HUE035692T2 - Use of C1 inhibitor for the prevention of ischemia-reperfusion injury - Google Patents

Use of C1 inhibitor for the prevention of ischemia-reperfusion injury Download PDF

Info

Publication number
HUE035692T2
HUE035692T2 HUE11172880A HUE11172880A HUE035692T2 HU E035692 T2 HUE035692 T2 HU E035692T2 HU E11172880 A HUE11172880 A HU E11172880A HU E11172880 A HUE11172880 A HU E11172880A HU E035692 T2 HUE035692 T2 HU E035692T2
Authority
HU
Hungary
Prior art keywords
inh
ischemia
rhc1
inhibitor
ischemic
Prior art date
Application number
HUE11172880A
Other languages
Hungarian (hu)
Inventor
Maurice Mannesse
Johannes Henricus Nuijens
Frank Pieper
Simoni Maria Grazia De
Gijsbertus Johannes Ziere
Original Assignee
Pharming Intellectual Property B V
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 Pharming Intellectual Property B V filed Critical Pharming Intellectual Property B V
Publication of HUE035692T2 publication Critical patent/HUE035692T2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • A61K38/57Protease inhibitors from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Description

• AKITA NOBUHISA ETAL: "PROTECTIVE EFFECT Remarks: OF C1 ESTERASE INHIBITOR ON REPERFUSION Thefile contains technical information submitted after INJURY IN THE RAT MIDDLE CEREBRAL the application was filed and not included in this ARTERY OCCLUSION MODEL", specification NEUROSURGERY, WILLIAMS & WILKINS, BALTIMORE, MD, US, vol. 52, no. 2,1 February 2003 (2003-02-01), pages 395-401, XP009083810, ISSN: 0148-396X, DOI: 10.1227/01. NEU.0000043710.61233.B4
Description
Field of the invention [0001] The present invention relates to the therapeutic and prophylactic use of C1 inhibitor for preventing, reducing and treating ischemia-reperfusion injury.
Background of the invention [0002] Ischemia-reperfusion injury is a well-known occurring pathologic condition. It may either represent a foreseen pathologic condition or an unforeseen pathologic condition. Stroke is one of the most common types of unforeseen ischemia-reperfusion injury. Stroke is the third cause of death and the leading cause of long-term disability in industrialized countries. Stroke is a type of cardiovascular disease that affects the arteries leading to and within the brain. A stroke occurs when such arteries are blocked by a clot or bursts and results in ischemia of the cerebral tissues that are served by the blocked artery. Direct damage to the brain is caused by the interruption of the blood flow, mainly due to loss of oxygenation to the viable tissue, ultimately leading to infarction if not reversed. However if the insult is reversed (either physiologically or therapeutically) then reperfusion of the ischemic tissue may paradoxically cause further indirect damage. When there is a long duration of ischemia, the "direct" damage resulting from hypoxia alone is the predominant mechanism. For shorter duration’s of ischemia, the indirect or reperfusion mediated damage becomes increasingly more important to the final outcome.
[0003] C1 inhibitor (C1INH), the inhibitor of complement C1, has been reported to display neuro-protective action by reducing ischemia-reperfusion injury in rodent models for cerebral ischemia-reperfusion. (De Simoni et al., 2003, J Cereb Blood Flow Metab. 23: 232-9; Akita et al., 2003, Neurosurgery 52: 395-400). The neuro-protective action of C1INH on brain ischemia-reperfusion injury does not require C1q (De Simoni et al., 2004, Am J Pathol. 164:1857-63). More recently Storini et al. (2005, Neurobiol Dis. 19: 10-7) reported that C1INH exerts an anti-inflammatory and anti-apoptotic action on ischemia-reperfusion injury through inhibition of inflammation and cell recruitment from the vasculature to the ischemic site. However, the window in time around the stroke during which administration of C1 INH is therapeutically effective is rather narrow. WO 2004/100982 relates to the manufacture of a recombinant C1 inhibitor prepared from the milk of transgenic rabbits; it has not explored the specific properties of the C1 inhibitor and has neither explored the time frame of administration of the C1 inhibitor. It is therefore an object of the present invention to provide for C1 INH with a broader window in terms of time of administration.
Description of the invention [0004] The present invention is based on the surprising finding that where naturally occurring plasma derived C1 inhibitor (C1 INH), has lost most of its ability to reduce ischemia reperfusion injury in a mouse model for transient cerebral focal ischemia when administered after ischemia, a recombinant preparation of C1 INH is still able to exert its neuropro-tective effects also when injected at least 1 hour after ischemia and/or reperfusion. Surprisingly, a neuroprotective effect can still be reached when the C1 INH is injected 18 hours after ischemia and/or reperfusion. The difference between the naturally occurring plasma derived C1 INH and the recombinant preparation of C1 INH is that the first has a plasma half life of at least 24 hours and is fully sialylated glycoprotein, and the latter has a reduced plasma half life and has a different glycosylation as compared to the plasma derived product.
[0005] A difference known between the naturally occurring plasma derived C1INH and the recombinant preparation of C1 INH is the extent and type of glycosylation. The recombinant glycoprotein contains a broad array of different N-glycans, comprising oligomannose-, hybrid-, and complex-type structures, whereas the N-glycans of plasma derived C1 INH are mainly composed of fully sialylated complex-type structures. As a result of the differences in glycosylation, the plasma derived glycoprotein has a plasma half life of at least 24 hours and the recombinant C1 INH has a reduced plasma half life.
[0006] Disclosed is a method for the prevention, reduction or treatment of at least one of ischemia and reperfusion injury, whereby the C1 inhibitor is administered after the ischemia and/or after the reperfusion. The method preferably comprises the step of administering an effective amount of a C1 INH having a plasma half life that less than the plasma half life of a plasma derived C1 INH. Alternatively, the method preferably comprises the step of administering an effective amount of a C1 INH that has a different glycosylation as compared to the plasma derived C1 INH. This method relates to a therapeutic and/or prophylactic use of C1 inhibitor for preventing, reducing and/or treating any type of ischemia-reperfusion injury.
[0007] A C1 inhibitor, also referred to as C1 esterase inhibitor is herein defined, as an inhibitor of complement C1. C1 INH belongs to the superfam ily of serine proteinase inhibitors and is the only inhibitor of C1 rand Clsofthe complement system and is the major inhibitor of factor XI la and kallikrein of the contact system. In addition C1 INH also inhibits other serine proteases of the coagulation and fibrinolytic systems like factorXI, tissue type plasminogen activator and plasmin (Schapira et al. 1985, Complement 2: 111 ; Davis, 1988, Ann. Rev. Immunol. 6: 595). Human C1INH is a protein of 500 amino acids, including a 22 amino acid signal sequence (Carter et al. 1988, Euro. J. Biochem. 173; 163). Plasma C1 INH is a glycoprotein of approximately 76 kDa and is heavily glycosylated, up to 26% of its molecular mass consists of carbohydrate (Perkins et al., 1990, J. Mol. Biol. 214 , 751). A C1INH for use in the methods of the present invention preferably is a protein with an amino acid sequence that has at least 65, 67, 68, 69, 70, 75, 80, 85, 90, 95, 98 or 99% identity with the amino acid sequence of the mature human C1 INH as depicted in SEQ ID NO:1.
[0008] For the purpose of the present invention, the degree of identity between two amino acid sequences refers to the percentage of amino acids that are identical between the two sequences. First, homologous polypeptide sequences are searched using the Basic Local Alignment Search Tool (BLAST) algorithm, which is described in Altschul, et al., J. Mol. Biol. 215:403-410 (1990). Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). The BLAST algorithm parameters W, B, and E determine the sensitivity and speed of the alignment. The BLAST program uses as defaults a word length (W) of 3, the BLOSUM62 scoring matrix (see Henikoff & Henikoff, Proc. Natl. Acad. Sei. USA 89: 10915 (1989)) alignments (B) of 50, expectation (E) of 10, M=5, N=-4. Next, the degree of identity (as defined above) of homologous sequences is determined using the CLUSTALW alignment algorithm (Higgins D. et al (1994). Nucleic Acids Res. 22:4673-4680) using the following parameters; Gap size: 5, Gap open: 11, Gap extension: 1, Mismatch: -15, Word size: 3.
[0009] The C1INH preferably has C1INH activity as may e.g. be assayed as described by Drouet et al. (1988, Clin Chim Acta. 174:121-30). More preferably, the C1INH is a human C1INH (hCIINH) which is understood to mean that the C1 INH has an amino acid sequence that naturally occurs in man (as e.g. SEQ ID NO:1 or CAA30314) but does not mean that the C1 INH is produced in and obtained from e.g. human plasma.
