WO1995025540A1 - Method for reducing immune and hemostatic dysfunctions during extracorporeal circulation - Google Patents

Method for reducing immune and hemostatic dysfunctions during extracorporeal circulation Download PDF

Info

Publication number
WO1995025540A1
WO1995025540A1 PCT/US1995/003614 US9503614W WO9525540A1 WO 1995025540 A1 WO1995025540 A1 WO 1995025540A1 US 9503614 W US9503614 W US 9503614W WO 9525540 A1 WO9525540 A1 WO 9525540A1
Authority
WO
WIPO (PCT)
Prior art keywords
activation
complement
platelet
blood
antibody
Prior art date
Application number
PCT/US1995/003614
Other languages
French (fr)
Inventor
Scott A. Rollins
Brian R. Smith
Stephen P. Squinto
Original Assignee
Alexion Pharmaceuticals, Inc.
Yale University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alexion Pharmaceuticals, Inc., Yale University filed Critical Alexion Pharmaceuticals, Inc.
Priority to EP95914820A priority Critical patent/EP0751787B1/en
Priority to DE69533921T priority patent/DE69533921T2/en
Priority to AU21917/95A priority patent/AU2191795A/en
Publication of WO1995025540A1 publication Critical patent/WO1995025540A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3687Chemical treatment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans

Definitions

  • the present invention relates to reducing the dysfunction of the immune and hemostatic systems associated with extracorporeal circulation.
  • the invention relates to the use of antibodies specific to human complement component C5 to effect inhibition of the complement arm of the immune system in order to accomplish such therapeutic prophylaxis.
  • Extracorporeal circulation (ECC) of the blood is an important medical technology that is used in a variety of life saving medical procedures. Such procedures include hemodialysi ⁇ , plasmapheresis, plateletpheresis, leukophereses, extracojrporeal membrane oxygenation (ECMO) , heparin-induced extracorporeal LDL precipitation (HELP) , and cardiopulmonary bypass (CPB) . As such, ECC is widely used in modern medical practice.
  • CPB Cerebrasian pulmonary disease
  • Hemostatic problems during and after ECC can be attributed to several factors, including complement-mediated platelet dysfunction, and can result in both excessive thrombosis and excessive bleeding as platelets first become activated and then become spent and non ⁇ functional, and are removed from the circulation.
  • Management of abnormal bleeding associated with CPB often requires re-operation and is frequently associated with excessive, and sometimes inappropriate, blood product administration, occasionally exceeding the available blood supply.
  • open heart surgery accounts for more than 25% of the total blood product use (Woodman and Harker, Blood 76:1690. 1990) .
  • complement component C3 a soluble recombinant form of human complement receptor type 1 (sCRl, designated BRL55730 by SmithKline Beecham) to reduce complement activation associated with CPB (Gillinov, et al. , Ann Thorac Surg 55:619, 1993) .
  • sCRl acts by blocking the conversion of complement component C3 into activated components C3a and C3b.
  • key measures of complement and platelet activation were not evaluated in this study, and the neutrophil and other physiological endpoint results reported were disappointing.
  • sCRl blocks C3, and as di ⁇ cu ⁇ ed in detail below, the blockade of C3 activation interrupts the most important antimicrobial actions of the complement system, exposing patients to increased risk of infection.
  • Aprotinin abroad-based serine pr ⁇ teinase inhibitor (TRASYLOL, Bayer AG) has recently been studied for its effects on CPB as ⁇ ociated pathology.
  • Aprotinin inhibit ⁇ kallikrein, a proteolytic enzyme that attenuates the release of neutrophil elastase, another protease, and diminishes the production of complement component C3a.
  • aprotinin may also partially inhibit C3 convertase activity (Wachtfogel, et al. , Blood 69:324, 1989).
  • C3a plays a key role in generating ECC-as ⁇ ociated immune and hemostatic dysfunctions
  • CPB-induced activation of platelets which is (at lea ⁇ t in part) ⁇ econdary to complement activation, is unaffected by aprotinin therapy (Bidstrup, et al. , J Thorac Cardiovasc Surg 973:364, 1989)
  • platelet dysfunction is directly involved in the pathogenesi ⁇ of ECC- associated hemostatic problems.
  • aprotininin administration of aprotinin
  • Efficacy of aprotinin in reducing blood lo ⁇ during CPB ha ⁇ been demon ⁇ trated when aprotinin is administered by continuous infusion.
  • Single dose bolus administration of aprotinin has proven ineffective.
  • Enthusiasm for aprotinin use during CPB has been dampened by recent clinical results showing an increased incidence of perioperative myocardial infarction (16.9% vs. 8.9% for placebo) and a significant incidence of postoperative renal dysfunction associated with aprotinin administration during CPB (Cosgrove, et al. , Ann Thorac Surg 54:1031, 1992).
  • postmortem findings in some patients who died after elective coronary operations using aprotinin included acute vein graft occlusions as well as wide ⁇ pread thrombosis in the kidney, native coronary vessel ⁇ and cerebral ve ⁇ sels.
  • the benefits of pharmaceutical modulation of adverse effects associated with CPB have recently led to FDA approval of aprotinin for the treatment of patients receiving CPB.
  • EACA The synthetic lysine analogue EACA (AMICAR, Lederle Laboratories) has been used frequently as an antifibrinolytic agent during CPB.
  • EACA is effective in reducing bleeding in a variety of clinical circumstances, its use in CPB has been controversial with regard to its potential to reduce postoperative blood loss (Copeland, et al. , Ann Thorac Surg 47:88, 1989). Additionally, both arterial and venous thrombosis have complicated EACA therapy in a number of clinical trials and have generally discouraged its clinical use (Sonntag and Stein, J Neurosurg 40:480) .
  • the method of the invention involves the use of antibodies to human complement component C5 as a pharmaceutical agent. More particularly, the invention provides for the use of an anti-C5 antibody preparation that binds to native plasma C5, and thereby blocks the generation of activated complement components C5a and C5b from C5.
  • the antibody is a monoclonal antibody.
  • the invention is used during CPB.
  • the inhibition of dysfunction is accomplished by the administration of a single dose of the anti-C5 antibody preparation.
  • anti-C5 antibodies can completely block important aspects of human complement activity, while maintaining important anti-infective functions of the complement system.
  • the antibodies completely block the generation of the cytolytic assemblage of activated complement components known as the membrane attack complex.
  • the antibodies inhibit platelet activation, inhibit leukocyte activation, inhibit platelet-leukocyte interactions, specifically, platelet- monocyte and platelet-polymorphonuclear cell interactions, and inhibit the removal of key glycoproteins from platelet surfaces during extracorporeal circulation of human blood.
  • the antibodies help maintain the immune and hemostatic sy ⁇ tems in their normal state ⁇ during ECC.
  • the invention comprises a method for performing a therapeutic procedure on a patient comprising: (a) passing circulating blood from a blood vessel of the patient, through a conduit, and back to a blood vessel of the patient, the conduit having a luminal surface comprising a material capable of causing at least one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion in the patient's blood; and
  • step (b) introducing an antibody that specifically binds to complement component C5 and has the following properties in vitro: (1) the antibody substantially inhibits complement hemolytic activity; (2) the antibody substantially prevents the generation of activated complement component C5a, as shown by a substantial reduction in the expression of the leukocyte activation marker CDllb/CD18; and (3) the antibody does not sub ⁇ tantially inhibit C3 convertase activity; into the patient's bloodstream in an amount effective to reduce at least one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion resulting from pas ⁇ age of the circulating blood through said conduit, wherein step (a) occurs in at least one temporal relation to step (b) selected from the group consisting of before step (b) , during step (b) , and after ⁇ tep (b) .
  • FIG. 2 As ⁇ ay of level ⁇ of complement component C3a demon ⁇ trating that the generation of complement component C3a in whole human blood circulated through an extracorporeal circuit is not inhibited by the addition of an anti-C5 monoclonal antibody to ⁇ uch whole blood.
  • Assays were performed before the addition of the antibody or the commencement of the CPB circuit ("Pre Tx") using undiluted blood (“undil”) and diluted blood (“dil”) as described in Example 1. Samples of diluted blood to which the antibody had been added (“Post Tx”) were assayed at the times indicated after starting the CPB circuit.
  • Figure 3 Assay of the level ⁇ of soluble C5b-9 in human blood circulated through an extracorporeal circuit demon ⁇ trating that the addition of an anti-C5 monoclonal antibody to such whole blood inhibits the formation of the C5b-9 terminal complement assembly.
  • Post Tx Samples of diluted blood to which the antibody had been added
  • FIG. 4 FACS analysis of level ⁇ of GPIb ("GPlb") on all platelets, GPIIb-IIIa on all platelets (regardless of P-selectin expression, "GPIIb-IIIa”), P-selectin on all platelets ("%PLT activ”), GPIIb-IIIa on P-selectin positive platelets (“Ilb-IIIa- act”) , GPIIb-IIIa on P-selectin negative platelets (“Ilb-IIIa- inact”), platelet neutrophil binding ("PLT/PMN”), platelet monocyte binding (“PLT/MONO”) , neutrophil CDllb levels (“PMN CDllb”) , and monocyte CDllb levels (“MONO CDllb”) , demonstrating inhibition of all except the last of these parameters by the addition of an anti-C5 monoclonal antibody to whole human blood during ECC.
  • GPlb GPIIb-IIIa on all platelets
  • %PLT activ P-selectin on
  • the present invention relates to inhibition of dysfunctions of the immune and hemostatic systems during ECC.
  • ECC electronic circuitry
  • the relevant aspects of the immune and hemostatic systems we turn first to general discussions in the context of ECC of the relevant aspects of the immune and hemostatic systems, in particular, certain aspects of the complement arm of the immune system, the cellular arm of the immune system, and the physiology of platelet ⁇ .
  • Activation of complement components that mediate inflammation and impact the hemostatic properties of the blood occurs when blood comes in direct contact with the various non-biological components of the ECC circuit (Videm, et al., J. Thorac. Cardiovasc. Surg., 97:764-770, 1989) , and can be inhibited to some extent by certain drugs, particularly heparin and protamine, that are administered to patients during ECC procedures (Jones, et al., Anaesthesia. 37:629-633, 1982) .
  • ECC In addition to its effects on platelets and the complement arm of the immune system, ECC also effects the cellular arm of the immune system both through effects on leukocytes and through effects on platelet-leukocyte interactions.
  • Platelet ⁇ are anuclear, cellular elements of the blood that are vitally important for the formation of blood clots and the prevention
  • Platelet dysfunction has been linked with the contact of platelets with the non-biological surfaces of the extracorporeal oxygenator and the hypothermia associated with CPB.
  • CPB extracorporeal oxygenator
  • Activated complement components are arguably the most important factors contributing to the platelet dysfunction associated with ECC.
  • CPB adversely affects platelet count as well as function. Hemodilution during CPB causes platelet counts to rapidly decrease soon after ⁇ tarting CPB, declining to about 50% of preoperative levels. This level of circulating platelet ⁇ , if occurring in the context of normal individual platelet function, is unlikely to contribute to clinical bleeding. Of greater significance to the development of CPB associated morbidity, however, i ⁇ the progre ⁇ sive los ⁇ of platelet function seen during and after CPB. Within minute ⁇ after initiating CPB, bleeding time i ⁇ prolonged significantly and platelet aggregation is impaired. These changes in bleeding time are independent of platelet count and worsen as CPB progresses. Bleeding times, normally less than 10 minute ⁇ , can approach 30 minutes after 2 hours of CPB.
  • Platelet ⁇ undergo profound biochemical and morphological alterations when activated by certain stimuli. When caused by stimuli associated with conditions calling for rapid hemostasis, these alterations are associated with the normal functions of platelet ⁇ . When cau ⁇ ed by ECC, pathophy ⁇ iologic outcomes result.
  • Activation-induced alterations in platelet characteri ⁇ tic ⁇ include exocytotic degranulation with the release of the contents of various storage organelles, shape change ⁇ , and the induction of adhe ⁇ ivene ⁇ , aggregation, and thromboxane production.
  • P- ⁇ electin (al ⁇ o known as CD62, CD62P, GMP-140, and PADGEM) , which i ⁇ located on the inner surfaces of platelet alpha granules, i ⁇ translocated to the platelet surface as these organelles turn inside-out during exocytosis.
  • the appearance of P-selectin on the platelet surface thus serves as a marker of platelet activation.
  • Various lines of evidence ⁇ ugge ⁇ t that the expre ⁇ ion of P- ⁇ electin and the temporary depletion or modification of ⁇ ome functional platelet membrane component ⁇ are key measures of platelet functionality.
  • the removal of platelet membrane glycoproteins may be mediated by the activity of proteolytic enzymes such as pla ⁇ min and elastase.
  • proteolytic enzymes such as pla ⁇ min and elastase.
  • the ability of the protease inhibitor aprotinin (discussed above) to minimize glycoprotein (specifically, GPIb) loss after CPB provides evidence implicating direct proteoly ⁇ is of platelet membrane glycoproteins in the development of platelet dy ⁇ function (Wenger, et al., J Thorac Cardiova ⁇ c Surg. 97:235, 1989) .
  • the P-selectin molecule which appears on the membrane surface of activated platelet ⁇ , i ⁇ known to mediate the binding of platelets to various types of white blood cells (WBCs, leukocytes) without requiring the activation of the WBCs for such binding to occur.
  • WBCs white blood cells
  • These WBCs include polymorphonuclear leukocytes (PMNs, neutrophil ⁇ , granulocytes) , and monocytes, and the P-selectin mediated binding results in platelet-PMN, and platelet-monocyte conjugate formation (Lar ⁇ en, et al. , J Biol Chem 267:11104-11110, 1992; Corral, et al.
  • leukocyte-platelet adhesion i ⁇ also believed to be of physiologic importance in the targeting of leukocytes to appropriate inflammatory and/or hemostatic sites and in modulating leukocyte function.
  • the relevance of ⁇ uch targeting has been recently demonstrated in vivo in a baboon model where blockade of P-selectin with a monoclonal antibody resulted in decreased monocyte accumulation on an artificial vascular graft and decrea ⁇ ed procoagulant activity (Palabrica, et al. , Nature 359:848-851, 1992) .
  • Such leukocyte-platelet adhe ⁇ ion caused by P-selectin has been found to be induced by CPB (Rinder, et al., Blood. 79:1201-1205, 1992).
  • TF tissue factor
  • Circ Res 72:1075-1081, 1993 have recently demonstrated CPB-as ⁇ ociated upregulation of both the quantity and activity of TF on circulating monocyte ⁇ .
  • the expre ⁇ sion of monocyte tissue factor several hours after the conclusion of a CPB procedure, combined with other procoagulatory events occurring during CPB (Evangelista, et al. , Blood 77:2379-2388, 1991; Higuchi, et al.. Blood 79:1712-1719, 1992; Weitz, et al.
  • Such upregulation is particularly relevant to CPB-induced injury since CDllb/CD18 is responsible for leukocyte adherence to and penetration (diapede ⁇ is) through the endothelium via binding to the intercellular adhe ⁇ ion molecules ICAM-l and ICAM-3 on "activated" endothelium (Staunton, et al. , Cell 52:925-933, 1988; Staunton, et al., Nature 339:61-64, 1989; Furie, et al. , J Immunol 148:2395-2404, 1992) .
  • CDllb/CD18 may contribute to ECC associated medical problems.
  • the CDllb moiety function ⁇ a ⁇ the receptor for complement component iC3b and for elements of the contact dependant soluble coagulation system, including fibrinogen, fibronectin, and Factor X (Altieri, et al., Proc Natl Acad Sci USA 85:7462-7465, 1988; Nathan, et al. , J Cell Biol 109:1341-1349. 1989).
  • CDllb with fibrinogen may be of particular importance to ECC-associated pathology, since, a ⁇ di ⁇ cu ⁇ ed above, the platelet ⁇ urface glycoprotein complex GPIIb-IIIa is also associated with fibrinogen binding, thus providing a potential means for platelet-leucocyte adhesion, i.e., via a CDllb--fibrinogen-- GPIIb-IIIa linkage (Altieri, et al. , Proc Natl Acad Sci USA 85:7462- 7465, 1988) . Induction of an inflammation-associated respiratory burst in granulocytes in response to a variety of cytokines also appears to require CDllb/CD18 in the local adherent microenvironment.
  • the complement ⁇ ystem acts in conjunction with other im unological systems of the body to defend against intrusion of cellular and viral pathogens.
  • complement proteins There are at least 25 complement proteins, which are found as a complex collection of plasma proteins and membrane cofactors.
  • the plasma protein ⁇ make up about 10% of the globulins in vertebrate serum.
  • Complement components achieve their immune defensive functions by interacting in a series of intricate but precise enzymatic cleavage and membrane binding events.
