently co-expressed with common alpha subunit in COS7 cells. These regions were also deleted in a betaalphaeLH/CG single chain also expressed in COS7 cells. The hormones produced were characterized by different ELISAs and in vitro LH and FSH bioassays. Mutation of the 26-110 disulphide bond and deletion of the betaCTP led to a decrease in eLH/CG heterodimer production. Double mutation promoted an additive effect on production of the heterodimer and of the corresponding tethered eLH/CG. The elimination of the beta26-110 disulphide bond in the betaalpha single chain had no effect on its production. However, neither the 26-110 disulphide bond nor the CTP mutations affected dimer stability and bioactivities of the secreted heterodimers and/or single chain molecules. Therefore, in contrast to hCG, the 26-110 S-S bond of the recombinant eLH/CG beta subunit does not seem to be essential for eLH/CG dimer stability upon secretion and expressing LH and FSH bioactivities.


1979 ◽  
Author(s):  
Laurence A. Sherman ◽  
Joan Lee

Fibronectin (cold insoluble globulin, CIG) binds to fibrin in vitro by both H+ bonds and F.XIII crosslinkinq. Potential in vivo significance was studied in a rabbit model. 131I-CIG purified by usual methods was ≥ 40% denatured as evinced by rapid blood clearance and complex t½. In contrast, the remaining nondenatured 131I-CIG in animals given 3H-amino acids had the same blood t½ (70-75 hours) and C1(0.8) as did the 3H-CIG. The CIG t½ was greater than that found for fibrinogen (55-65 hours). A second series received 131I-fibrinogen and nondenatured 125I-CIG i.V. Subsequently, thromboplastin or thrombin (50 u/Kg/lhr) was injected and both 125I-CIG and 13I-fibrinogen levels dropped during injection to 50-70% of preinjection levels. When ancrod was injected instead, 131I-fibrinogen fell by >90%, but 125I-CIG levels did not change. When animals were initially dofibrinated with ancrod CIG levels remained unchanged during subsequent thrombin injection. The results indicate that CIG has a longer normal blood t½ than fibrinogen, but the body distributions were approximately the same. When fibrin is formed, CIG and fibrin complex in vivo, and are rapidly cleared from the blood. The abnormal fibrin formed by ancrod apparently does not bind to CIG in vivo, or else does so reversibly prior to clearance from the blood.


1989 ◽  
Vol 62 (03) ◽  
pp. 909-916 ◽  
Author(s):  
Edward T A Fry ◽  
David L Mack ◽  
Burton E Sobel

SummaryEnhancement of thrombolysis with combinations of tissue-type and single chain urokinase plasminogen activators (t-PA and scu-PA) has been demonstrated in vivo but has not been seen consistently in vitro. This study was designed to characterize interactions between t-PA and scu-PA with respect to rate of and extent of thrombolysis in vitro and to delineate mechanisms responsible. Combinations of t-PA and scu-PA at selected concentrations synergistically enhanced thrombolysis in vitro compared with thrombolysis induced by either activator alone. Enhanced thrombolysis did not occur at the expense of fibrin specificity since the extent of fibrinogenolysis and consumption of α2-antiplasmin were significantly less with synergistic combinations of t-PA and scu-PA compared with equi-effective concentrations of either activator alone. Attenuation of complex formation of t-PA and two chain u-PA (tcu-PA), formed from scu-PA, with plasma proteins did not appear to contribute to enhancement of thrombolysis as assessed by fibrin autography. Binding of 125I-t-PA to thrombi was increased by 27% at 1 hr and by 21% at 2 hr in the presence of scu-PA (p <0,001 for both). Conversion of scu-PA to tcu-PA was enhanced when thrombi were exposed to scu-PA in the presence of t-PA. Results of this study indicate that t-PA and scu-PA at selected concentrations enhance thrombolysis in vitro synergistically without compromising fibrin specificity. Enhanced binding of t-PA to thrombi in the presence of scu-PA and enhanced conversion of scu-PA to tcu-PA appear to contribute to synergy between t-PA and scu-PA for thrombolysis.