[0010] According to one aspect of the invention the C1 INH for use in the methods of the invention preferably has a reduced plasma half life as compared to the plasma half life of plasma derived C1 INH, more preferably the plasma half life of the C1 INH of the invention is less than the plasma half life of C1 INH derived from human plasma. By "a reduced plasma half life" is meant the negative change in circulating half life of a C1 INH of the invention relative to the circulating half life of a plasma derived C1 INH. In this context, a plasma derived C1 INH refers to naturally occurring C1 INH which is typically derived from plasma and which may be purified from plasma but is not modified in chemically or enzymatically. [0011] Plasma half life is measured by taking blood samples at various time points after administration of the C1 INH, and determining the concentration of the C1 INH in each sample. Correlation of the serum concentration with time allows calculation of the plasma half life. The reduction of plasma half life of a C1 INH of the invention relative to the circulating half life of a plasma derived C1 INH preferably is at least about twofold, at least about three-fold, at least about four-fold, at least about six-fold, more preferably at least about eight-fold, and most preferably at least about ten-fold. In other words, plasma half life of a C1 INH of the invention preferably is less than 60, 50, 40, 30, 25, 20, 15, 12.5 or 10% of the plasma half life of a plasma derived C1INH, i.e. its naturally occurring counterpart.
[0012] E.g. the plasma half life of the C1 INH of the invention that is used in the Examples herein, which is obtained from the milk of transgenic rabbits, exhibits a plasma half life in humans of about 3 hours, which about four- to eightfold less than the average plasma half life of a plasma derived C1 INH in man. It is understood that the determination of the reduction of plasma half life of a C1 INH of the invention as compared to that of plasma derived C1 INH is preferably performed under similar if not identical conditions, i.e. preferably at corresponding dosages, sampling regimes, in the same organism, which may be a laboratory animal such as a mouse or human subjects, and in about the same number of test subjects. Furthermore, it is understood that the average plasma half lives of both C1 INH preparation are compared as may be determined by standard method of statistical analysis.
[0013] A C1 INH with shorter half life, be it a naturally occurring or a recombinantly produced C1 INH, may be prepared by any convenient method. It may for example be prepared in vivo in a recombinant host cell or organism that results in a C1INH with a modified carbohydrate structure (as compared to the plasma derived C1INH) or the carbohydrate structure of a naturally occurring C1 INH may be chemically or enzymatically modified in vitro. Preferably, the C1 INH of the invention is modified compared to the plasma derived C1 INH the following way: removal of a carbohydrate moiety (from a naturally occurring variant or recombinantly expressed variant of the glycoprotein), preferably the removal of sialic acid and/or galactose from a N-linked carbohydrate chain and/or the removal of a carbohydrate chain resulting in exposure of mannose, galactose, N-acetylglucosamine and/or fucose residues.
[0014] According to another aspect of the invention the C1 INH for use in the methods of the invention preferably has a different glycosylation as compared to the plasma derived C1INH. Modifications to the carbohydrate structure of a C1 INH of the invention include modifications which lead to underglycosylation, overglycosylation, to the asialio form of C1 INH, or any other modifications which lead to a different glycosylation pattern.
[0015] In vitro, underglycosylation may be the result of a deletion of a carbohydrate moiety or of a complete carbohydrate chain of C1 INH. Modifications may involve both N- or O-linked carbohydrate chains, or only one type of chain. It may involve all the chains, or only some of the chains. Overglycosylation may for instance be the result of the addition of an extra carbohydrate moiety or a complete carbohydrate chain to the C1 INH molecule. An asialo-form of C1 INH or a form having a reduced level of terminal sialic acid residues may typically be obtained by removal of a sialic acid group. It is well-known that the half life of a glycoprotein in the blood is highly dependent on the composition and structure of its N-and O-linked carbohydrate groups. In general, maximal half life of a glycoprotein requires that its N- and O-linked carbohydrate groups have a terminal sialic acid. If this terminal sialic acid is not present, the glycoprotein is rapidly cleared from the blood due to the exposure of galactose residues. It is well-established that the presence of terminal galactose residues in carbohydrate moieties of glycoproteins results in enhanced plasma clearance by the asialoglycoprotein receptor in the liver. Thus, C1INH for use in the methods of the present invention has a reduced level of terminal sialic acid residues as compared to plasma derived human C1 inhibitor. Sialic acid may be removed in several ways. For instance, it may be removed chemically or enzymatically, for example, by treatment with sialidase. Suitable sialidases for this purpose are described by Chou et al. (1996, J Biol Chem. 271 (32): 19219-24; and 1994, J Biol Chem. 269(29):18821-6) and may e.g. be obtained from V-labs, Inc. (Covington, Louisiana, USA). In a further preferred embodiment, C1 INH for use in the methods of the present invention preferably has exposed mannose, N-acetylglucosamine phosphomannose, galactose and/or N-acetylgalactosamine residues. An exposed sugar residue will usually be a terminal sugar residue on a glycan branch or at least a sugar residue that is accessible for interactions with a moiety having affinity forthe residue (such as a carbohydrate binding domain). AC1 INH with exposed galactose, N-acetylgalactosamine, N-acetylglucosamine, mannose, fucose or phosphomannose residues may e.g. be obtained by enzymatic treatment with one or more of ß-D-/V-acetylhexosaminidase, endo-ß-D-galactosidase, and/or a-D-/V-acetylgalactosaminidase (also obtainable form e.g. V-labs, Inc., Covington, Louisiana, USA).
[0016] In vivo, modifications of carbohydrate chains of C1INH may be introduced by using recombinant production systems. Both prokaryotic and eukaryotic cell cultures may be used, such as yeast cells, fungal cells, insect cells and mammalian cells. For example, COS cells and CHO cells are suitable mammalian production systems. Although mammalian cell culture systems have the capacity to produce glycoproteins with sialylated carbohydrate groups, optimal, natural or complete glycosylation is often difficult to achieve and consequently, recombinantly produced glycoproteins in general have a different glycosylation pattern than their natural counterparts. Usually this different glycosylation pattern is incomplete (as compared to the natural counterparts) having exposed galactose, N-acetylglucosamine and/or mannose residues. Likewise, production of C1INH in eukaryotic microorganisms like yeasts or fungi will result in C1INH with exposed mannose residues.
[0017] C1INH with modified carbohydrate structures may also be prepared in transgenic animals, preferably in nonhuman animals, such as in transgenic rabbits, bovine, mice, rats, goats and sheep. Preferably, such glycoproteins are expressed in the mammary glands of these non-human transgenic animals such that the glycoproteins may be obtained from the milk of the animal. The skilled person will understand that it will depend on the specific glycoprotein to be produced and on the amount which has to be produced, which transgenic animal is best usedfor production. A particularly preferred C1 INH for use in the present invention is a C1 INH that is obtained from the milk of a transgenic bovine or an animal of the order Lagomorpha, preferably of the family Leporidae, more preferably of the genus Oryctolagus and most preferably a rabbit of the species Oryctolagus cuniculus.
[0018] Different types of modifications to the structure of the carbohydrate chain of the C1 INH protein as compared to its natural plasma-derived counterpart may be obtained from recombinant production systems, such as different glycosylation, underglycosylation or overglycosylation may be introduced separately or in combination, simultaneously or consecutively, some types may be introduced to one part of the molecule, while others are introduced to another part of the molecule. Preferred combinations of modifications contribute to the therapeutic efficacy of the protein include exposed galactose, N-acetylgalactosamine, N-acetylglucosamine, mannose, fucose and/or phosphomannose residues on the C1 INH of the invention. The C1 INH of the invention may e.g. have glycans of the oligomannose type or of the highmannose type. Preferably at least about 5, 10, 15, 20, 40 or 60% of the terminal residues on the glycans on the C1INH are selected from galactose, N-acetylgalactosamine, N-acetylglucoseamine, mannose, fucose and phosphomannose residues. E.g. a preferred C1 INH for use in the present invention contains about 2, 4, 5, 6-fold less sialic acid as compared to its natural counterpart and/or at least about 5, 10, 15, 20, 40 or 60% of its N-linked glycans are neutral carrying terminal hexoses with equatorial 3- and 4-OH groups, such as mannose and N-acetylglucosamine. In contrast, plasma derived C1 INH has no oligomannose type glycosylation. A preferred C1 INH for use in the present invention e.g. is a recombinant human C1INH produced in the mammary glands of rabbits which has 5-6 fold less sialic acid as compared to its natural counterpart and about 15% of its N-linked glycans are neutral carrying terminal mannose residues. [0019] In a preferred embodiment, the different glycosylation of the C1INH for use in the present invention results in a higher affinity for a mannose binding protein as compared to its plasma derived counterpart. The mannose binding protein (MBP) is also referred to as mannan-binding protein, mannose-binding lectin (MBL), mannan-binding lectin, or bactericidal Ra-reactive factor. MBP is a collectin that belongs to a group of soluble Ca2+-dependent (C-type) lectins. MBP is an activator of complement via the lectin pathway (that differs from the classical and alternative pathways of complement activation). The complement system is an important component of the innate immune defense and is activated by three pathways: the classical pathway, the alternative pathway, and the recently discovered lectin or Mannose binding lectin (MBL) pathway. The activation of the classical pathway begins when the catalytic domains C1r and C1s bind to immune complexes via the recognition protein C1q (see figure 11).