  • the resulting complement cascade lead ⁇ to the production of product ⁇ with op ⁇ onic, immunoregulatory, and lytic function ⁇ .
  • the complement cascade converge ⁇ es via the cla ⁇ ical pathway or the alternative pathway. These pathways share many components, and while they differ in their initial steps, they converge and share the same "terminal complement” components responsible for the activation, attack, and destruction of target cells.
  • the classical complement pathway is typically initiated by antibody recognition of and binding to an antigenic site on a target cell.
  • the alternative pathway is usually antibody independent, and can be initiated by certain molecule ⁇ on pathogen ⁇ urface ⁇ . Both pathway ⁇ converge at the point where complement component C3 i ⁇ cleaved by an active protease (which is different in each pathway) to yield C3a and C3b.
  • Other pathways activating complement attack can act later in the sequence of events leading to variou ⁇ a ⁇ pect ⁇ of complement function.
  • C3a is an anaphylotoxin (see below) .
  • C3b binds to bacterial and other cells and tags them for removal from the circulation. (C3b in this role is known as opsonin.)
  • Patient ⁇ with genetic le ⁇ ion ⁇ that block C3b function are prone to infection by a broad variety of pathogenic organisms, while patients with lesions later in the complement cascade sequence, i.e., patient ⁇ with le ⁇ ion ⁇ that block C5 functions, are found to be more prone only to Neisseria infection, and then only somewhat more prone (Fearon, in Intensive Review of Internal Medicine. 2nd Ed. Fanta and Minaker, ed ⁇ . Brigham and Women's and Beth Israel Hospitals, 1983) .
  • C3b also forms a complex with other component ⁇ unique to each pathway to form classical or alternative C5 convertase, which cleaves C5 into C5a and C5b.
  • C3 is thus regarded as the central protein in the complement reaction ⁇ equence ⁇ ince it i ⁇ e ⁇ ential to both the alternative and clas ⁇ ical pathways (Wurzner, et al. , Complement Inflamm 8:328-340, 1991) .
  • This property of C3b i ⁇ regulated by the serum protease Factor I, which acts on C3b to produce iC3b. While still functional a ⁇ opsonin, iC3b cannot form an active C5 convertase.
  • C5a is another anaphylatoxin (see below) .
  • C5b combines with C6, C7, and C8 to form the C5b-8 complex at the surface of the target cell.
  • the membrane attack complex (MAC, C5b-9, terminal complement complex -- TCC) is formed.
  • MAC membrane attack complex
  • C5b-9 terminal complement complex -- TCC
  • MAC pore ⁇ When sufficient numbers of MACs insert into target cell membranes the openings (MAC pore ⁇ ) they create mediate rapid o ⁇ motic ly ⁇ i ⁇ of the target cells. Lower, non-lytic concentrations of MACs can produce other effects.
  • membrane insertion of small numbers of the C5b-9 complexes into endothelial cells and platelets can cause deleterious cell activation. In some case ⁇ activation may precede cell lysis.
  • platelet activation by MAC pores can predispose a CPB patient to late thrombotic events, such as vascular graft re-occlusion.
  • C3a and C5a are anaphylatoxins. These activated complement components can trigger mast cell degranulation, which releases histamine and other mediators of inflammation, resulting in smooth muscle contraction, increased vascular permeability, leukocyte activation, and other inflammatoryphenomena.
  • C5a also function ⁇ as a chemotactic peptide that seirves to attract pro-inflammatory granulocytes to the site of complement activation.
  • C3a anaphylatoxin have been found to increase dramatically during CPB, and there is a strong statistical association between elevated C3a levels and postoperative organ system dysfunction manifest by impairment and/or failure of cardiac, renal and pulmonary sy ⁇ tem ⁇ , bleeding diathe ⁇ i ⁇ , and the need for artificial ventilation
  • platelets can be activated by the assembly of terminal complement components C5b-9 on their surfaces.
  • the assembly of these complement components on platelets is known to occur during ECC (Finn, et al. , J Thorac Cardiovasc Surg.
  • Complement-mediated platelet activation leads ⁇ to alpha-granule relea ⁇ e, increa ⁇ ed expression of P-selectin, and the lo ⁇ s of GPIb.
  • the generation of products of complement activation such as C3a, C5a, and C5b-9 further results in platelet membrane vesiculation and consequent microparticle formation.
  • Other damaging effects of complement activation during CPB can include the activation of granulocytes, leading to partial degranulation and up-regulation of CDllb/CD18, and to damage to organs.
  • Such damaging effects are largely due to the actions of certain products of complement activation, specifically the anaphylatoxins C3a and C5a, which, in turn, can be converted to desArg forms with altered activity levels by plasma carboxypeptidase.
  • These activated complement components cause activation and aggregation of neutrophils.
  • Such activated cells accumulate in the pulmonary vessels and vascular beds, as ha ⁇ been demonstrated by serial biopsies of lung tissue before and after CPB (Howard, et al. , Arch Surg. 123:1496-1501, 1988). Liver, brain and pancreas, also suffer such damage, which can result in postoperative dysfunction of these organs.
  • the present invention relates to the use of anti-C5 antibodies in ECC. More generally, the invention relates to the use of anti-C5 antibodies in any procedure which involves circulating the patient's blood from a blood vessel of the patient, through a conduit, and back to a blood ves ⁇ el of the patient, the conduit having a luminal surface comprising a material capable of causing at least one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion.
  • Such procedures include, but are not limited to, all forms of ECC, as well a ⁇ procedure ⁇ involving the introduction of an artifical or foreign organ, tissue, or vessel into the blood circuit of a patient.
  • Conduit materials which have the foregoing effects on blood include those now known in the art as well as those which may be developed in the future. Such materials include synthetic materials such as the various forms of plastics and synthetic polymers used in CPB circuits, hemodialy ⁇ is circuits, vascular grafts, and artificial organs, as well a ⁇ biological materials of xenogeneic origin.
  • the anti-C5 antibody is used to reduce at least one of complement activation, platelet activation, leukocyte activation, or platele -leukocyte adhesion resulting from the circulation of the patient's blood through such a conduit.
  • Reduction of complement activation can be measured by method ⁇ well known in the art, for example, by the chicken erythrocyte hemolysi ⁇ method described below in Example 1.
  • a sufficient amount of antibody is used to reduce the increase in plasma hemolytic activity resulting from complement activation caused by contact with the conduit by at least 50%.
  • Reduction of platelet activation can be mea ⁇ ured by various methods well known in the art, for example, by immunofluore ⁇ cence analysis of the expression of platelet activation markers GPIIb-IIIa or P- ⁇ electin by the methods described below in Examples 1 and 4. Again, it is preferred to use a sufficient amount of antibody so a ⁇ to achieve at lea ⁇ t a 50% reduction in the increase in the expre ⁇ sion of at least one of the platelet activation markers resulting from contact with the conduit.
  • Reduction of leukocyte activation can be mea ⁇ ured by various methods well known in the art, for example, by immunofluorescence analysis of the expre ⁇ ion of the leukocyte activation marker CDllb/CD18.
  • the expression of this marker can be determined by the measurement of CDllb expression. Again, it i ⁇ preferred to u ⁇ e a ⁇ ufficient amount of antibody ⁇ o a ⁇ to achieve at lea ⁇ t a 50% reduction in the increa ⁇ e in the expression of the leukocyte activation marker by at least one type of leukocyte, e.g., PMNs, re ⁇ ulting from contact with the conduit.
  • a type of leukocyte e.g., PMNs
  • Reduction of the level ⁇ of platelet-leukocyte adhesion, specifically platelet-monocyte and platelet-neutrophil adhesion, can be measured by the fluorescence analysis techniques described below in Example 4, e.g., as the percent increase in leukocytes that read as GPIb or GPIIb/IIIa positive. Again, it is preferred to use a sufficient amount of antibody so as to achieve at least a 50% reduction in the increase in the percentage of GPIb or GPIIb/IIIa positive monocytes or neutrophils resulting from contact with the conduit.
  • the anti-C5 antibodies can be administered in a variety of unit dosage forms.
  • the dose will vary according to, e.g., the particular antibody, the manner of administration, the particular procedure being performed and its duration, the nature of the conduit or conduits used in the procedure, the overall health, condition, size, and age of the patient, and the judgment of the prescribing physician.
  • Dosage levels for human subjects are generally between about 1 mg per kg and about 100 mg per kg per patient per treatment, and preferably between about 5 mg per kg and about 50 mg per kg per patient per treatment. In some cases, only a single dose of the anti-C5 antibody can be administered to the patient in order to achieve a therapeutic effect.
  • the anti-C5 antibody is preferably administered prior to contact of the blood with the conduit.
  • administration can be continued for as long as the blood is in contact with the conduit, and even after that time, if desired.
  • a single dose can be used.
  • periodic do ⁇ es or a continuous infusion of the anti-C5 antibody can be used.
  • a typical procedure thus includes the following steps.
  • the patient i ⁇ prepared for surgery in accordance with standard practice and within an hour of the commencement of surgery, but generally before the commencement of the surgery, a first dose of the pharmaceutical preparation containing the anti-C5 antibody is given.
  • a series of doses may also be administered during and after the procedure.
  • levels of serum complement activity available in the patient's blood are monitored using the techniques set forth below in Example 1 to determine if additional doses are required, with such doses being administered as required to maintain at least a 50% reduction, and preferably about a 95% reduction of serum complement activity.
  • Administration of the anti-C5 antibodies will generally be performed by an intravascular route, e.g., via intravenous infusion by injection. Other routes of administration may be used if desired.
  • Formulations suitable for injection are found in Remington's Pharmaceutical Sciences. Mack Publishing Company, Philadelphia, PA, 17th ed. (1985) .
  • Such formulations must be sterile and non-pyrogenic, and generally will include a pharmaceutically effective carrier, such as saline, buffered (e.g., phosphate buffered) saline, Hank's solution. Ringer's solution, dextrose/saline, glucose ⁇ olution ⁇ , and the like.
  • the formulation ⁇ may contain pharmaceutically acceptable auxiliary ⁇ ub ⁇ tances as required, such as, tonicity adjusting agents, wetting agents, bactericidal agents, preservatives, stabilizers, and the like.
  • the formulations (pharmaceutical agents) of the invention can be distributed a ⁇ articles of manufacture comprising packaging material and the anti-C5 antibody.
  • the packaging material will include a label which indicates that the formulation is for use in as ⁇ ociation with an extracorporeal circulation procedure, e.g., a cardiopulmonary bypas ⁇ procedure.
  • the anti-C5 antibody i ⁇ preferably a monoclonal antibody, although polyclonal antibodies produced and ⁇ creened by conventional techniques can also be used if desired.
  • the anti-C5 antibodies are immunoreactive against epitopes on the beta chain of purified human complement component C5 (i.e., the part of the C5 molecule that become ⁇ C5b) and are capable of blocking the conversion of C5 into C5a and C5b by C5 convertase. This capability can be measured u ⁇ ing the technique ⁇ described in Wurzner, et al. , Complement Inflamm 8:328-340, 1991.
  • the amount of reduction of C5 converta ⁇ e activity i ⁇ preferably at least about 50% of the increase in convertase activity resulting from contact with the conduit.
  • Hybridomas producing monoclonal antibodies reactive with complement component C5 can be obtained according to the teachings of Sims, et al. , U.S. Patent No. 5,135,916. A ⁇ discussed therein, antibodie ⁇ are prepared u ⁇ ing purified component ⁇ of the complement membrane attack complex a ⁇ immunogens.
  • complement component C5 or C5b is preferably used as the immunogen.
  • the antibodies preferably should prevent the cleavage of C5 to form C5a and C5b, thus preventing the generation of the anaphylatoxic activity as ⁇ ociated with C5a and preventing the assembly of the membrane attack complex associated with C5b.
  • these anti-C5 antibodies will not impair the opsonization function as ⁇ ociated with the activation of complement component C3 by a C3 convertase.
  • Plasma C3 convertase activity can be measured by assaying plasma for the presence of C3a as described below in Example 2.
  • the anti-C5 antibody produces essentially no reduction in plasma C3a levels.
  • the term "monoclonal antibodies” refers to immunoglobulins produced by a hybridoma and antigen binding fragments (e.g., Fab' preparations) of such immunoglobulins, as well as to recombinantly expressed antigen binding proteins, including immunoglobulins, chimeric immunoglobulins, "humanized” immunoglobulins, antigen binding fragments of such immunoglobulins, ⁇ ingle chain antibodie ⁇ , and other recombinant proteins containing antigen binding domain ⁇ derived from immunoglobulin ⁇ .
  • Fab' preparations e.g., Fab' preparations
  • the blood was then used to prime the extracorporeal circuit of a COBE CML EXCEL membrane oxygenator CPB machine (Cobe BCT, Inc., Lakewood, CO)and the circuit wa ⁇ ⁇ tarted.
  • the circuit was cooled to 28°C and circulated for 60 min.
  • the circuit was then warmed to 37°C and circulated for an additional 30 min.
  • the experiment wa ⁇ then terminated. Sample ⁇ were taken at several time point ⁇ and evaluated for complement activity (Figure 1) .
  • Anti- chicken erythrocyte antibody (IgG fraction of anti-chicken-RBC antiserum, Intercell Technologies, Hopewell, NJ) was added to the cells at a final concentration of 25 ⁇ g/ml and the cells were incubated for 15 min. at 23°C. The cells were washed 2x with GVBS and 5xl0 6 cells were resuspended in a final volume of 130 ⁇ l GVBS containing 5% plasma. After incubation for 30 min. at 37°C, % hemolysis was calculated relative to a human serum control sample (Figure 1) . Hemolysis was determined by spinning the cells down and measuring released hemoglobin in the supernatant as the optical density at 4l5nm.
  • C5b-9 in each sample was determined using the manufacturers specification ⁇ and i ⁇ expressed in arbitrary ab ⁇ orbance units (AU) .
  • the anti-C5 antibody completely inhibited C5b-9 generation during extracorporeal circulation so that
  • Example l were washed, labeled with fluorescent antibodies directed against various cell surface marker molecules, and analyzed by flow cytometry (FACS) .
  • the primary antibodies used in these experiments were anti-GPIIb-IIIa (clone RUUSP2.41, AMAC, Inc., Westbrook, ME); anti-CDllb (Anti-CR3 (leu-15) clone D12, Becton-Dickinson
  • anti-GPIb CD42b
  • anti-CD45 a marker for both monocytes and PMNs, which are distinguished by the forward and side ⁇ catter characteri ⁇ tics of the FACS profiles of the CD45 positive cells
  • the anti-C5 antibody is a potent inhibitor of complement-induced platelet activation, leukocyte activation, specifically, PMN activation, and platelet-leukocyte adhesion during extracorporeal circulation and that terminal complement components, and not C3a, are largely re ⁇ ponsible for complement-mediated inflammation and platelet dysfunction.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Vascular Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Cardiology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Anesthesiology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The use of anti-C5 antibodies to reduce the dysfunction of the immune and hemostatic systems associated with extracorporeal circulation procedures, such as, cardiopulmonary bypass procedures, is disclosed. The antibodies have been found to significantly reduce complement activation, platelet activation, leukocyte activation, and platelet-leukocyte adhesion associated with such procedures.