1979 ◽  
Author(s):  
Laurence Sherman ◽  
Joan Lee

Fibronectin (cold insoluble globulin, CIG) binds to fibrin in vitro by both H+ bonds and F.XIII crosslinking. Potential in vivo significance was studied in a rabbit model. 131 I-ciG purified by usual methods was ≥ 40% denatured as evinced by rapid blood clearance and complex t½. in contrast, the remaining nondenatured 131 I-CIG in animals given 3 H-amino acids had the same blood t½ (70-75 hours) and Cl(0.8) as did the 3 H-CIG. The CIG t½ was greater than that found for fibrinogen (55-65 hours). A second series received 131-fibrinogen and nondenatured 125I-CIG i.v. Subsequently, thromboplastin or thrombin (50 u/Kg/lhr) was injected and both 125 I-CIG and 131I-fibrinogen levels dropped during injection to 50-70% of preinjection levels. When ancrod was injected instead, 131I-fibrinogen fell by >90%, but 125I-CIG levels did not change. When animals were initially defibrinated with ancrod CIG levels remained unchanged during subsequent thrombin injection. The results indicate that CIG has a longer normal blood t½ than fibrinogen, but the body distributions were approximately the same. When fibrin is formed, CIG and fibrin complex in vivo, and are rapidly cleared from the blood. The abnormal fibrin formed by ancrod apparently does not bind to CIG in vivo, or else does so reversibly prior to clearance from the blood.


2005 ◽  
Vol 93 (04) ◽  
pp. 627-630 ◽  
Author(s):  
Roger Lijnen ◽  
Désiré Collen

SummaryThrombolytic agents are plasminogen activators that convert the zymogen plasminogen to the active enzyme plasmin, which degrades fibrin. Elucidation of the molecular mechanism of physiological fibrinolysis opened up a new era of fibrin-specific thrombolysis. Fibrin-specific plasminogen activators, including tissue-type plasminogen activator (t-PA), single-chain urokinase-type plasminogen activator (scu-PA) and staphylokinase (Sak), preferentially activate fibrin-associated plasminogen. Generated plasmin remains associated with fibrin, where it is protected from rapid inhibition and can efficiently degrade fibrin, avoiding systemic activation of the fibrinolytic system. Following a decade of clinical investigation t-PA and variants thereof are routinely used for treatment of patients with thromboembolic disease.


1999 ◽  
Vol 81 (04) ◽  
pp. 605-612 ◽  
Author(s):  
Dmitry V. Sakharov ◽  
Marrie Barrett-Bergshoeff ◽  
Rob T. Hekkenberg ◽  
Dingeman C. Rijken

SummaryIn a number of cases, thrombolytic therapy fails to re-open occluded blood vessels, possibly due to the occurrence of thrombi resistant to lysis. We investigated in vitro how the lysis of hardly lysable model thrombi depends on the choice of the plasminogen activator (PA) and is accelerated by ultrasonic irradiation. Lysis of compacted crosslinked human plasma clots was measured after addition of nine different PAs to the surrounding plasma and the effect of 3 MHz ultrasound on the speed of lysis was assessed.Fibrin-specific PAs showed bell-shaped dose-response curves of varying width and height. PAs with improved fibrin-specificity (staphylokinase, the TNK variant of tissue-type PA [tPA], and the PA from the saliva of the Desmodus rotundus bat) induced rapid lysis in concentration ranges (80-, 260-, and 3,500-fold ranges, respectively) much wider than that for tPA (a 35-fold range). However, in terms of speed of lysis, these three PAs exceeded tPA only slightly. Reteplase and single-chain urokinase were comparable to tPA, whereas two-chain urokinase, anistreplase, and streptokinase were inferior to tPA. In the case of fibrin-specific PAs, ultrasonic treatment accelerated lysis about 1.5-fold. For streptokinase no acceleration was observed. The effect of ultrasound correlated with the presence of plasminogen in the outer plasma, suggesting that it was mediated by facilitating the transport of plasminogen to the surface of the clot.In conclusion, PAs with improved fibrin-specificity induce rapid lysis of plasminogen-poor compacted plasma clots in much wider concentration ranges than tPA. This offers a possibility of using single-or double-bolus administration regimens for such PAs. However, it is not likely that administration of these PAs will directly cause a dramatic increase in the rate of re-opening of the occluded arteries since they are only moderately superior to tPA in terms of maximal speed of lysis. Application of high-frequency ultrasound as an adjunct to thrombolytic therapy may increase the treatment efficiency, particularly in conjunction with fibrin-specific PAs.