The alternative pathway is continuously turning over at a slow rate in an antibody-independent manner and will attack particles that are not specifically protected against complement.
The lectin or MBL pathway is initiated or activated upon binding of MBL to carbohydrate structures present on various pathogens or other cellular structures. Two serine proteases: mannan-binding lectin associated serine protease (MASP)-1 and -2 (see figure 11) are associated with MBL and show striking similarities with the serine proteases C1s and C1r. The complex has C4- and C3-activating capacities upon binding to mannan. The complex contains two serine proteases MASP-1 and MASP-2 linked by a disulfide bond. In this form, MASP is capable of cleaving C4 and C3 resulting in their activation. The C1INH of the invention preferably has a higher affinity for a human MBP as compared to its plasma derived counterpart.
[0020] MBP recognizes exposed hexoses with equatorial 3- and 4-OH groups, such as mannose and N-acetylglu-cosamine and/or N-acetyl-hexosamines. A preferred C1INH of the invention therefore carries such terminal hexoses. The higher affinity for MBP, preferably human MBP of the C1 INH of the invention preferably is such that it allows a more efficient targeting, binding and/or inhibition of MBP as compared to its natural plasma derived counterpart that lacks exposed mannose and N-acetylglucosamine residues. Human MBP is herein understood to refer to the protein characterized by Kawasaki et al. (1983, J. Biochem 94:937-47), having an amino acid sequence as described by Taylor et al. (1989, Biochem. J. 262 (3), 763-771; NCBI accession no. CAA34079). The structure of rat MBP complexed with an oligosaccharide is described by Weis et al. (1992, Nature. 360:127-34). For a further description of human MBP see e.g US 6,846,649 and references cited therein.
[0021] All of these pathways (classical, alternative and lectin or MBL) generate a crucial enzymatic activity that eventually leads to the assembly of the membrane attack complex (MAC or C5b-C9) (see Figure 11). Under physiological conditions, activation of the complement system is effectively controlled by the coordinated action of soluble and membrane-associated regulatory proteins. One of these proteins is C 1 inhibitor (C1INH), a serine protease inhibitor that binds to C1s and C1r and currently the only known physiological inhibitor of the classical pathway. In addition, C1INH is able to inactivate MBL-mediated complement activation by binding and inhibiting MASP-1 and MASP-2.
[0022] The activation of the different complement pathways is preferably measured in human sera by the Wielisa kit (product no. COMPL 300, Wieslab, Sweeden). This is a commercially available enzyme immuno assay, specific for the detection of each of the three complement pathways with deposition of C5b-C9 as a common read-out. Briefly, wells of microtitre strips are coated with specific activators of each of the three complement pathways. Human serum is diluted in diluent containing specific blocker to ensure that only the respective pathway is activated. C1 INH of the invention or its plasma-derived counterpart is further added in a concentration ranged between 0 and 75 μίτιοΙ, incubated for 30 minutes at room temperature and added to the wells. During a subsequent incubation of the diluted human serum in the well for 60 minutes at 37°c, complement is activated by the specific coating. The wells are then washed and C5b-C9 formed is detected with a specific alkaline phosphatase labelled anti C5b-C9 antibody. After a further washing step, detection of specific antibodies is obtained by incubation with alkaline phosphatase substrate solution. The amount of complement activation correlates with the colour intensity and is measured in terms of absorbance (optical density OD). Using this kit, both recombinant human C1 INH (rhC1 INH) of the invention and plasma-derived C1 INH (pdC1 INH) were found to have similar inhibiting capacities for the classical pathway. However, the CIINH of the invention was found to have approximatively 20% more inhibiting capacitiy for the MBL pathway than plasma-derived C1INH (see example 3). Therefore accordingly, in this preferred embodiment, the different glycosylation of the C1INH for use in the present invention results in a higher affinity for a MBP as compared to its plasma derived counterpart, which results in a more efficient inhibition of MBP, leading to a more efficient inhibition of the lectin pathway. More efficient inhibition of the lectin pathway preferably means at least 5% more inhibition, even more preferably at least 10% more inhibition, even more preferably at least 15% more inhibition even more preferably at least 20% even more preferably at least 25% even more preferably at least 30% even more preferably at least 35% even more preferably at least 40% even more preferably at least 45% even more preferably at least 50% even more preferably at least 55% even more preferably at least 60% even more preferably at least 65% even more preferably at least 70% even more preferably at least 75% even more preferably at least 80% even more preferably at least 85% even more preferably at least 90% even more preferably at least 95% and most preferably at least 98% more inhibition. The activation of the lectin pathway is preferably measured by the Wielisa kit as described above.
[0023] The method of the invention may be applied to prevent, reduce or treat any type of ischemia and reperfusion injury. Preferably, the method of the invention is applied wherein the ischemia and reperfusion injury is known to arise at least in part, more preferably mostly via the lectin pathway. For myocardial ischemia and reperfusion injury (J Immunology 2005, 175: 541-546), renal ischemia-reperfusion injury (Am J Pathol. 2004 165(5):1677-88), gastrointestinal ischemia reperfusion injury (J Immunol. 2005 15:174(10):6373-80), and for stroke (deSimoni et al, 2004 Am J. Pathol. 164:1857-63) it has been shown that reperfusion injury arises mostly via the lectin pathway and hardly via the classical pathway. Hence, a C1INH of the invention preferably is a more potent inhibitor of the lectin pathway as compared to its natural plasma derived counterpart. Preferably a C1 INH of the invention is a more potent in vivo inhibitor of the lectin pathway in man as compared to its natural plasma derived counterpart.
Unlike the experimental model used in the Examples herein, the occurrence of ischemia in real life often is an unforeseen event. Therefore administration of C1INH prior to the occurrence of ischemia and/or subsequent reperfusion is not generally a feasible option and inevitably in practice C1 INH will have to be administered some time if not several hours after ischemia and/or subsequent reperfusion. This, however, seriously limits the therapeutic usefulness of conventional plasma derived C1 INH because it is mostly ineffective when administered subsequent to ischemic reperfusion and only has a very small time window for therapeutic efficacy (see Figure 2 and deSimoni et al, 2004 Am J. Pathol. 164:1857-63). In contrast, a C1INH for use in the present invention as defined above, is still able to exert its neuroprotective effects also when injected at least 1 hour after ischemia or after the onset of ischemia and/or 30 minutes after the start of the reperfusion. Therefore, in a disclosure of the method for the prevention, reductionortreatmentofat least one of unforeseen or foreseen occurrence of ischemia and reperfusion injury, the C1 INH is administered at least at the end or after the ischemic period, i.e. when the ischemic tissue is reperfused. More preferably, the C1INH is administered at least 10, 15, 20, 30, 45, 60, 90 or 120 minutes after the ischemic period or after the start of reperfusion. Preferably, the C1 INH is administered no more than 24, 12, 6, 4 or 3 hours after ischemia or after the onset of ischemia and/or reperfusion. In a disclosure, the C1 inhibitor is administered at least 3 hours after ischemia or after the onset of ischemia and/or reperfusion, preferably at least 6 hours, more preferably at least 9 hours, even more preferably at least 18 hours. [0024] In a disclosure, the method is applied to prevent, reduce or treat an unforeseen, sudden or acute occurrence of ischemic reperfusion. Conditions and disorders associated with an unforeseen, sudden or acute occurrence of ischemic reperfusion injury include but are not limited to ischemic reperfusion injury after acute myocardial infarction (AMI), after stroke, including perinatal stroke, after hemorrhagic shock, after intestinal ischemia, after emergency coronary surgery for failed percutaneous transluminal coronary angioplasty (PCTA), after any vascular surgery with blood vessel cross clamping (e.g. of aorta, leading to skeletal muscle ischemia), or after pancreatitis after manipulation of pancreatic or bile duct (ERCP). In such instances the C1INH preferably is administered at least 1, 5, 10, 15, 20, 30, 45, 60, 90 or 120 minutes after the acute myocardial infarction (AMI), after stroke, including perinatal stroke, after hemorrhagic shock, after intestinal ischemia, after emergency coronary surgery for failed percutaneous transluminal coronary angioplasty (PCTA), after any vascular surgery with blood vessel cross clamping (e.g. of aorta, leading to skeletal muscle ischemia), or after pancreatitis after manipulation of pancreatic or bile duct (ERCP). Alternatively, the time of administering the C1 INH may be defined as preferably at least 1,5, 10, 15, 20, 30, 45, 60, 90 or 120 minutes after the start of reperfusion. In addition, unforeseen ischemic reperfusion injury is preferably defined as an ischemic reperfusion injury wherein a therapy or surgery induces a reperfusion but not an ischemia. Such therapy or surgery include but not limited to: pharmacological thrombolysis, including intravenous and endovascular therapies for stroke, acute coronary syndromes, peripheral arterial occlusion, pulmonary embolus, renal artery occlusion, mechanical thrombolysis, e.g. percutaneous coronary intervention, peripheral arterial angioplasty, visceral arterial angioplasty, coronary artery bypass grafting, carotid endarterectomy, mesenteric ischemia, shock including hemorrhagic, cardiogenic, neurogenic, analphylactic, flap-failure, e.g. plastic surgery, re-implantation of digits and limbs, strangulated bowel.