Description

METHOD FOR REDUCING IMMUNE AND HEMOSTATIC DYSFUNCTIONS
DURING EXTRACORPOREAL CIRCULATION
The U.S. Government has a paid-up license in this invention and the right in limited circumstances to require the patent owner to license others on reasonable terms as provided for by the terms of Grant No. HL47193 awarded by The National Institutes of Health, Bethesda, Maryland. FIELD OF THE INVENTION
The present invention relates to reducing the dysfunction of the immune and hemostatic systems associated with extracorporeal circulation. In particular, the invention relates to the use of antibodies specific to human complement component C5 to effect inhibition of the complement arm of the immune system in order to accomplish such therapeutic prophylaxis. BACKGROUND OF THE INVENTION
I. Extracorporeal Circulation
Extracorporeal circulation (ECC) of the blood is an important medical technology that is used in a variety of life saving medical procedures. Such procedures include hemodialysiε, plasmapheresis, plateletpheresis, leukophereses, extracojrporeal membrane oxygenation (ECMO) , heparin-induced extracorporeal LDL precipitation (HELP) , and cardiopulmonary bypass (CPB) . As such, ECC is widely used in modern medical practice.
One of ECC's most common uses is in CPB. Nearly 400,000 CPB surgical procedures are carried out in the United States each year (Rinn, A., N Enαl J Med 312:119, 1985). The primary medical application of CPB is the facilitation of coronary artery bypass grafting, but it is also utilized during other types of open heart surgery, including procedures to correct congenital heart defects, heart valve disease, or other heart defects. With improvements in surgical techniques and extracorporeal oxygenation, the overall mortality for this procedure is low (1% to 4%) (Allen CM., Br Med J 297:1485, 1988) .
Because the most extensive research to date on the effects of ECC on the immune and hemostatic systems has been in the area of CPB, much of the discussion of the detailed effects of ECC presented herein is in connection with CPB. This discussion, however, is not intended to and should not be interpreted as limiting the present invention in any way to CPB. II . Medical Problems Caused by Extracorporeal Circulation Although death during ECC procedures is rare, several acute and chronic complications during and subsequent to these procedures result in potentially life-threatening medical problems and cause significant expense to the health care system. Many of these complications have been associated with activation of the immune system, with the complement arm of the immune system playing a particularly important role in the development of inflammation, platelet dysfunction, thrombocytopeni , and other ECC complications. Hemostatic problems during and after ECC can be attributed to several factors, including complement-mediated platelet dysfunction, and can result in both excessive thrombosis and excessive bleeding as platelets first become activated and then become spent and non¬ functional, and are removed from the circulation. Management of abnormal bleeding associated with CPB often requires re-operation and is frequently associated with excessive, and sometimes inappropriate, blood product administration, occasionally exceeding the available blood supply. In some hospitals, open heart surgery accounts for more than 25% of the total blood product use (Woodman and Harker, Blood 76:1690. 1990) .
Activation of the complement system occurs when blood plasma contacts foreign surfaces during ECC. Activated complement components can initiate inflammatory responses, with associated vasoconstriction, capillary leakage and platelet activation. A discussion of various of the physiological mechanisms involved in ECC-associated immune and hemostatic dysfunctions is presented below tinder the heading "Background Physiology." III. Previous Approaches to Complement Modulation During ECC
The many previous approaches taken in pursuit of controlling activation of the complement system and associated problems arising during ECC illustrate the perceived importance in the medical research community of accomplishing these goals. These approaches have included attempts to alter the mechanical components of CPB circuits, including the use of membrane (as opposed to bubble) oxygenatorε, and of heparin-coated bypass circuits, as well as various approaches to pharmacological modulation of complement activation. Unfortunately, these efforts have not eliminated the immune and hemostatic system problems associated with ECC, and have, in many cases, themselves been responsible for additional adverse effects. As mentioned above, foreign surfaces of the ECC circuit cause complement activation. In CPB, the oxygenator constitutes the most significant complement activating surf ce. Although some studies have shown that membrane oxygenators are associated with less activation of certain complement components than bubble oxygenatorε, there is no consensus that they cause less overall complement activation during CPB. Rather, complement activation varies with individual oxygenators, whether bubble or membrane (Videm, et al., Ann Thorac Surer 50:387, 1990) .
Recently, heparin-coated bypass circuits have been shown to reduce complement activation during CPB. Unfortunately, this approach only partially inhibits complement activation (Nilsson, et al., Artif Organs 14:46, 1990). No previous pharmacological approach to specifically reduce complement activation during CPB has yielded satisfactory results. Anti-inflammatory steroid drugs are im unosuppressive agents that can modulate complement function. Although steroid use has been associated with a moderate decrease in complement activation in some studies (Cavarocchi, et al. , J Thorac Cardiovasc Surg 91:252, 1986), others have found no significant beneficial effect (Miyamoto, et al. , ASAIO J 31:508, 1985), and the overall immune suppression caused by steroids, and other debilitating side effects associated with steroid administration, further limit their utility in treating ECC patients.
To date, most efforts to modulate the activation of complement in ECC have focused on complement component C3. A recent study evaluated the potential of a soluble recombinant form of human complement receptor type 1 (sCRl, designated BRL55730 by SmithKline Beecham) to reduce complement activation associated with CPB (Gillinov, et al. , Ann Thorac Surg 55:619, 1993) . sCRl acts by blocking the conversion of complement component C3 into activated components C3a and C3b. Unfortunately, key measures of complement and platelet activation were not evaluated in this study, and the neutrophil and other physiological endpoint results reported were disappointing. In addition, sCRl blocks C3, and as diεcuεεed in detail below, the blockade of C3 activation interrupts the most important antimicrobial actions of the complement system, exposing patients to increased risk of infection.
While ECC clearly causes complement activation, it is also associated with other problems, including kinin generation, loss of coagulation factors by hemodilution, fibrinolysis, liberation of thromboxane A-., and the activation of platelets and neutrophilε. Many of these phenomena are, at leaεt in part, secondary consequences of complement activation. Current therapies used to address these specific phenomena have, to date, relied on rather ineffective, broad-based antifibrinolytics and hemoεtatic agents, examples of which are as follows.
Aprotinin, abroad-based serine prσteinase inhibitor (TRASYLOL, Bayer AG) has recently been studied for its effects on CPB asεociated pathology. Aprotinin inhibitε kallikrein, a proteolytic enzyme that attenuates the release of neutrophil elastase, another protease, and diminishes the production of complement component C3a. In the absence of any direct evidence, it has been suggested that aprotinin may also partially inhibit C3 convertase activity (Wachtfogel, et al. , Blood 69:324, 1989). Although, aε mentioned above, there is a consensus in the art that C3a plays a key role in generating ECC-asεociated immune and hemostatic dysfunctions, CPB-induced activation of platelets, which is (at leaεt in part) εecondary to complement activation, is unaffected by aprotinin therapy (Bidstrup, et al. , J Thorac Cardiovasc Surg 973:364, 1989), and it is clear that platelet dysfunction is directly involved in the pathogenesiε of ECC- associated hemostatic problems. The beneficial effects of aprotinin administration are probably related to its ability to inhibit kallikrein, thereby diminishing the proteolytic conversion of plasminogen to plasmin, a key step in blood coagulation. Efficacy of aprotinin in reducing blood loεε during CPB haε been demonεtrated when aprotinin is administered by continuous infusion. Single dose bolus administration of aprotinin, however, has proven ineffective.
Enthusiasm for aprotinin use during CPB has been dampened by recent clinical results showing an increased incidence of perioperative myocardial infarction (16.9% vs. 8.9% for placebo) and a significant incidence of postoperative renal dysfunction associated with aprotinin administration during CPB (Cosgrove, et al. , Ann Thorac Surg 54:1031, 1992). In this study, postmortem findings in some patients who died after elective coronary operations using aprotinin included acute vein graft occlusions as well as wideεpread thrombosis in the kidney, native coronary vesselε and cerebral veεsels. Despite the risks associated with these deleterious clinical events, the benefits of pharmaceutical modulation of adverse effects associated with CPB have recently led to FDA approval of aprotinin for the treatment of patients receiving CPB.
The synthetic lysine analogue EACA (AMICAR, Lederle Laboratories) has been used frequently as an antifibrinolytic agent during CPB. Although EACA is effective in reducing bleeding in a variety of clinical circumstances, its use in CPB has been controversial with regard to its potential to reduce postoperative blood loss (Copeland, et al. , Ann Thorac Surg 47:88, 1989). Additionally, both arterial and venous thrombosis have complicated EACA therapy in a number of clinical trials and have generally discouraged its clinical use (Sonntag and Stein, J Neurosurg 40:480) . Tranexeamic acid has also been used for its anti-fibrinolytic effect, but has also been associated with excessive thrombotic complications (Orum, et al. , J Thorac Cardiovasc Surg, 105:78, 1993). The relatively non-specific hemostatic properties of the synthetic vasopreεsin analogue, deεmopressin acetate, has made it a candidate pharmaceutical agent for treating the hemodynamic alterations associated with CPB. However, randomized double-blind studies of 150 consecutive patients undergoing elective CPB found no significant differences in blood loss or postoperative transfusion requirements in patients receiving desmopreεεin (Hackmann, et al. , N Engl J Med 321:1437, 1989; Rocha, et al. , Circulation 77:1319, 1988) . SUMMARY OF THE INVENTION
In view of the foregoing state of the art, it is an object of the present invention to provide a method for reducing the dysfunction of the immune and hemostatic systems associated with extracorporeal circulation. The method of the invention involves the use of antibodies to human complement component C5 as a pharmaceutical agent. More particularly, the invention provides for the use of an anti-C5 antibody preparation that binds to native plasma C5, and thereby blocks the generation of activated complement components C5a and C5b from C5. Preferably, the antibody is a monoclonal antibody.
In certain preferred embodiments, the invention is used during CPB. In a further preferred embodiment, the inhibition of dysfunction is accomplished by the administration of a single dose of the anti-C5 antibody preparation. As shown in the examples presented below, anti-C5 antibodies can completely block important aspects of human complement activity, while maintaining important anti-infective functions of the complement system. In particular, the antibodies completely block the generation of the cytolytic assemblage of activated complement components known as the membrane attack complex. In addition, the antibodies inhibit platelet activation, inhibit leukocyte activation, inhibit platelet-leukocyte interactions, specifically, platelet- monocyte and platelet-polymorphonuclear cell interactions, and inhibit the removal of key glycoproteins from platelet surfaces during extracorporeal circulation of human blood. As a result of these activities, the antibodies help maintain the immune and hemostatic syεtems in their normal stateε during ECC.
In certain embodiments, the invention comprises a method for performing a therapeutic procedure on a patient comprising: (a) passing circulating blood from a blood vessel of the patient, through a conduit, and back to a blood vessel of the patient, the conduit having a luminal surface comprising a material capable of causing at least one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion in the patient's blood; and
(b) introducing an antibody that specifically binds to complement component C5 and has the following properties in vitro: (1) the antibody substantially inhibits complement hemolytic activity; (2) the antibody substantially prevents the generation of activated complement component C5a, as shown by a substantial reduction in the expression of the leukocyte activation marker CDllb/CD18; and (3) the antibody does not subεtantially inhibit C3 convertase activity; into the patient's bloodstream in an amount effective to reduce at least one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion resulting from pasεage of the circulating blood through said conduit, wherein step (a) occurs in at least one temporal relation to step (b) selected from the group consisting of before step (b) , during step (b) , and after εtep (b) .
The accompanying figureε, which are incorporated in and constitute part of the specification, illustrate certain aspects of the invention, and together with the description, serve to explain the principles of the invention. It is to be underεtood, of courεe, that both the figureε and the description are explanatory only and are not restrictive of the invention. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 -- Hemolytic assay demonstrating anti-C5 monoclonal antibody inhibition of complement activity associated with human blood circulated through an extracorporeal circuit. Assays were performed before the addition of the antibody or the commencement of the CPB circuit ("Pre Tx") using undiluted blood ("undil") and diluted blood ("dil") as described in Example 1. Samples of diluted blood to which the antibody had been added ("Post Tx") were assayed at the times indicated after starting the CPB circuit.
Figure 2 -- Asεay of levelε of complement component C3a demonεtrating that the generation of complement component C3a in whole human blood circulated through an extracorporeal circuit is not inhibited by the addition of an anti-C5 monoclonal antibody to εuch whole blood. Assays were performed before the addition of the antibody or the commencement of the CPB circuit ("Pre Tx") using undiluted blood ("undil") and diluted blood ("dil") as described in Example 1. Samples of diluted blood to which the antibody had been added ("Post Tx") were assayed at the times indicated after starting the CPB circuit.
Figure 3 -- Assay of the levelε of soluble C5b-9 in human blood circulated through an extracorporeal circuit demonεtrating that the addition of an anti-C5 monoclonal antibody to such whole blood inhibits the formation of the C5b-9 terminal complement assembly.
Assays were performed before the addition of the antibody or the commencement of the CPB circuit ("Pre Tx") using undiluted blood
("undil") and diluted blood ("dil") as described in Example 1.
Samples of diluted blood to which the antibody had been added ("Post Tx") were assayed at the times indicated after starting the CPB circuit.
Figure 4 -- FACS analysis of levelε of GPIb ("GPlb") on all platelets, GPIIb-IIIa on all platelets (regardless of P-selectin expression, "GPIIb-IIIa"), P-selectin on all platelets ("%PLT activ"), GPIIb-IIIa on P-selectin positive platelets ("Ilb-IIIa- act") , GPIIb-IIIa on P-selectin negative platelets ("Ilb-IIIa- inact"), platelet neutrophil binding ("PLT/PMN"), platelet monocyte binding ("PLT/MONO") , neutrophil CDllb levels ("PMN CDllb") , and monocyte CDllb levels ("MONO CDllb") , demonstrating inhibition of all except the last of these parameters by the addition of an anti-C5 monoclonal antibody to whole human blood during ECC. BACKGROUND PHYSIOLOGY
I. Introduction
Aε described above, the present invention relates to inhibition of dysfunctions of the immune and hemostatic systems during ECC. To provide background for the description of the preferred embodiments and the examples presented below, we turn first to general discussions in the context of ECC of the relevant aspects of the immune and hemostatic systems, in particular, certain aspects of the complement arm of the immune system, the cellular arm of the immune system, and the physiology of plateletε.
II. Pathophvεiologv Associated With ECC
A key pathophysiologic change in the blood that iε aεεociated with ECC iε the rapid activation of the complement cascade. Activation of complement components that mediate inflammation and impact the hemostatic properties of the blood occurs when blood comes in direct contact with the various non-biological components of the ECC circuit (Videm, et al., J. Thorac. Cardiovasc. Surg., 97:764-770, 1989) , and can be inhibited to some extent by certain drugs, particularly heparin and protamine, that are administered to patients during ECC procedures (Jones, et al., Anaesthesia. 37:629-633, 1982) .
Interdependent disturbances in both the immune system and the hemostatic system are seen during CPB and other types of ECC. Many of these changes intersect at the level of platelet function, as the actions of the complement arm of the immune system can reεult in platelet activation.