1987 ◽  
Vol 58 (03) ◽  
pp. 921-926 ◽  
Author(s):  
E Seifried ◽  
P Tanswell

SummaryIn vitro, concentration-dependent effects of rt-PA on a range of coagulation and fibrinolytic assays in thawed plasma samples were investigated. In absence of a fibrinolytic inhibitor, 2 μg rt-PA/ml blood (3.4 μg/ml plasma) caused prolongation of clotting time assays and decreases of plasminogen (to 44% of the control value), fibrinogen (to 27%), α2-antiplasmin (to 5%), FV (to 67%), FVIII (to 41%) and FXIII (to 16%).Of three inhibitors tested, a specific polyclonal anti-rt-PA antibody prevented interferences in all fibrinolytic and most clotting assays. D-Phe-Pro-Arg-CH2Cl (PPACK) enabled correct assays of fibrinogen and fibrinolytic parameters but interfered with coagulometric assays dependent on endogenous thrombin generation. Aprotinin was suitable only for a restricted range of both assay types.Most in vitro effects were observed only with rt-PA plasma concentrations in excess of therapeutic values. Nevertheless it is concluded that for clinical application, collection of blood samples on either specific antibody or PPACK is essential for a correct assessment of in vivo effects of rt-PA on the haemostatic system in patients undergoing fibrinolytic therapy.


1994 ◽  
Vol 72 (06) ◽  
pp. 906-911 ◽  
Author(s):  
D C Rijken ◽  
E Groeneveld ◽  
M M Barrett-Bergshoeff

SummaryBM 06.022 is a non-glycosylated mutant of human tissue-type plasminogen activator (t-PA) comprising only the kringle-2 and proteinase domains. The in vivo half-life of BM 06.022 antigen is 4- to 5-fold longer than that of t-PA antigen. The in vitro half-life of the activity of BM 06.022 at therapeutic concentrations in plasma is shorter than that of t-PA. In this study the inactivation of BM 06.022 in plasma was further investigated.Varying concentrations of BM 06.022 were incubated in plasma for 0-150 min. Activity assays on serial samples showed a dose-dependent decline of BM 06.022 activity with a half-life from 72 min at 0.3 μg/ml to 38 min at 10 μg/ml. SDS-polyacrylamide gel electrophoresis (SDS-PAGE) followed by fibrin autography showed the generation of several BM 06.022-complexes. These complexes could be completely precipitated with antibodies against Cl-inactivator, α2-antiplasmin and α1-antitrypsin.During the incubation of BM 06.022 in plasma, plasmin was generated dose-dependently as revealed by varying degrees of a2-anti-plasmin consumption and fibrinogen degradation. SDS-PAGE and immunoblotting showed that single-chain BM 06.022 was rapidly (i. e. within 45 min) converted into its two-chain form at concentrations of 5 μg/ml BM 06.022 and higher.In conclusion, BM 06.022 at therapeutic concentrations in plasma was inactivated by Cl-inactivator, a2-antiplasmin and a j-antitrypsin. The half-life of the activity decreased at increasing BM 06.022 concentrations, probably as a result of the generation of two-chain BM 06.022 which may be inactivated faster than the single-chain form.


Sign in / Sign up

Export Citation Format

Share Document