[0025] According to the invention, the method is applied to prevent, reduce or treat a foreseen occurrence of ischemic reperfusion. A foreseen occurrence of ischemia reperfusion injury preferably includes a setting in which a therapy or surgery induce both an ischemia and subsequently a reperfusion. A non-limiting list is given below of therapy or surgery in which there is an induced temporary period of no or low blood flow, i.e. ischemia or hypoxia, followed by reperfusion: cardiopulmonary bypass, aneurysm repair, including aortic, cererbral, carotid endarterectomy in which a clamp is used during the surgery, deep hypothermic circulatory arrest, tourniquet use, i.e. in trauma settings, solid organ transplantation, any other iatrogenic disruption of blood flow.
In addition, conditions and disorders associated with a foreseen occurrence of ischemic reperfusion injury include but are not limited to ischemic reperfusion injury after organ transplantation (lung, liver, kidney, heart), after any vascular surgery with blood vessel cross clamping (e.g. of aorta, leading to skeletal muscle ischemia), or after pancreatitis after manipulation of pancreatic or bile duct (ERCP), after or during extra corporal circulation (ECC).
In a disclosure, reduction or treatment of at least one of foreseen occurrence of ischemia and reperfusion injury, the C1 INH of the invention is administered at least at the end or after the ischemic period, i.e. when the ischemic tissue is reperfused. In a disclosure, the C1 INH is administered at least 10,15,20, 30, 45, 60, 90 or 120 minutes after the ischemic period or after the start of reperfusion. Preferably, the C1 INH is administered no more than 24, 12, 6, 4 or 3 hours after ischemia or after the onset of ischemia and/or reperfusion. In a disclosure, the C1 inhibitor is administered at least one hour after ischemia or after the onset of ischemia and/or reperfusion, 3 hours after ischemia or after the onset of ischemia and/or reperfusion, preferably at least 6 hours, more preferably at least 9 hours, even more preferably at least 18 hours. [0026] A method is provided for the prevention, reduction or treatment of at least one of foreseen occurrence of ischemia and reperfusion injury, wherein the C1INH of the invention is administered before or during the ischemia and reperfusion. The skilled person will understand that depending upon the plasma half life of the C1 INH of the invention, the earliest possible time point, wherein the C1INH of the invention may be administered may be adjusted to obtain the best possible result.
According to one preferred embodiment, the C1INH of the invention is continuously administered to a subject in the need thereof and/or in case of an organ transplantation to the organ to be transplanted. The organ to be transplanted is preferably conserved in a composition with a suitable medium and suitable amount of C1 INH.
According to the invention, before the occurrence of a foreseen type of ischemia and reperfusion injury means that the administration is performed at the most 3 hours before at least one foreseen occurrence of ischemia and reperfusion injury, preferably at the most 2 hours, more preferably at the most one hour, and most preferably at the most 30 minutes. A subject in the need thereof is a subject wherein a foreseen occurrence of ischemia and reperfusion injuries may occur. Foreseen occurrence of ischemia and reperfusion injuries have been already described herein.
The administration of the C1 INH before the foreseen occurrence of ischemia and reperfusion injury is attractive since it may prevent the occurrence of most if not all damages associated with the ischemia and reperfusion injury the same way as presented when the C1 INH is administered after the occurrence of ischemia and reperfusion injury, if not better. [0027] In a disclosure, the method is applied to an unforeseen occurrence of ischemic reperfusion, such as, an ischemic reperfusion injury occurring after a stroke or a perinatal stroke. In these types of unforeseen occurrence of ischemic reperfusion, we demonstrated that the C1 inhibitor of the invention exerts a neuroprotective effect in the ischemic penumbra. The ischemic penumbra preferably means the hippocampus and/or cortex. A neuroprotective effect preferably means that neurodegeneration is counteracted in the hippocampus and/or cortex after treatment with the C1 inhibitor of the invention up to 3 hours after the onset of ischemia in the hippocampus and up to 9 hours after the onset of ischemia in the cortex. More preferably, neurodegeneration is counteracted up to 4, 5, 6 hours or more in the hippocampus and up to 10,11,12 hours or more in the ischemia. Neurodegeneration is preferably assessed as in example 2: brain sections are stained with a marker specific for neuronal degeneration, preferably Jade (Schmued LC, et al, reference 4) and analyzed by fluroescent microscopy. Using this method, counteraction of neurodegeneration means at least 2% less stained cells are visualized in the treated sample compared to the untreated sample. Preferably, counteraction of neurodegeneration means at least 5% less stained cells, at least 7%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40% or more.
[0028] In a disclosure, the use of the C1 inhibitor exerts a reduction of the lesion induced by the ischemia and/or reperfusion. More preferably, when the ischemic reperfusion injury occurred after a stroke or a perinatal stroke, the use of the C1 inhibitor of the invention exerts a reduction of the infarct size. Even more preferably, the infarct size is quantified as presented in example 2. Even more preferably, using this quantification method, at least 3 hours after the onset of ischemia, a reduction of at least 10% of the infarct size is reached, even more preferably at least 20%, even more preferably at least 40%, even more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%,and most preferably at least 90%.
[0029] A C1 INH for use in the methods of the invention may be part of or combined with state of the art pharmaceutical compositions. These pharmaceutical compositions typically comprise the C1INH and a pharmaceutically acceptable carrier or excipient. These pharmaceutical compositions may be administered in a number of ways depending on whether local or systemic treatment is desired, the area to be treated and the stability of the active compound. Suitable formulations will depend on the method of administration. The pharmaceutical composition is preferably administered by parenteral administration, such as for example by intravenous, intra-arterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or by intrathecal or intracranial administration. In a preferred embodiment it is administered by intravenous infusion. Suitable formulations for parenteral administration are known in the art and are typically liquid formulations. C1 INH preparations for parental administration must be sterile. Sterilization is readily accomplished by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution. C1 INH preparations may be administered continuously by infusion or by bolus injection. Liquid C1 INH formulations may for example be administered by an infusion pump. Atypical composition for intravenous infusion could be made up to contain 100 to 500 ml of sterile 0.9% NaCI or 5% glucose optionally supplemented with a 20% albumin solution and 100 to 500 mg of the C1 INH. A typical pharmaceutical composition for intramuscular injection would be made up to contain, for example, 1 - 10 ml of sterile buffered water and 1 to 250 mg of the C1 INH of the present invention. Methods for preparing parenterally administrable compositions are well known in the art and described in more detail in various sources, including, for exam pie, Remington’s Pharmaceutical Science (15th ed., Mack Publishing, Easton, PA, 1980).
[0030] The effective dose, i.e. effective concentration and frequency, of the C1INH when used in the methods of the invention will depend on the specific pharmaceutical composition which is used, the severity of the condition and the general state of the patient’s health. In general, the effective dose of a pharmaceutical composition which is based on a C1INH for use in the methods of the invention may be found by routine optimisation. A suitable starting point is the dose which is used for the equivalent pharmaceutical composition which is based on plasma-derived C1INH. A great advantage of a pharmaceutical composition of the invention is that a high initial dose may be used in treatment, which enhances the likelihood of successful treatment. This high initial dose is possible because the C1 INH in the pharmaceutical composition of the invention shows a faster clearance than its natural counterpart. In particular for the treatment of acute cases, a high initial dose of the C1 INH of the invention may be advantageous. This high initial dose may be at least 1.5, at least 2, 3 or 4 times the dose of the natural occurring counterpart which would be administered.