In addition to its effects on platelets and the complement arm of the immune system, ECC also effects the cellular arm of the immune system both through effects on leukocytes and through effects on platelet-leukocyte interactions.
(a) Effects of ECC on Platelets
Plateletε are anuclear, cellular elements of the blood that are vitally important for the formation of blood clots and the prevention
" of excessive bleeding. An abnormally severely low platelet count in the blood, a condition known as thrombocytopenia, often results in severe bruising, hemorrhage from mucosal membranes, and considerable loss of blood following surgery or other injury.
Platelet dysfunction has been linked with the contact of platelets with the non-biological surfaces of the extracorporeal oxygenator and the hypothermia associated with CPB. Several other mechanisms, alone or in combination, have also been implicated as contributing to platelet dysfunction. For example, mechanical trauma due to shear stress, surface adherence, and turbulence within the extracorporeal oxygenator may cause fragmentation of platelet membranes. Activated complement components, however, are arguably the most important factors contributing to the platelet dysfunction associated with ECC.
CPB adversely affects platelet count as well as function. Hemodilution during CPB causes platelet counts to rapidly decrease soon after εtarting CPB, declining to about 50% of preoperative levels. This level of circulating plateletε, if occurring in the context of normal individual platelet function, is unlikely to contribute to clinical bleeding. Of greater significance to the development of CPB associated morbidity, however, iε the progreεsive losε of platelet function seen during and after CPB. Within minuteε after initiating CPB, bleeding time iε prolonged significantly and platelet aggregation is impaired. These changes in bleeding time are independent of platelet count and worsen as CPB progresses. Bleeding times, normally less than 10 minuteε, can approach 30 minutes after 2 hours of CPB.
Plateletε undergo profound biochemical and morphological alterations when activated by certain stimuli. When caused by stimuli associated with conditions calling for rapid hemostasis, these alterations are associated with the normal functions of plateletε. When cauεed by ECC, pathophyεiologic outcomes result. Activation-induced alterations in platelet characteriεticε include exocytotic degranulation with the release of the contents of various storage organelles, shape changeε, and the induction of adheεiveneεε, aggregation, and thromboxane production. A εpecific integral membrane protein, P-εelectin (alεo known as CD62, CD62P, GMP-140, and PADGEM) , which iε located on the inner surfaces of platelet alpha granules, iε translocated to the platelet surface as these organelles turn inside-out during exocytosis. The appearance of P-selectin on the platelet surface thus serves as a marker of platelet activation. Various lines of evidence εuggeεt that the expreεεion of P- εelectin and the temporary depletion or modification of εome functional platelet membrane componentε are key measures of platelet functionality. Several studies have reported significant decreaseε in the amount of glycoprotein membrane antigens GPIb (CD45b) , GPIIb, and GPIIIa on circulating platelets following CPB. The loss of platelet GPIIIa in particular iε associated with a reduction in total fibrinogen binding sites. Such surface altered platelets thus demonstrate poor hemostatic capacity.
The removal of platelet membrane glycoproteins may be mediated by the activity of proteolytic enzymes such as plaεmin and elastase. The ability of the protease inhibitor aprotinin (discussed above) to minimize glycoprotein (specifically, GPIb) loss after CPB provides evidence implicating direct proteolyεis of platelet membrane glycoproteins in the development of platelet dyεfunction (Wenger, et al., J Thorac Cardiovaεc Surg. 97:235, 1989) .
Effects of ECC on platelets are particularly significant because platelets can only be activated once, i.e., activation of platelets decreaseε the number of functional platelets available when platelet functions are subsequently required. The importance of the effects of ECC on platelets is demonεtrated by the finding that the impaired hemostasis observed after cardiac operations is mainly attributable to platelet dysfunction (Mohr, et al., J. Thorac. Cardiovasc. Surg. 96:530, 1988).
(b) Platelet-Leukocyte Interactions Associated with ECC The P-selectin molecule, which appears on the membrane surface of activated plateletε, iε known to mediate the binding of platelets to various types of white blood cells (WBCs, leukocytes) without requiring the activation of the WBCs for such binding to occur. These WBCs include polymorphonuclear leukocytes (PMNs, neutrophilε, granulocytes) , and monocytes, and the P-selectin mediated binding results in platelet-PMN, and platelet-monocyte conjugate formation (Larεen, et al. , J Biol Chem 267:11104-11110, 1992; Corral, et al. , Biochem Biophvs Res Commun 172:1349-1353, 1990) . One result of such conjugate formation is the removal of platelets from the circulation, a phenomenon that can contribute to the development of thrombocytopenia (Rinde, et al. , Transfusion, 31:408-414, 1991).
Such leukocyte-platelet adhesion iε also believed to be of physiologic importance in the targeting of leukocytes to appropriate inflammatory and/or hemostatic sites and in modulating leukocyte function. The relevance of εuch targeting has been recently demonstrated in vivo in a baboon model where blockade of P-selectin with a monoclonal antibody resulted in decreased monocyte accumulation on an artificial vascular graft and decreaεed procoagulant activity (Palabrica, et al. , Nature 359:848-851, 1992) . Such leukocyte-platelet adheεion caused by P-selectin has been found to be induced by CPB (Rinder, et al., Blood. 79:1201-1205, 1992).
The modulation of leukocyte functionmediated by ECC-associated platelet-leukocyte binding includes the upregulation of the major monocyte procoagulant molecule known as "tissue factor" (TF) . Data reported by Catalett, et al. , Blood 78 (Suppl 1) :279a, 1991, suggests that P-selectin induces TF upregulation on monocytes over a four hour period. Such TF upregulation enhances the pro-coagulatory effects of leukocytes (Altieri, Blood 81:569-579, 1993). In a simulated closed-loop CPB model, Kappelmayer, et al. , Circ Res 72:1075-1081, 1993, have recently demonstrated CPB-asεociated upregulation of both the quantity and activity of TF on circulating monocyteε. The expreεsion of monocyte tissue factor several hours after the conclusion of a CPB procedure, combined with other procoagulatory events occurring during CPB (Evangelista, et al. , Blood 77:2379-2388, 1991; Higuchi, et al.. Blood 79:1712-1719, 1992; Weitz, et al. , J Exp Med 166:1836-1850, 1987) iε believed to prediεpoεe the patient to late thrombotic events, such as vascular graft re-occlusion, (c) Other Effects of ECC on Leukocvteε
In addition to upregulation of TF on monocyteε, CPB-aεsociated upregulation of cell adhesion ligands that contain the β2 integrin CD18, in particular, the heterodimeric adhesive ligand that contains CD18 and CDllb (known as CDllb/CD18 or MAC-1) , has been described on monocytes and neutrophils (Rinder, et al. , Blood 79:1201-1205, 1992) . Such upregulation is particularly relevant to CPB-induced injury since CDllb/CD18 is responsible for leukocyte adherence to and penetration (diapedeεis) through the endothelium via binding to the intercellular adheεion molecules ICAM-l and ICAM-3 on "activated" endothelium (Staunton, et al. , Cell 52:925-933, 1988; Staunton, et al., Nature 339:61-64, 1989; Furie, et al. , J Immunol 148:2395-2404, 1992) .
In experimental systems, administration of monoclonal antibodies blocking the activity of CDllb/CD18 haε been εhown to prevent reperfusion injury (Simpson, et al. , J Clin Invest 81:624- 629, 1988; Arnaout, Blood 75:1037-1050, 1990; Dreyer, et al. , Circ 84:400-411, 1991) . In addition, increased CDllb/CD18 expression on leukocytes has been linked to complications associated with hemodialysiε (Arnaout, et al. , N Engl J Med 312:457, _985) . Thuε, CDllb/CD18 may contribute to ECC associated medical problems.
In addition to the foregoing, the CDllb moiety functionε aε the receptor for complement component iC3b and for elements of the contact dependant soluble coagulation system, including fibrinogen, fibronectin, and Factor X (Altieri, et al., Proc Natl Acad Sci USA 85:7462-7465, 1988; Nathan, et al. , J Cell Biol 109:1341-1349. 1989). The interactions of CDllb with fibrinogen may be of particular importance to ECC-associated pathology, since, aε diεcuεεed above, the platelet εurface glycoprotein complex GPIIb-IIIa is also associated with fibrinogen binding, thus providing a potential means for platelet-leucocyte adhesion, i.e., via a CDllb--fibrinogen-- GPIIb-IIIa linkage (Altieri, et al. , Proc Natl Acad Sci USA 85:7462- 7465, 1988) . Induction of an inflammation-associated respiratory burst in granulocytes in response to a variety of cytokines also appears to require CDllb/CD18 in the local adherent microenvironment.
Thuε this β2 integrin complex exerts its effects at critical junctures of the complement system, the cellular immune εyεtem, and the soluble cellular coagulation pathway. CDllb/CD18 expression on leukocyte surfaces can thuε be assayed as a predictive indicator related to the probability of the occurrence of complement, immune, and coagulation asεociated eventε. (d) The Complement System
The complement εystem acts in conjunction with other im unological systems of the body to defend against intrusion of cellular and viral pathogens. There are at least 25 complement proteins, which are found as a complex collection of plasma proteins and membrane cofactors. The plasma proteinε make up about 10% of the globulins in vertebrate serum. Complement components achieve their immune defensive functions by interacting in a series of intricate but precise enzymatic cleavage and membrane binding events. The resulting complement cascade leadε to the production of productε with opεonic, immunoregulatory, and lytic functionε.
The complement cascade progresεes via the claεεical pathway or the alternative pathway. These pathways share many components, and while they differ in their initial steps, they converge and share the same "terminal complement" components responsible for the activation, attack, and destruction of target cells.
The classical complement pathway is typically initiated by antibody recognition of and binding to an antigenic site on a target cell. The alternative pathway is usually antibody independent, and can be initiated by certain moleculeε on pathogen εurfaceε. Both pathwayε converge at the point where complement component C3 iε cleaved by an active protease (which is different in each pathway) to yield C3a and C3b. Other pathways activating complement attack can act later in the sequence of events leading to variouε aεpectε of complement function.
C3a is an anaphylotoxin (see below) . C3b binds to bacterial and other cells and tags them for removal from the circulation. (C3b in this role is known as opsonin.) The opεonic function of C3b iε conεidered to be the moεt important anti-infective action of the complement εyεtem. Patientε with genetic leεionε that block C3b function are prone to infection by a broad variety of pathogenic organisms, while patients with lesions later in the complement cascade sequence, i.e., patientε with leεionε that block C5 functions, are found to be more prone only to Neisseria infection, and then only somewhat more prone (Fearon, in Intensive Review of Internal Medicine. 2nd Ed. Fanta and Minaker, edε. Brigham and Women's and Beth Israel Hospitals, 1983) .
C3b also forms a complex with other componentε unique to each pathway to form classical or alternative C5 convertase, which cleaves C5 into C5a and C5b. C3 is thus regarded as the central protein in the complement reaction εequence εince it iε eεεential to both the alternative and clasεical pathways (Wurzner, et al. , Complement Inflamm 8:328-340, 1991) . This property of C3b iε regulated by the serum protease Factor I, which acts on C3b to produce iC3b. While still functional aε opsonin, iC3b cannot form an active C5 convertase.
C5a is another anaphylatoxin (see below) . C5b combines with C6, C7, and C8 to form the C5b-8 complex at the surface of the target cell. Upon binding of several C9 molecules, the membrane attack complex (MAC, C5b-9, terminal complement complex -- TCC) is formed. When sufficient numbers of MACs insert into target cell membranes the openings (MAC poreε) they create mediate rapid oεmotic lyεiε of the target cells. Lower, non-lytic concentrations of MACs can produce other effects. In particular, membrane insertion of small numbers of the C5b-9 complexes into endothelial cells and platelets can cause deleterious cell activation. In some caseε activation may precede cell lysis.
Certain aspects of the mechanism of C5b-9 mediated platelet activation have been described (Sims, et al. , J Biol Chem. 264:19228, 1989) . Sublytic concentrations of MAC pores on platelet surfaces allow an influx of extracellular calcium ions into the platelet. The resulting elevated levels of intraplatelet calcium ions act as an activating signal and induce the surface expression of P-selectin that iε characteriεtic of activated platelets. Since, as described above, P-selectin mediates platelet-monocyte binding, and such binding triggers the release of tissue factor from monocytes, the formation of MAC pores in platelet surface membranes can have deleterious effects on patients. For example, platelet activation by MAC pores can predispose a CPB patient to late thrombotic events, such as vascular graft re-occlusion. As mentioned above, C3a and C5a are anaphylatoxins. These activated complement components can trigger mast cell degranulation, which releases histamine and other mediators of inflammation, resulting in smooth muscle contraction, increased vascular permeability, leukocyte activation, and other inflammatoryphenomena.
C5a also functionε as a chemotactic peptide that seirves to attract pro-inflammatory granulocytes to the site of complement activation.
In addition, direct activation of endothelial cells by C5a induces the release of the coagulation-inhibiting glycoprotein heparan sulfate. The release of heparan sulfate can have the effect of decreasing the levels of this coagulation-inhibiting molecule at the endothelial cell surface, with the conεequence of increasing the thrombogenic potential of the endothelial cell surface (Platt, et al., J Exp Med 171:1363, 1990). (e) Complement and Platelet Function and ECC
The activation and consumption of complement components during ECC, εpecifically CPB, is evidenced by levels of hemolytic complement activity that are much lower at the end of CPB than can be explained by hemodilution alone. A rapid effect on complement components accompanies the initiation of extracorporeal circulation; evidence of alternative pathway activation is observed minutes after the onεet of CPB. Claεsical pathway activation has also been obεerved during CPB (Haεlam, et al. , Anaesthesia 35:22, 1980).
The levelε of C3a anaphylatoxin have been found to increase dramatically during CPB, and there is a strong statistical association between elevated C3a levels and postoperative organ system dysfunction manifest by impairment and/or failure of cardiac, renal and pulmonary syεtemε, bleeding diatheεiε, and the need for artificial ventilation
(Kirklin, et al. , J Thorac Cardiovasc Surg 86:845-847, 1983). This asεociation haε led to the belief that C3a is a key mediator of the deleterious effects linked to complement activation during CPB.
It is well' eεtablished that platelets can be activated by the assembly of terminal complement components C5b-9 on their surfaces. The assembly of these complement components on platelets is known to occur during ECC (Finn, et al. , J Thorac Cardiovasc Surg.
105:234, 1993). Complement-mediated platelet activation, in turn, leadε to alpha-granule releaεe, increaεed expression of P-selectin, and the loεs of GPIb. The generation of products of complement activation such as C3a, C5a, and C5b-9 further results in platelet membrane vesiculation and consequent microparticle formation. Other damaging effects of complement activation during CPB can include the activation of granulocytes, leading to partial degranulation and up-regulation of CDllb/CD18, and to damage to organs. Such damaging effects are largely due to the actions of certain products of complement activation, specifically the anaphylatoxins C3a and C5a, which, in turn, can be converted to desArg forms with altered activity levels by plasma carboxypeptidase. These activated complement components cause activation and aggregation of neutrophils. Such activated cells, in turn, accumulate in the pulmonary vessels and vascular beds, as haε been demonstrated by serial biopsies of lung tissue before and after CPB (Howard, et al. , Arch Surg. 123:1496-1501, 1988). Liver, brain and pancreas, also suffer such damage, which can result in postoperative dysfunction of these organs.
Although circumstantial data indicates that activated complement components C3a, C5a, and C5b-9 play a critical role in the pathophyεiology of CPB, to date definitive evidence haε been lacking. Additionally, the relative contribution of theεe individual complement componentε to the generation of the inflammatory proceεε aεsociated with CPB haε remained undefined. DESCRIPTION OF THE PREFERRED EMBODIMENTS
As discussed above, the present invention relates to the use of anti-C5 antibodies in ECC. More generally, the invention relates to the use of anti-C5 antibodies in any procedure which involves circulating the patient's blood from a blood vessel of the patient, through a conduit, and back to a blood vesεel of the patient, the conduit having a luminal surface comprising a material capable of causing at least one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion. Such procedures include, but are not limited to, all forms of ECC, as well aε procedureε involving the introduction of an artifical or foreign organ, tissue, or vessel into the blood circuit of a patient. Conduit materials which have the foregoing effects on blood include those now known in the art as well as those which may be developed in the future. Such materials include synthetic materials such as the various forms of plastics and synthetic polymers used in CPB circuits, hemodialyεis circuits, vascular grafts, and artificial organs, as well aε biological materials of xenogeneic origin.
The anti-C5 antibody is used to reduce at least one of complement activation, platelet activation, leukocyte activation, or platele -leukocyte adhesion resulting from the circulation of the patient's blood through such a conduit. Reduction of complement activation can be measured by methodε well known in the art, for example, by the chicken erythrocyte hemolysiε method described below in Example 1. Preferably, a sufficient amount of antibody is used to reduce the increase in plasma hemolytic activity resulting from complement activation caused by contact with the conduit by at least 50%.
Reduction of platelet activation can be meaεured by various methods well known in the art, for example, by immunofluoreεcence analysis of the expression of platelet activation markers GPIIb-IIIa or P-εelectin by the methods described below in Examples 1 and 4. Again, it is preferred to use a sufficient amount of antibody so aε to achieve at leaεt a 50% reduction in the increase in the expreεsion of at least one of the platelet activation markers resulting from contact with the conduit. Reduction of leukocyte activation can be meaεured by various methods well known in the art, for example, by immunofluorescence analysis of the expreεεion of the leukocyte activation marker CDllb/CD18. As described in Example 4, the expression of this marker can be determined by the measurement of CDllb expression. Again, it iε preferred to uεe a εufficient amount of antibody εo aε to achieve at leaεt a 50% reduction in the increaεe in the expression of the leukocyte activation marker by at least one type of leukocyte, e.g., PMNs, reεulting from contact with the conduit.
Reduction of the levelε of platelet-leukocyte adhesion, specifically platelet-monocyte and platelet-neutrophil adhesion, can be measured by the fluorescence analysis techniques described below in Example 4, e.g., as the percent increase in leukocytes that read as GPIb or GPIIb/IIIa positive. Again, it is preferred to use a sufficient amount of antibody so as to achieve at least a 50% reduction in the increase in the percentage of GPIb or GPIIb/IIIa positive monocytes or neutrophils resulting from contact with the conduit.
To achieve the desired reductions, the anti-C5 antibodies can be administered in a variety of unit dosage forms. The dose will vary according to, e.g., the particular antibody, the manner of administration, the particular procedure being performed and its duration, the nature of the conduit or conduits used in the procedure, the overall health, condition, size, and age of the patient, and the judgment of the prescribing physician. Dosage levels for human subjects are generally between about 1 mg per kg and about 100 mg per kg per patient per treatment, and preferably between about 5 mg per kg and about 50 mg per kg per patient per treatment. In some cases, only a single dose of the anti-C5 antibody can be administered to the patient in order to achieve a therapeutic effect.
The anti-C5 antibody is preferably administered prior to contact of the blood with the conduit. In addition, administration can be continued for as long as the blood is in contact with the conduit, and even after that time, if desired. In the caεe of prior administration, a single dose can be used. For adminiεtration continuing throughout the procedure and/or thereafter, periodic doεes or a continuous infusion of the anti-C5 antibody can be used.
A typical procedure, in accordance with the present invention, thus includes the following steps. The patient iε prepared for surgery in accordance with standard practice, and within an hour of the commencement of surgery, but generally before the commencement of the surgery, a first dose of the pharmaceutical preparation containing the anti-C5 antibody is given. Usually, as determined by the judgement of the physician, a series of doses may also be administered during and after the procedure. Alternatively, during and after the procedure is conducted, levels of serum complement activity available in the patient's blood are monitored using the techniques set forth below in Example 1 to determine if additional doses are required, with such doses being administered as required to maintain at least a 50% reduction, and preferably about a 95% reduction of serum complement activity.
Administration of the anti-C5 antibodies will generally be performed by an intravascular route, e.g., via intravenous infusion by injection. Other routes of administration may be used if desired. Formulations suitable for injection are found in Remington's Pharmaceutical Sciences. Mack Publishing Company, Philadelphia, PA, 17th ed. (1985) . Such formulations must be sterile and non-pyrogenic, and generally will include a pharmaceutically effective carrier, such as saline, buffered (e.g., phosphate buffered) saline, Hank's solution. Ringer's solution, dextrose/saline, glucose εolutionε, and the like. The formulationε may contain pharmaceutically acceptable auxiliary εubεtances as required, such as, tonicity adjusting agents, wetting agents, bactericidal agents, preservatives, stabilizers, and the like.
The formulations (pharmaceutical agents) of the invention can be distributed aε articles of manufacture comprising packaging material and the anti-C5 antibody. The packaging material will include a label which indicates that the formulation is for use in asεociation with an extracorporeal circulation procedure, e.g., a cardiopulmonary bypasε procedure.
The anti-C5 antibody iε preferably a monoclonal antibody, although polyclonal antibodies produced and εcreened by conventional techniques can also be used if desired. Preferably, the anti-C5 antibodies are immunoreactive against epitopes on the beta chain of purified human complement component C5 (i.e., the part of the C5 molecule that becomeε C5b) and are capable of blocking the conversion of C5 into C5a and C5b by C5 convertase. This capability can be measured uεing the techniqueε described in Wurzner, et al. , Complement Inflamm 8:328-340, 1991. The amount of reduction of C5 convertaεe activity iε preferably at least about 50% of the increase in convertase activity resulting from contact with the conduit.
Hybridomas producing monoclonal antibodies reactive with complement component C5 can be obtained according to the teachings of Sims, et al. , U.S. Patent No. 5,135,916. Aε discussed therein, antibodieε are prepared uεing purified componentε of the complement membrane attack complex aε immunogens. In accordance with the present invention, complement component C5 or C5b is preferably used as the immunogen. In accordance with the present invention, the antibodies preferably should prevent the cleavage of C5 to form C5a and C5b, thus preventing the generation of the anaphylatoxic activity asεociated with C5a and preventing the assembly of the membrane attack complex associated with C5b. In a particularly preferred embodiment, these anti-C5 antibodies will not impair the opsonization function asεociated with the activation of complement component C3 by a C3 convertase. Plasma C3 convertase activity can be measured by assaying plasma for the presence of C3a as described below in Example 2. Preferably, the anti-C5 antibody produces essentially no reduction in plasma C3a levels. General methods for the immunization of animals (in this caεe with C5 or C5b) , iεolation of antibody producing cellε, fuεion of εuch cells with immortal cellε (e.g., myeloma cellε) to generate hybridomaε εecreting monoclonal antibodieε, screening of hybridoma supernatants for reactivity of secreted monoclonal antibodies with a desired antigen (in this case C5 or C5b) , the preparation of quantities of such antibodies in hybridoma supernatants or asciteε fluidε, and for the purification and storage of such monoclonal antibodieε, can be found in numerous publications. These include: Coligan, et al. , eds. Current Protocolε In Immunology. John Wiley & Sonε, New York, 1992; Harlow and Lane, Antibodieε. A Laboratory Manual. Cold Spring Harbor Laboratory, New York, 1988; Liddell and Cryer, A Practical Guide To Monoclonal Antibodieε. John Wi_ ' & Sonε, Chichester, West Sussex, England, 1991; Montz, et al. , Cellular Immunol. 127:337-351, 1990; Wurzner, et al. , Complement Inflamm 8:328-340, 1991; and Mollnes, et al., Scand. J. Immunol. 28:307-312, 1988.
Aε used herein, the term "monoclonal antibodies" refers to immunoglobulins produced by a hybridoma and antigen binding fragments (e.g., Fab' preparations) of such immunoglobulins, as well as to recombinantly expressed antigen binding proteins, including immunoglobulins, chimeric immunoglobulins, "humanized" immunoglobulins, antigen binding fragments of such immunoglobulins, εingle chain antibodieε, and other recombinant proteins containing antigen binding domainε derived from immunoglobulinε. Publications describing methods for the preparation of such monoclonal antibodieε, in addition to those listed immediately above, include: Reichmann, et al., Nature, 332:323-327, 1988; Winter and Milstein, Nature, 349:293- 299, 1991; Clackson, et al . , Nature. 352:624-628, 1991; Morrison, Annu Rev Immunol, 10:239-265, 1992; Haber, Immunol Rev. 130:189-212, 1992; and Rodrigueε, et al . , J Immunol. 151:6954-6961, 1993.
Without intending to limit it in any manner, the present invention will be more fully described by the following examples.
EXAMPLE 1 Anti-C5 Monoclonal Antibody Inhibition of Complement Activity During
Extracorporeal Circulation The effectε of a monoclonal antibody raised to human C5 on complement activation was evaluated using a closed-loop CPB model for the extracorporeal circulation of human blood. The C5 monoclonal antibody was raised in mice against purified human C5 protein (Wurzner, et al. , Complement Inflamm 8:328-340, 1991; N19-8 mAb) and was propagated, recovered and purified aε an IgG fraction from mouse asciteε fluid (Antibodieε. A Laboratory Manual, Cold Spring Harbor Laboratory, New York, 1988; Current Protocols In Immunology, John Wiley & Sons, New York, 1992) .
To carry out theεe experimentε, 300 ml of whole human blood was drawn from a healthy human donor and additionally a 1 ml sample was removed aε a control εample for later analysiε. The blood waε diluted to 600 ml by the addition of Ringer's lactate solution containing lOU/ml heparin. The blocking anti-C5 mAb (30 mg in sterile PBS) was added to the diluted blood to a final concentration of 50 μg/ml. In a control experiment, an equal volume of sterile PBS was added to the diluted blood. The blood was then used to prime the extracorporeal circuit of a COBE CML EXCEL membrane oxygenator CPB machine (Cobe BCT, Inc., Lakewood, CO)and the circuit waε εtarted. The circuit was cooled to 28°C and circulated for 60 min. The circuit was then warmed to 37°C and circulated for an additional 30 min. The experiment waε then terminated. Sampleε were taken at several time pointε and evaluated for complement activity (Figure 1) . At each time point an aliquot of whole blood waε taken, divided into 3 εampleε and A) diluted 1:1 in 2% paraformaldehyde in PBS to evaluate platelet and blood cell activation parameterε (Example 4) ; B) centrifuged to remove all cellε and plaεma diluted 1:1 in Quidel εa ple preεervation εolution (Quidel Corporation, San Diego, CA) and εtored at -80°C to evaluate C3a and C5b-9 generation (Exampleε 2 and
3, reεpectively) , and C) centrifuged to remove all cellε and plasma frozen at -80°C for hemolytic assays to evaluate complement activity (Example 1) . To evaluate complement activity, the frozen plasma samples were thawed and hemolytic assayε were performed aε follows. Chicken erythrocytes were washed well in GVBS (Rollins, et al. , J Immunol 144:3478-3483, 1990) and resuspended to 2xl08/ml in GVBS. Anti- chicken erythrocyte antibody (IgG fraction of anti-chicken-RBC antiserum, Intercell Technologies, Hopewell, NJ) was added to the cells at a final concentration of 25 μg/ml and the cells were incubated for 15 min. at 23°C. The cells were washed 2x with GVBS and 5xl06 cells were resuspended in a final volume of 130 μl GVBS containing 5% plasma. After incubation for 30 min. at 37°C, % hemolysis was calculated relative to a human serum control sample (Figure 1) . Hemolysis was determined by spinning the cells down and measuring released hemoglobin in the supernatant as the optical density at 4l5nm.
As can be seen in Figure 1, addition of the anti-C5 antibody to the extracorporeal circuit resulted in a 95% reduction in serum complement activity. The complement activity remained inhibited throughout the course (90 minuteε) of the experiment.
EXAMPLE 2 Generation of C3a During Extracorporeal Circulation The fresh frozen plaεma samples that had previously been diluted in Quidel sample preservation solution (see Example 1) were assayed for the presence of the complement split product C3a by using the Quidel C3a EIA kit (Quidel Corporation, San Diego, CA) . These measurements were carried out according to the manufacturers specifications. C3a released is expressed in ng/well aε determined by comparison to a standard curve generated from samples containing known amounts of human C3a.
As seen in Figure 2, addition of the anti-C5 antibody had no effect on the production of C3a during the CPB experiment. C3a generation was dramatically increased during the final 30 min. of the experiment and correlates with sample warming.
EXAMPLE 3 Prevention of the Generation of C5b-9 During Extracorporeal
Circulation Fresh frozen plaεma samples that had been previously diluted in
Quidel sample preservation εolution (εee Example 1) were aεsayed for the presence of the terminal human complement complex C5b-9 using the
Quidel C5b-9 kit (Quidel Corporation, San Diego, CA) . The amount of
C5b-9 in each sample was determined using the manufacturers specificationε and iε expressed in arbitrary abεorbance units (AU) .
As can be seen in Figure 3, the anti-C5 antibody completely inhibited C5b-9 generation during extracorporeal circulation so that
C5b-9 levelε during the full courεe of the run were equivalent to control (t0) time pointε. The reεults of this experiment and those of Examples 1 and 2 εhow that the addition of an anti-C5 mAb during extracorporeal circulation effectively inhibitε both complement hemolytic activity (Figure 1) and C5b-9 generation (Figure 3) , but not
C3a generation (Figure 2) .
EXAMPLE 4 Prevention of Platelet and Leukocyte Activation and Adheεion During
Extracorporeal Circulation
Paraformaldehyde fixed samples of whole blood taken at T=0 and
T=90 minutes during the extracorporeal circulation experiment (see
Example l) were washed, labeled with fluorescent antibodies directed against various cell surface marker molecules, and analyzed by flow cytometry (FACS) . The primary antibodies used in these experiments were anti-GPIIb-IIIa (clone RUUSP2.41, AMAC, Inc., Westbrook, ME); anti-CDllb (Anti-CR3 (leu-15) clone D12, Becton-Dickinson
Immunocytometry Syεtemε, San Jose, CA) ; anti-P-selectin, (1E3 obtained from Dr. Kenneth Ault, Maine Medical Center Research Institute,
Portland, ME); anti-GPIb (CD42b) , (IOP42b clone SZ2, AMAC, Inc., Westbrook, ME) , and anti-CD45 (a marker for both monocytes and PMNs, which are distinguished by the forward and side εcatter characteriεtics of the FACS profiles of the CD45 positive cells) (Anti-Leucocyte (HLe-1; CD45) clone 2D1, Becton-Dickinson Irnmunocytometry Systemε, San Jose, CA) . The percent change in surface expresεion of variouε cell εurface receptors (Platelet GPIb, Platelet GPIIb-IIIa, P-selectin, CDllb) waε calculated by comparison of mean FL-1 values obtained for each antibody by the following calculation: T=90 / T=0 x 100. Platelet monocyte binding (PLT/MONO) and platelet neutrophil binding (PLT/PMN) waε determined by calculating the percentage of neutrophilε or monocyteε that read positive for the platelet-specific receptor GPIb.
As can be seen in Figure 4, the addition of anti-C5 antibody to the blood in the ECC circuit had a dramatic effect on various aspects of platelet and blood cell activation. In the presence of the anti-C5 antibody, platelets retained 50% more GPIb as compared to non- reated controls. Similarly, platelets retained virtually all of their cell surface GPIIb-IIIa while untreated plateletε lost 80% of this receptor. Platelet activation as determined by expression of P- selectin was completely inhibited by the addition of the anti-C5 antibody. Conjugate formation by plateletε and monocyteε and/or plateletε and neutrophilε waε inhibited by addition of the anti-C5 antibody. Additionally, expression of CDllb on neutrophilε was blocked by 75% by anti-C5 while CDllb expresεion on monocytes was unaffected.
These resultε εhow that the anti-C5 antibody is a potent inhibitor of complement-induced platelet activation, leukocyte activation, specifically, PMN activation, and platelet-leukocyte adhesion during extracorporeal circulation and that terminal complement components, and not C3a, are largely reεponsible for complement-mediated inflammation and platelet dysfunction.
Throughout this application various publicationε, patents, and patent applications are referred to. The teachings and discloεures of these publications, patents, and patent applications in their entireties are hereby incorporated by reference into this application to more fully describe the state of the art to which the present invention pertains.
Although preferred and other embodiments of the invention have been described herein, further embodiments may be perceived by those skilled in the art without departing from the scope of the invention as defined by the following claims.