[0031] In a preferred embodiment, C1INH of the invention is administered intravenously at a dose of more than 50, 100, 200, 400, 600, 800, or 1000 U /kg body weight of the individual, preferably in the range of 50 - 2000, 100 -1000, 200 - 800, 400-700 or 500-700 U/kg body weight of the individual. One unit (U) of C1 INH is the amount of C1 INH present in 1 millilitre of human blood. One such unit corresponds to approximately 275 microgram plasma derived C1INH. Assuming a molecular weight of 110,000 dalton, the concentration in human plasma of C1 INH is 2.5 micromol per litre (Nuijens et al. (1989), J. Clin. Invest. 84:443).
[0032] In a further preferred embodiment of the method of the invention the pharmaceutical composition further contains a thrombolytic agent or is for use in combination with a thrombolytic agent or aftersubsequent treatment with such agent. A thrombolytic agent is herein understood to mean an agent (drug) that is able to dissolve a blood clot (thrombus) and reopen an artery orvéin. Thrombolytic agents are usually serine proteases and convert plasminogen to plasmin which breaks down the fibrinogen and fibrin and dissolves the clot. Preferred thrombolyic agents include reteplase (r-PA or Retavase), alteplase (t-PA or Activase), urokinase (Abbokinase), prourokinase, anisoylated purified streptokinase activator complex (APSAC), and streptokinase.
[0033] In a further aspect, particularly for jurisdictions other than the USA, the invention pertains to the use of a C1 INH of the invention as defined herein above for the manufacture of a medicament for the prevention, reduction or treatment of reperfusion injury in accordance with any of the methods defined herein above.
[0034] In this document and in its claims, the verb "to comprise" and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition, reference to an element by the indefinite article "a" or "an" does not exclude the possibility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements. The indefinite article "a" or "an" thus usually means "at least one".
Disclosures [0035] 1. Use of a C1 inhibitor having a plasma half life of less than 6 hours in the preparation of a pharmaceutical composition for the prevention, reduction or treatment of at least one of ischemia and reperfusion injury, whereby the C1 inhibitor is administered after the ischemia and reperfusion. 2. The use according to embodiment 1, wherein the C1 inhibitor has a reduced level of terminal sialic acid residues as compared to plasma derived human C1 inhibitor. 3. The use according to embodiment 1 or 2, wherein the C1 inhibitor comprises a glycan that has a terminal residue selected from galactose, N-acetylgalactosamine, N-acetylglucosamine, mannose and fucose. 4. The use according to any one of embodiments 1 to 3, wherein the C1 inhibitor has an amino acid sequence that has at least 65% identity with the amino acid sequence of SEQ ID NO: 1. 5. The use according to any one of embodiments 1 to 4, wherein the C1 inhibitor is obtained from genetically engineered cell or organism. 6. The use according to embodiment 5, wherein the C1 inhibitor is obtained from a transgenic non-human animal. 7. The use according to embodiment 6, wherein the C1 inhibitor is obtained from the milk of a transgenic non-human animal. 8. The use according to embodiment 6 or 7, wherein the transgenic non-human animal is a bovine or an animal of the order Lagomorpha, preferably a rabbit. 9. The use according to any one of embodiments 1 to 8, wherein the C1 inhibitor is used in an amount in the range of 50 - 2000 units per kg body weight. 10. The use according to any one of embodiments 1 to 9, wherein the C1 inhibitor is administered at least one hour after ischemia and/or reperfusion, preferably at least 3 hours, more preferably at least 6 hours, more preferably at least 9 hours, even more preferably at least 18 hours. 11. The use according to anyone of embodiments 1 to 10, wherein the pharmaceutical composition also contains a thrombolytic agent or is for use in combination with a thrombolytic agent or after subsequent treatment with such agent. 12. The use according to any one of embodiments 1 to 11, wherein the pharmaceutical composition is for the prevention, reduction or treatment of at least one unforeseen sudden or acute occurrence of ischemia and reperfusion injury. 13. The use according to embodiment 12, wherein the pharmaceutical composition is for the prevention, reduction or treatment of at least one of ischemia and reperfusion injury after stroke or perinatal stroke. 14. The use according to any one of embodiments 1 to 11, wherein the pharmaceutical composition is for the prevention, reduction or treatment of at least one foreseen occurrence of ischemia and reperfusion injury, preferably after organ transplantation. 15. Use of a C1 inhibitor, said C1 inhibitor being as defined in any one of embodiments 1 to 9, in the preparation of a pharmaceutical composition for the prevention, reduction or treatment of at least one of ischemia and reperfusion injury, whereby the C1 inhibitor is administered before or during at least one foreseen occurrence of ischemia and reperfusion injury. 16. The use according to embodiment 15, wherein the C1 inhibitor is administered at the most 3 hours before at least one foreseen occurrence of ischemia and reperfusion injury, preferably at the most 2 hours, more preferably at the most one hour, and most preferably at the most 30 minutes and/or wherein the C1 inhibitor of the invention is continuously administered to a subject in the need thereof and/or in case of an organ transplantation to the organ to be transplanted. 17. The use according to embodiment 15 or 16, wherein the foreseen occurrence of ischemia and reperfusion injury is organ transplantation. 18. The use according to any one of embodiments 1 to 17, wherein the pharmaceutical composition is for the prevention, reduction or treatment of at least one of ischemia and reperfusion injury known to arise via the lectin pathway, preferably myocardial, renal, gastrointestinal ischemia and reperfusion injury or stroke. 19. The use according to embodiment 13, wherein the C1 inhibitor exerts a neuroprotective effect preferably in the hippocampus and/or cortex. 20. The use according to any one of embodiments 1 to 19, wherein the C1 inhibitor exerts a reduction of a lesion induced by the ischemia and/or reperfusion.
Description of the figures [0036]
Figure 1 Assessment of infarct size forty-eight hours after ischemia in mice treated with saline or with 15U rhC1 INH (recombinant human C1 INH, see Example 1.2) per mouse pre, post and 1 h post ischemia.
Figure 2 Assessment of infarct size twenty-four hours after ischemia in mice treated with saline or with 15U plasma derived hCIINH per mouse pre, post and 1h post ischemia.
Figure 3 Infarct volume assessed 48h after ischemia in mice receiving saline or 15U/mouse of rabbit rhC1-INH at different time points from the beginning of ischemia. Data are expressed as mean ± SEM (n=6 mice per group). *P<0.05, **P<0.01 versus saline, one way ANOVA and Dunnett as post-hoc test.
Figure 4 Semi-quantitative evaluation of Fluoro-Jade staining.- = no positivity, + = low positivity, ++ = intermediate positivity, +++ = high positivity.
Figure 5 Representative images of neurodegeneration by Fluoro-Jade staining in the striatum of ischemic mice receiving saline or 15U/mouse of rabbit rhC1-INH at different time points from the onset of ischemia. Bar: 100 pim.
Figure 6 Representative images of neurodegeneration by Fluoro-Jade staining in the dentate gyrus of ischemic mice receiving saline or 15U/mouse of rabbit rhC1 -INH at different time points from the onset of ischemia. Bar: ΙΟΟμίτι.
Figure 7 Representative images of neurodegeneration by Fluoro-Jade staining in the cortex of ischemic mice receiving saline or 15U/mouse of rabbit rhC1-INH at different time points from the onset of ischemia. Bar: 100 μη.
Figure 8 Infarct volume assessed 48h after ischemia in mice receiving saline or 5, 10, 15U/mouse of rabbit rhC1-INH 3 hours after the onset of ischemia. Data are expressed as mean ± SEM (n=6 mice per group). **P<0.01 versus saline, one way ANOVA and Dunnett as post-hoc test.
Figure 9 Infarct volume assessed 48h after ischemia in mice receiving saline or 15U/mouse of pdC1-INH or cow, or rabbit rhC1-INH three hours after the onset of ischemia. Data are expressed as mean ± SEM (n=6 mice per group). *P<0.05, **P<0.01 versus saline, one way ANOVA and Dunnett as post-hoc test.
Figure 10 General (upper pannel 10a) and focal (lower pannel 10b) deficits assessed 48h after ischemia in mice receiving saline or 15U/mouse of pdC1-INH or cow, or rabbit rhC1-INH three hours after the onset of ischemia. (n=6 mice per group). **P<0.01 versus saline, one way ANOVA and Kruskal-Wallis as post-hoc test.