Claims

What iε claimed iε:
1. A method for performing a therapeutic procedure on a patient comprising:
(a) passing circulating blood from a blood vessel of the patient, through a conduit, and back to a blood veεεel of the patient, the conduit having a luminal εurface comprising a material capable of causing at least one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion in the patient's blood; and
(b) introducing an antibody that specifically binds to complement component C5 into the patient's bloodstream in an amount effective to reduce at leaεt one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion resulting from passage of the circulating blood through said conduit, wherein step (a) occurs before and/or during and/or after step (b) .
2. The method of Claim 1 wherein the antibody reduces the conversion of complement component C5 into complement components C5a and C5b.
3. The method of Claim 1 wherein the antibody specifically binds to C5b. . The method of Claim 1 wherein the antibody does not substantially inhibit formation of complement component C3b.
5. The method of Claim 1 wherein the therapeutic procedure is an extracorporeal circulation procedure.
6. The method of Claim 5 wherein the extracorporeal circulation procedure is a cardiopulmonary bypasε procedure.
7. An article of manufacture compriεing packaging material and a pharmaceutical agent contained within said packaging material, wherein:
(a) said pharmaceutical agent comprises an antibody that specifically binds to complement component C5, said antibody being effective for reducing at least one of complement activation, platelet activation, leukocyte activation, or platele -leukocyte adhesion caused by passage of circulating blood from a blood vessel of a patient, through a conduit, and back to a blood vessel of the patient, the conduit having a luminal surface comprising a material capable of causing at least one of complement activation, platelet activation, leukocyte activation, or platelet-leukocyte adhesion in the patient's blood; and
(b) said packaging material comprises a label which indicates that said pharmaceutical agent is for use in association with an extracorporeal circulation procedure.
8. The article of manufacture of Claim 7 wherein the label indicates that said pharmaceutical agent is for use in association with an cardiopulmonary bypass procedure.
PCT/US1995/003614 1994-03-23 1995-03-22 Method for reducing immune and hemostatic dysfunctions during extracorporeal circulation WO1995025540A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP95914820A EP0751787B1 (en) 1994-03-23 1995-03-22 Method for reducing immune and hemostatic dysfunctions during extracorporeal circulation
DE69533921T DE69533921T2 (en) 1994-03-23 1995-03-22 METHOD FOR REDUCING MALFUNCTIONS OF IMMUNE SYSTEM AND HEMOSTASE DURING EXTRACORPOREAL CIRCULATION
AU21917/95A AU2191795A (en) 1994-03-23 1995-03-22 Method for reducing immune and hemostatic dysfunctions during extracorporeal circulation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US21739194A 1994-03-23 1994-03-23
US08/217,391 1994-03-23

Publications (1)

Publication Number Publication Date
WO1995025540A1 true WO1995025540A1 (en) 1995-09-28

Family

ID=22810875

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/003614 WO1995025540A1 (en) 1994-03-23 1995-03-22 Method for reducing immune and hemostatic dysfunctions during extracorporeal circulation

Country Status (7)

Country Link
US (1) US5853722A (en)
EP (1) EP0751787B1 (en)
AU (1) AU2191795A (en)
CA (1) CA2186108A1 (en)
DE (1) DE69533921T2 (en)
ES (1) ES2236706T3 (en)
WO (1) WO1995025540A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0777474A1 (en) * 1994-09-23 1997-06-11 Alexion Pharmaceuticals, Inc. Methods for the treatment of inflammatory joint disease
WO1999010009A1 (en) * 1997-08-26 1999-03-04 Gliatech Inc. A process for inhibiting complement activation via the alternative pathway
WO2003061765A1 (en) * 2002-01-22 2003-07-31 Alexion Pharmaceuticals, Inc. Method of prophylaxis against large myocardial infarctions
US7361339B2 (en) 2003-01-09 2008-04-22 Alexion Pharmaceuticals, Inc. Methods for reducing morality associated with acute myocardial infarction
EP2886140A1 (en) * 2013-12-17 2015-06-24 University of Limerick An apparatus for the extracorporeal treatment of blood

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7112327B2 (en) 1998-02-20 2006-09-26 Tanox, Inc. Inhibition of complement activation
US6956107B2 (en) * 1998-02-20 2005-10-18 Tanox, Inc. Inhibitors of complement activation
EP3056516A1 (en) * 1998-02-20 2016-08-17 Genentech, Inc. Inhibitors of complement activation
WO1999047190A1 (en) * 1998-03-16 1999-09-23 Medtronic, Inc. Hemostatic system and components for extracorporeal circuit
DE19913707A1 (en) 1999-03-26 2000-10-05 Privates Inst Bioserv Gmbh Immunosorbent for sepsis therapy
DE60118362T2 (en) * 2000-05-19 2007-05-24 The Center for Blood Research, Inc., Boston METHOD FOR TREATING HEMOSTATIC INTERFERENCE BY SOLUBLE P-SELECTIN
PT1325033E (en) * 2000-10-10 2010-04-15 Genentech Inc Inhibition of complement c5 activation for the treatment and prevention of delayed xenograft or acute vascular rejection
KR100899970B1 (en) 2001-02-19 2009-05-28 메르크 파텐트 게엠베하 Method for identification of t-cell epitopes and use for preparing molecules with reeduced immunogenicity
CA2454562A1 (en) * 2001-07-26 2003-02-06 Alexion Pharmaceuticals, Inc. Method of improving cognitive function
WO2003012396A2 (en) * 2001-08-01 2003-02-13 Chauhan Anil K Immune complexes
EP3372243A1 (en) 2001-08-17 2018-09-12 Genentech, Inc. Complement pathway inhibitors binding to c5 and c5a without preventing formation of c5b
ES2283594T5 (en) 2001-08-17 2016-03-15 Genentech, Inc. Inhibitors of the complement pathway that bind to C5 and C5a without preventing the formation of C5b
WO2004071446A2 (en) * 2003-02-12 2004-08-26 Chauhan Anil K Immune complexes
US7135538B2 (en) 2003-11-12 2006-11-14 General Electric Company Transparent polycarbonate-polysiloxane copolymer blend, method for the preparation thereof, and article derived therefrom
WO2005118020A1 (en) * 2004-04-21 2005-12-15 Allvivo, Inc. Surface coating comprising bioactive compound
NZ554734A (en) 2004-12-08 2009-03-31 Sirion Therapeutics Inc Methods, Assays And Compositions For Treating Retinol-Related Diseases
CN107929731A (en) * 2005-11-04 2018-04-20 健泰科生物技术公司 Utilize complement pathway inhibitors to treat ocular diseases
ES2700609T3 (en) 2006-11-02 2019-02-18 Genentech Inc Humanized anti-factor D antibodies and their uses
CR20170001A (en) 2008-04-28 2017-08-10 Genentech Inc ANTI FACTOR D HUMANIZED ANTIBODIES
DE102008045127A1 (en) 2008-09-01 2010-03-04 Heinrich, Hans-Werner, Prof. Dr. Filter system for extracorporeal depletion of activated polymorphonuclear granulocytes (polymorphonuclear leukocytes PMNs)
US8758287B2 (en) 2008-11-12 2014-06-24 Marv Enterprises, LLC Utilization of stents for the treatment of blood borne carcinomas
WO2010107789A1 (en) 2009-03-17 2010-09-23 Marv Enterprises Llc Sequential extracorporeal treatment of bodily fluids
US9420770B2 (en) 2009-12-01 2016-08-23 Indiana University Research & Technology Corporation Methods of modulating thrombocytopenia and modified transgenic pigs
KR102336895B1 (en) 2012-02-20 2021-12-08 스위디쉬 오르펀 바이오비트럼 에이비 (피유비엘) Polypeptides binding to human complement c5
US20150027950A1 (en) * 2012-03-27 2015-01-29 Marv Enterprises, LLC Treatment for atherosclerosis
WO2013177099A1 (en) * 2012-05-21 2013-11-28 Felder Mitchell S Method for treating infectious diseases using emissive energy
US9132171B2 (en) 2012-05-23 2015-09-15 Wisconsin Alumni Research Foundation Test of insulin as a drug to reduce restenosis of vessels
KR20180021234A (en) 2013-08-12 2018-02-28 제넨테크, 인크. Compositions and method for treating complement-associated conditions
WO2015168468A1 (en) 2014-05-01 2015-11-05 Genentech, Inc. Anti-factor d antibody variants and uses thereof
EP3302529A4 (en) 2015-06-03 2019-02-06 The Children's Hospital Medical Center Compositions and methods for treating neonatal biliary atresia
EP3368090A1 (en) 2015-10-30 2018-09-05 H. Hoffnabb-La Roche Ag Anti-factor d antibody variant conjugates and uses thereof
JP2018534930A (en) 2015-10-30 2018-11-29 ジェネンテック, インコーポレイテッド Anti-factor D antibodies and conjugates
JP7102353B2 (en) 2016-06-14 2022-07-19 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Anti-C5 antibodies and their use
MA51147A (en) 2017-12-13 2021-03-24 Regeneron Pharma ANTI-C5 ANTIBODY ASSOCIATIONS AND ASSOCIATED USES
US20190247560A1 (en) * 2018-02-13 2019-08-15 Gambro Lundia Ab Extracorporeal devices and methods of treating complement factor related diseases
WO2021063708A1 (en) 2019-09-30 2021-04-08 Hemotune Ag Assembly for extracorporeal treatment of body fluids

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5135916A (en) * 1989-06-12 1992-08-04 Oklahoma Medical Research Foundation Inhibition of complement mediated inflammatory response

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4686100A (en) * 1985-04-02 1987-08-11 The Board Of Trustees Of The Leland Stanford Junior University Method for the treatment of adult respiratory distress syndrome
US5366986A (en) * 1988-04-15 1994-11-22 T Cell Sciences, Inc. Compounds which inhibit complement and/or suppress immune activity
US5506247A (en) * 1988-04-15 1996-04-09 T Cell Sciences, Inc. Compounds that inhibit complement and/or suppress immune activity
SE9100142L (en) * 1991-01-17 1992-07-18 Bengt Sandberg A METHOD AND A SYSTEM FOR PREPARING VIVO REDUCTION OF DIAGNOSTIC AND / OR THERAPEUTIC SUBSTANCES BY EXTRACORAL REMOVAL AND THE USE OF THESE SUBSTANCES FOR THIS PURPOSE

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5135916A (en) * 1989-06-12 1992-08-04 Oklahoma Medical Research Foundation Inhibition of complement mediated inflammatory response

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
COMPLEMENT. INFLAMM., Volume 8, issued 1991, R. WURZNER et al., "Inhibition of Terminal Complement Complex Formation and Cell Lysis by Monoclonal Antibodies", pages 328-340. *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0777474A1 (en) * 1994-09-23 1997-06-11 Alexion Pharmaceuticals, Inc. Methods for the treatment of inflammatory joint disease
EP0777474A4 (en) * 1994-09-23 2000-07-26 Alexion Pharma Inc Methods for the treatment of inflammatory joint disease
US7279158B2 (en) 1994-09-23 2007-10-09 Alexion Pharmaceuticals, Inc. Methods for the treatment of inflammatory joint disease
WO1999010009A1 (en) * 1997-08-26 1999-03-04 Gliatech Inc. A process for inhibiting complement activation via the alternative pathway
US6333034B1 (en) 1997-08-26 2001-12-25 Gliatech, Inc. Process for inhibiting complement activation via the alternative pathway
WO2003061765A1 (en) * 2002-01-22 2003-07-31 Alexion Pharmaceuticals, Inc. Method of prophylaxis against large myocardial infarctions
US7361339B2 (en) 2003-01-09 2008-04-22 Alexion Pharmaceuticals, Inc. Methods for reducing morality associated with acute myocardial infarction
EP2886140A1 (en) * 2013-12-17 2015-06-24 University of Limerick An apparatus for the extracorporeal treatment of blood
EP2886142A1 (en) * 2013-12-17 2015-06-24 University of Limerick An apparatus for the extracorporeal treatment of blood
US9422541B2 (en) 2013-12-17 2016-08-23 University Of Limerick Apparatus for the extracorporeal treatment of blood

Also Published As

Publication number Publication date
DE69533921D1 (en) 2005-02-17
US5853722A (en) 1998-12-29
EP0751787B1 (en) 2005-01-12
EP0751787A4 (en) 1999-06-16
ES2236706T3 (en) 2005-07-16
DE69533921T2 (en) 2005-12-01
AU2191795A (en) 1995-10-09
CA2186108A1 (en) 1995-09-28
EP0751787A1 (en) 1997-01-08

Similar Documents

Publication Publication Date Title
US5853722A (en) Use of C5-specific antibodies for reducing immune and hemostatic dysfunctions during extracorporeal circulation
Kroshus et al. Complement inhibition with an anti-C5 monoclonal antibody prevents acute cardiac tissue injury in an ex vivo model of pig-to-human xenotransplantation.
US6333034B1 (en) Process for inhibiting complement activation via the alternative pathway
Andonegui et al. Platelets express functional Toll-like receptor-4
Herzig et al. Granulocyte collection by continuous-flow filtration leukapheresis
Fung et al. Inhibition of complement, neutrophil, and platelet activation by an anti-factor D monoclonal antibody in simulated cardiopulmonary bypass circuits
JP2017057234A (en) Method of treating hemolytic disease
Weerwind et al. Influence of Duraflo II heparin-treated extracorporeal circuits on the systemic inflammatory response in patients having coronary bypass
US20170166640A1 (en) Cd89 activation in therapy
CA2134966C (en) Antibodies with specificity for multiple adhesion molecules
Gawaz et al. Changes in platelet membrane glycoproteins and platelet-leukocyte interaction during hemodialysis
Salvatierra et al. C1-esterase inhibitor prevents early pulmonary dysfunction after lung transplantation in the dog.
US5567443A (en) Method of treating inflammatory diseases
Gorlin Therapeutic plasma exchange and cytapheresis in pediatric patients
Bengtsson et al. Extracorporeal (“ex vivo”) connection of pig kidneys to humans. III. Studies of plasma complement activation and complement deposition in the kidney tissue
Johnell et al. Cell adhesion and tissue factor upregulation in oxygenators used during coronary artery bypass grafting are modified by the Corline Heparin Surface
Apter et al. An approach to immunologic reactions associated with plasma exchange
Kalmin et al. Pheresis with the IBM 2997
Lim et al. Clinical analysis of single filtration plasmapheresis using continuous renal replacement therapy machines in kidney transplantation
Gurewich et al. The fate of soluble fibrin monomer in relation to intravascular fibrin formation and degradation in rabbits
Baumgartner-Parzer et al. Possible clinical effects of the interaction of hemodialysis membranes with adhesion proteins
Heideman et al. The role of complement
Watkins The role of replacement fluids in plasma exchange and therapeutic applications of apheresis
AU2006202394A1 (en) A process for inhibiting complement activation via the alternative pathway
HEIDEMAN¹ et al. The Role of Complement

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1995914820

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2186108

Country of ref document: CA

WWP Wipo information: published in national office

Ref document number: 1995914820

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1995914820

Country of ref document: EP