Figure 11 Overview of the different pathways of complement activation.
Figures 12, 13 Effect of rhc1 INH and pdC1 INH on activation of the classical complement pathway. Increasing doses of rhCHNH or pdCIINH (x-axis) was added to two different samples of normal human serum (sample 1 upper pannel. sample 2 lower pannel). As a control, the buffer in which rhCHNH is dissolved (20 mM citrate, 0.19 M sucrose pH 6.8; 0.22 μίτι filtered) was taken along in the same dilutions as rhdlNH. Readout was deposition of C5b-9, the normal serum control in the assay defining 100% (y-axis). Data are mean and SD (n = 3).
Figures 14, 15 Effect of rhc1 INH and pdC1 INH on activation of the MBL complement pathway. Increasing doses of rhC1 INH or pdC1 INH (x-axis) was added to two different samples of normal human serum serum (sample 1 upper pannel. sample 2 lower pannel). As a control, the buffer in which rhdlNH is dissolved (20 mM citrate, 0.19 M sucrose pH 6.8; 0.22 μίτι filtered) was taken along in the same dilutions as rhdlNH. Readout was deposition of C5b-9, the normal serum control in the assay defining 100% (y-axis). Data are mean and SD (n = 3).
Figure 16 Effect of rhdlNH and pdCIINH on activation of both the classical and MBL pathway of complement activation. Increasing doses of rhC1 INH or pdC1 INH was added to two different samples of normal human serum. As a control, the buffer in which rhC1 INH is dissolved (20 mM citrate, 0.19 M sucrose pH 6.8; 0.22 μπι filtered) was taken along in the same dilutions as rhC1 INH. Readout was deposition of C5b-9 and the percentage complement activation was calculated per measurement with this formula: (Sample-NC)/(PC-NC)x100. PC is set at 100%. Results shown are the mean SD of 3 independent verdunning at each concentration tested.
Examples
Example 1 [0037] Previous experiments showed that a single dose of rhdlNH (15U/mouse) administered at the beginning of the ischemic period, significantly reduces ischemic volume, as assessed 48 hours after ischemia in our mouse model of cerebral focal ischemia in a manner very similar to plasma derived C1 INH. In this Example we have explored the time window of efficacy for rhC1 INH neuro-protective activity on the ischemic volume and functional deficits. We have also studied the effect of rhC1 INH on seven-days outcome by assessing the neurodegeneration and glial response. 1. Methods 1.1 Transient focal cerebral ischemia [0038] Ischemia was achieved by middle cerebral artery occlusion (MCAO) as previously described (De Simoni étal., 2003 and 2004, supra). Anesthesia was induced by 5% isoflurane in N2O/O2 (70/30%) mixture and maintained by 1.5-2% isoflurane in the same mixture. To confirm the adequacy of the vascular occlusion in each animal, blood flow was measured by laser doppler flowmetry (Transonic BLF-21) using a flexible 0.5 mm fiberoptic probe (Transonic, Type M, 0.5 mm diameter) positioned on the brain surface and secured with impression material on the skull at the following coordinates: AP = - 1mm; L= -3,5mm. Briefly, the right common carotid artery was exposed and a siliconized filament (7-0), was introduced into the internal carotid artery through an incision performed on the common carotid artery and advanced to the anterior cerebral artery so as to block its bifurcation into the anterior cerebral artery and the MCA. The filament was advanced until a >70% reduction of blood flow, compared to preischemic baseline, was observed. After 30 min of ischemia, blood flow was restored by carefully removing the nylon filament. 1.2 Drug treatment [0039] Mice received a single iv injections of rhdlNH at the dose of 15U/mouse in 150 μΙ or the same volume of saline at different time from ischemia: at the beginning of ischemic period (rhC1 INH -pre), at the end of ischemic period (rhC1 INH -post). one hour after the beginning of the ischemic period (rhC1 INH 1 h -post). rhC1 INH used in this study was produced in transgenic rabbits that express human C1 INH in their mammary glands and purified from the milk obtained from these animals as described in WO 01/57079. 1.3 Evaluation of neurological deficits [0040] Forty-eight hours after ischemia, each mouse was rated on two neurological function scales unique to the mouse, by a trained investigator blinded to the experimental conditions. For general deficits mice were scored from 0 to 28 in each of the following categories: hair, ears, eyes, posture, spontaneous activity, epileptic behavior. For focal deficits mice were scored from 0 to 28 in each of the following categories: body symmetry, gait, climbing, circling behavior, front limb symmetry, compulsory circling, sensory response. Data are expressed as median and 25th to 75th percentiles. 1.4 Quantification of infarct size [0041] Forty-eight hours after ischemia, mice were deeply anesthetized with Equitensin (120 μΙ/mice, ip) and tran-scardially perfused with 30ml of PBS 0.1 mol/l, pH 7.4, followed by 60ml of chilled paraformaldheyde (4%) in PBS. After carefully removing the brains from the skull, they were transferred to 30% sucrose in PBS at 4°C overnight for cryoprotection. The brains were then rapidly frozen by immersion in isopentane at -45°C for 3 min before being sealed into vials and stored at -70°C until use. For lesion size determination, 20 μπι coronal brain sections were cut serially at 240 μπι intervals and stained with neutral red (Neutral Red Gurr Certistain, BDH, England). On each slice, infarcted areas were assessed blindly and delineated by the relative paleness of histological staining. The infarcted area was determined by subtracting the area of the healthy tissue in the ipsilateral hemisphere from the area of the contralateral hemisphere on each section. Infarct volumes were calculated by the integration of infarcted areas on each brain slice as quantified with computer-assisted image analyzer and calculated by Analytical Image System. 1.5 Open field test [0042] Seven days after ischemia mouse behavior was evaluated by the open field test. This test may be useful to dectect anxiety and exploratory behavior, and locomor activity in long-term ischemic mice. The open filed consisted of a plastic box (41 x 41 x 41cm) containing 4 different objects. The area of the open field was divided into a 28 x 28cm central zone and the surrounding border zone. Mice were individually placed into the centre of the open field and their behavior was observed for 5 minutes by an investigator blinded to the experimental conditions. The number of inside crossings (mainly related to anxiety behavior), outside crossings (mainly related to motor activity), rears (mainly related to exploratory behavior) and contacts with objects (mainly related to sensory/motor activity) was counted. 1.6 Neuronal count [0043] Seven days after ischemia, mice were transcardially perfused as previously described. For neuronal count determination, 20 μπι coronal brain sections were cut serially at 640 μπι intervals and stained with cresyl violet (Cresyl Violet acetate, Sigma, St. Louis, MO). Three 20 μπι sections from ipsi- and controlatéral hemispheres were selected for neuronal count. The first section was at stereotaxic coordinates anteroposterior +0.86 from bregma. The amount of neuronal loss was calculated by pooling the number of viable neurons in the three sections of both hemispheres and expressed as percentage of controlatéral hemisphere. An Olympus BX61 microscope, interfaced with Soft Imaging System Colorview video camera and Analysis software was used. The quantitative analysis was performed at 40X magnification by an investigator blinded to the treatment. 1.7 Immunohistochemistry for astrocytes and microglia [0044] Seven days after ischemia twenty-p.m-thick coronal sections from transcardially perfused ischemic mice were prepared and used for assessment of astrocytes and microglia/macrophages immunostaining. Briefly, the sections were rinsed for 30 minutes in 0.4% Triton X-100 in 0.1 mol/L PBS followed by 15 minutes in 0.1 % Triton X-100 and 3% normal goat serum (NGS) in PBS. The sections were then incubated overnight with antibody for astrocytes and microglia (anti-GFAP 1:1500, Chemicon; anti-CD11 b 1:250, kindly gift by Dr. A. Doni, Mario Negri Institute). The next day, the sections were washed in PBS and incubated with biotinylated secondary antibody for 1 h, washed and incubated with avidin-biotin-peroxidase. After reacting with 3’-3-diaminobenzidine tetrahydrochloride the sections were washed, dried, dehy-dratate through graded alcohols, fixed in xylene and coverslipped using DPX mountant before light microscopy analysis. 2. Results 2.1 Time-window of efficacy 2.1.1 Evaluation of neurological deficits [0045] Neurological deficits were evaluated in ischemic mice receiving rhC1 INH or saline 48h after ischemia. A slight, although not significant, decrease in every group of rhC1 INH-treated mice was observed compared to saline-treated ischemic mice (rhdlNH -pre: 9 and 12; rhC1 INH-post: 7 and 11; rhdlNH 1h -post: 9 and 13, saline: 10 and 12.5, median of general and focal deficits, respectively) (data not shown). 2.1.2 Assessment of infarct size [0046] Forty-eight hours after ischemia rhC1 INH-treated mice showed a marked reduction of the ischemic volume, at 15U/mouse -pre, -post and 1h -post doses (13.67 ± 2.59mm3, 9.06 ± 0.77mm3 and 8.24 ± 1.00 mm3, respectively), compared to saline-treated mice (41.51 ± 7.01mm3) (Figure 1, data are expressed as mean ± SEM). 2.2 Seven-days outcome 2.2.1 Open field test [0047] Ischemia induced a significant reduction in the number ofrears compared to naive animals while in the rhC1 INH-treated group this parameter was not different from non-ischemic mice. The other parameters evaluated did not show any difference among the three groups. 2.2.2 Neuronal count [0048] To evaluate if the protective effect of rhdlNH is long lasting, we assessed the neuronal loss 7 days after induction of ischemia and treatment with the drug. The results show that rhC1 INH protective effect is still present at this time: 14% ± 2.18% versus 4% ± 1.24% mean of saline- and rhC1 INH -treated mice, respectively (data not shown). 2.2.3 Immunohistochemistry for microglia/macrophages and astrocytes [0049] Seven days after ischemia a large amount of activated microglia and infiltrated macrophages were observed in the lesioned hippocampus and striatum of ischemic mice receiving saline (data not shown). Fifteen units of rhC1 INH-pre were able to counteract this activation and infiltration in both the areas considered (data not shown).
[0050] The ipsilateral hippocampus of saline-treated ischemic mice showed a slight astrocytosis which was not different from that observed in rhC1 INH- ischemic mice (data not shown). Other brain areas did not show any relevant astrocytic activation in either groups. 3. Conclusions [0051] The present data show that rhC1 INH at the dose of 15U/mouse is similarly effective in reducing the ischemic volume when given at the beginning (-pre) or at the end of ischemic period (-post, i.e. at reperfusion). More importantly the inhibitor is able to exert its neuroprotective effects also when injected 1 hour after the onset of ischemia (1 h - post). Furthermore, the protective action of rhC1 INH is still present 7 days after ischemia. These results are in sharp contrast with plasma derived hCIINH which when injected 1 hour after ischemia has nearly completely lost the ability to exert neuroprotective effects (see Figure 2).
[0052] The main results of this study are the following: 1. the half life of rhC1 INH in mouse plasma is about 3 hours (at a dose of 15U/mouse). The good correlation between antigen and functional activity indicates that the recombinant protein circulates in plasma in its active form only; it is possible that tissue distribution contributed to the reduction of plasma levels. 2. rhC1 INH, at the dose of 15U/mouse -pre is very effective in reducing the ischemic volume (reduction of 69%). 3. rhC1 INH at the dose of 15U/mouse is able to clearly reduce the number of degenerating neurons in the hippocampus as assessed by Fluoro-Jade staining thus indicating that the reduction in ischemic volume is due to sparing of neurons. 4. rhC1 INH is similarly effective in reducing ischemic volume when given at the beginning (-pre), at the end of the ischemic period (-post, i.e. at reperfusion) or 1 hour after the onset of ischemia (1 h -post, i.e. 30 min from beginning of reperfusion). Thus rhC1 INH has a wider time-window of efficacy than pdC1 INH (that is no more effective when given 1h after ischemia). 5. the neuroprotective effect of rhC1INH-pre dose is long-lasting, as showed by neuronal counting performed 7 days after the beginning of ischemia. 6. rhdlNH, induced a slight improvement of general and focal deficits assessed 48 hours after ischemia. This finding is similar to what observed with pdC1 INH. In order to evaluate the effect of rhC1 INH on long-term behavioral outcome, we analyzed mouse behavior by open field test. Seven days after ischemia the rearing behavior shows a significantly lower score in the ischemic compared to naive mice. This decrease is not present in rhC1 INH treated mice whose score is not different from control mice. 7. rhdlNH is able to counteract the activation/recruitment of microglia/macrophages in ischemic mice brain as assessed both at early (48 h) and at late (7 days) time points. These cells are an index of the inflammatory response of the brain tissue. 8. The strong astrocytic response elicited by ischemia at 48h is dampened by rhC1 INH. The astrocytic activation is markedly decreased at 7 days in both experimental groups and no difference between saline- and rhC1 INH-treated mice could be observed.
Example 2: study on the neuroprotective action of rhC1-INH in mouse models of focal cerebral ischemia [0053] We have previously demonstrated that 15U of rhC1-INH have a marked neuroprotective action in a model of murine cerebral ischemia/reperfusion also when administrated 1 hourafterthe onset of ischemia/reperfusion, atvariance with pdC1-ENH that, at this time of post-treatment, is no longer effective. This neuroprotection is long-lasting, in fact seven days after ischemia and treatment, ischemic brains of mice treated with rhC1-INH still show a decreased infarct size. In the following experiments we have determined the time window of efficacy (beyond 1 hour post) and the dose-response of rhC1-INH neuroprotective activity on the ischemic volume. In addition we have performed a direct comparison among pdC1-INH, rabbit and cow rhC1-INH (at the most effective dose and time-points for rabbit rhC1-INH) using the same protocol.
METHODS
Animals [0054] Procedures involving animals and their care was conducted in conformity with institutional guidelines that are in compliance with national (D.L. n.116, G.U. suppl. 40, 18 February 1992) and international laws and policies (EEC Council Directive 86/609, OJ L 358,1 ; Dec. 12,1987; NIH Guide for the Care and Useof Laboratory Animals, U.S. National Research Council 1996). Male C57B1/6 mice (26-28 g, Charles River, Calco, Italy) were housed 5 per cage and kept at constant temperature (21±14°C) and relative humidity (60 %) with regular light/dark schedule (7 am-7 pm). Food (Altromin pellets for mice) and water available ad libitum.
Transient focal cerebral ischemia [0055] Ischemia was achieved by middle cerebral artery occlusion (MCAO) as previously described1"3. Anesthesia was induced by 5% isoflurane in NaO/Oa (70/30%) mixture and maintained by 1.5-2% isoflurane in the same mixture. To confirm the adequacy of the vascular occlusion in each animal, blood flow was measured by laser doppler flowmetry (Transonic BLF-21) using a flexible 0.5 mm fiberoptic probe (Transonic, Type M, 0.5 mm diameter) positioned on the brain surface and secured with impression material on the skull at the following coordinates: AP = -1mm; L= -3,5mm. Briefly, the right common carotid artery was exposed and a siliconized filament (7-0) was introduced into the internal carotid artery through an incision performed on the common carotid artery and advanced to the anterior cerebral artery so as to block its bifurcation into the anterior cerebral artery and the MCA. The filament was advanced until a >70% reduction of blood flow, compared to preischemic baseline, was observed. After 30 min of ischemia, blood flow was restored by carefully removing the nylon filament.
Drug treatment [0056] Mice received a single iv injections of C1-INH (rabbit rhC1-INH, cow rhC1-INH or pdC1-INH) at different doses at different times from ischemia. Control mice received the same volume of saline.
Evaluation of neurological deficits.
[0057] Forty-eight hours after ischemia, each mouse was rated on two neurological function scales unique to the mouse, by a trained investigator blinded to the experimental conditions. For general deficits mice were scored from 0 to 28 in each of the following categories: hair, ears, eyes, posture, spontaneous activity, epileptic behavior. For focal deficits mice were scored from 0 to 28 in each of the following categories: body symmetry, gait, climbing, circling behavior, front limb symmetry, compulsory circling, sensory response. Data are expressed as median and percentiles.
Quantification of infarct size [0058] Forty-eight hours after ischemia, mice were deeply anesthetized with Equitensin (120jil/mice, ip) and transcardially perfused with 30ml of PBS O.1mol/I, pH 7.4, followed by 60ml of chilled paraformaldheyde (4%) in PBS. After carefully removing the brains from the skull, they were transferred to 30% sucrose in PBS at 4°C overnight for cryoprotection. The brains were then rapidly frozen by immersion in isopentane at -45°C for 3 min before being sealed into vials and stored at -70°C until use. For lesion size determination, 20fjm coronal brain sections were cut serially at 240(j,m intervals and stained with neutral red (Neutral Red Gurr Certistain, BDH, England). On each slice, infarcted areas were assessed blindly and delineated by the relative paleness of histological staining. The infarcted area was determined by subtracting the area of the healthy tissue in the ipsilateral hemisphere from the area of the contralateral hemisphere on each section. Infarct volumes were calculated by the integration of infarcted areas on each brain slice as quantified with computer-assisted image analyzer and calculated by Analytical Image System.
Assessment of neurodegeneration [0059] The presence of neurodegeneration was evaluated on 20 jam thick sections by staining with Fluoro-Jade4, a marker for neuronal degeneration. Briefly, sections were dried and rehydrated in ethanol (100% - 75%) and distilled water. Then, they were incubated in 0.06% potassium permanganate for 15 minutes, washed in distilled water and transferred to 0.001 % Fluoro-Jade staining solution for 30
minutes. After staining, the sections were rinsed in distilled water, dried, immerse in xylene and coverslipped using DPX mountant (BDH, Poole, UK) before fluorescent microscopy analysis.
RESULTS
TIME-WINDOW of EFFICACY IN TRANSIENT ISCHEMIA
[0060] In order to evaluate the time-window of efficacy, 15U of rabbit rhC1-INH or saline were given at 3, 6, 9, 18 and 24 hours from the beginning of ischemia. Forty-eight hours later, ischemic mice treated with rabbit rhC1-INH 3 and 6 hours after the onset of ischemia showed a marked decrease of ischemic volume (11.71 ± 0.63mm3 and 20.38±2.37mm3, respectively) compared to saline-treated ischemic mice (44.43±5.94mm3). Also when administrated 9 and 18 hours after ischemia, rabbit rhC1-INH was still effective, although to a minor extent (23.63±4.11 mm3 and 27.13±2.58mm3 respectively). Twenty-four hours after ischemia the inhibitor lost its beneficial action (41.92±2.76mm3). (Fig.3). In saline-treated mice, Fluoro-Jade staining showed that, neurodegeneration was present in striatum cortex and hippocampus. When administrated at early time points, rhC1-INH was able to counteract the neurodegeneration in hippocampus (up to 3 hours) and in cortex (up to 9 hours). When mice were treated with this inhibitor 6 and 9 hours after ischemia, some degenerating neurons were observed in hippocampus. At later time points oftreatment (18 and 24 hours), when the ischemic volume was larger, Fluoro-Jade staining showed the presence of neurodegenerating neirons in cortex. At all time points considered, striatum showed an extensive neurodegeneration, both in saline- and rhC1-INH- treated animals (Fig.5, 6, 7). A semi-quantitative evaluation of Fluoro-Jade staining for each animal was performed by an investigator blinded to the experimental conditions (figure 4).
DOSE-RESPONSE IN TRANSIENT ISCHEMIA
[0061] Since the dose of C1-INH used in humans for hereditary angioedema is lower than the one we used in mice for stroke treatment, lower doses were used in our ischemic model. Based on the results of the previous experiment we chose 3h post treatment for dose-response experiment. Different doses of rabbit rhC1-INH (5 and 10 units) were given 3 hours after the onset of ischemia and reperfusion. The dose of 1OU/mouse was still effective in reducing the ischemic volume (22.10± 3.65mm3), while 5U of rabbit rhC1-INH did not modify the extent of the brain damage (47.39±4.08mm3). These data show that rabbit rhC1-INH is able to modify the ischemic lesion in a dose-dependent manner (fig.8).
In mice treated with 10U of rhC1-INH some neurodegenerating neurons, as evidenced by Fluoro-Jade staining, were observed in striatum but not in hippocampus and cortex, while 5U-treated ischemic mice displayed a large neurodegeneration in striatum cortex (not shown).
[0062] General and focal neurological deficits did not show any significative variations either in time-window of efficacy or in dose-response experiment (not shown).
[0063] COMPARISON BETWEEN THE EFFECT OF pdC1-INH and rhC1-INH (from rabbits and cows) Our previous data on pdCI-INH were obtained with a different model of transient cerebral ischemia. In order to directly compare pdCIINH, cow rhC1-INH and rabbit rhC1-INH, these compounds were given to mice in which ischemia was induced with the same experimental protocol (silicone-coated filament). The inhibitors were administrated at the dose of 15U/mouse 3 hours after the onset of ischemia.
As expected, pdC1-INH was not able to exert a neuroprotective action at this time point (47.39±4.08mm3). At variance, cow rhC1-INH-treated ischemic mice showed a significantly reduced ischemic volume compared to saline-treated mice, even though to a lower extent than rabbit rhC1-INH-treated mice (Fig.9). Surprisingly both general and focal deficits were significantly improved by cow rhC1-INH (Fig. 10).
Fluoro-Jade staining showed a large neurodegeneration in the brain of ischemic mice treated with pdC1-INH in all the considered areas (cortex, striatum and hippocampus). The staining of the brain of cow rhC1-INH-treated mice showed a variable grade of neurodegeneration in cortex and hippocampus since in 3 out of 6 mice a marked neurodegeneration was observed in both these areas, while in the other 3 mice the neurodegeneration was present in a very little amount. The striatum displayed an extensive Fluoro-Jade staining in 6 out of 6 mice.
COMMENTS
[0064] The most relevant data of this work is the time-window of efficacy of rabbit rhC1-INH. The dose of 15U/mouse of rabbit rhC1-INH was able to significantly reduce the ischemic volume up to 18 hours after the onset of ischemia at variance with pdC1-INH that 3 hours after ischemia has already lost its neuroprotective effect. This surprising feature makes rhC1-INH a possible candidate for stroke therapy in humans. The different efficacy of pd and rhC1-INHs, could be due to the different glycosylation of the two molecules resulting in turn to a higher affinity for a mannose binding protein (MBP) of rhC1-INH as compared to the plasma derived one. Binding MBP, rhC1-MH causes the inhibition of the complement lectin pathway, involved in the pathogenesis of the damage in heart, kidney and gastrointestinal ischemia/reperfusion7"9. The role of this poorly characterized pathway is still unknown in brain ischemia and further experiments are required in order to clarify the mechanism of rhC1-INH neuroprotection.
The superior neuroprotective effect of rhC1 INH over pdC1 INH in the time-window after the onset of ischemia may further be explained by a more efficient targeting of the recombinant molecule to the site of tissue damage eitherthrough binding to cell-surface antigens and/or a more efficient tissue penetration. More research needs to be done to fully elucidate the exact molecular mechanism underlying the observation described in this invention.
Fluoro-Jade staining gives indirect evidence of how the lesion evolves in time. The early treatment with rhC1 -INH provides a complete rescue of the ischemic penumbra (hippocampus and cortex). The later the treatment is administrated, the more neurons in the penumbra degenerate. These findings confirm that rhC1-INH exerts its neuroprotective action on ischemic penumbra. Rabbit rhC1-INH is able to reduce ischemic volume in a dose dependent-manner. The most effective dose ofrhC1-INH (15U/mouse, corresponding about to 600U/kg), used for time-window of efficacy experiment, is much higher than the one used in humans for hereditary angioedema (about 25-100U/kg). In order to verify if a lower dose was still effective in reducing neurodegeneration and ischemic infarct, a dose-response experiment was performed. The results showed that 400U/kg (10 U/mouse) of rhC1-INH were still able to significantly counteract the ischemic insult, although to a lower extent. A dose 8 fold higher than the one used for HAE (5U/mouse, 200U/kg) was not effective. These findings are in line with evidence showing that large doses of C1-INH are required for therapeutic application in various inflammation settings5. In particularsuch doses are necessary to reach an important inhibitory effect on endothelial adhesion molecules6, a mechanism involved in the pathogenesis of ischemia/reperfusion brain damage. Lastly, cow rhC1-INH provided neuroprotection, when given 3 hours after ischemia at the dose of 15U/mouse, although less markedly than rabbit rhC1-INH. The inhibitor from cow was also able to improve neurological deficits compared to saline-treated mice. These findings indicate that this molecule is able to ameliorate the general conditions of ischemic mice.
Example 3 : Comparison of the ability of rhdlNH and plasma derived C1INH to inhibit activation of the classical and MBL pathways
Materials and Methods [0065] The effect of rhC1 INH and pdC1 INH (Cetor, Sanquin, Amsterdam, The Netherlands) on the function of the classical and lectin pathway was examined in the Wieslab TM complement system Screen (Euro-Diagnostica, Malmo, Sweeden) using two different sources of serum. One serum source is included in the kit, where it is used as a positive control (hereafter referred to as serum sample 1 ). The other serum sample was obtained from a commercially available pool of human serum (pool of 25 different donors; Kordia, Leiden, The Netherlands), hereafter referred to as serum sample 2. Both serum samples were incubated in independent triplo’s with 0, 15, 30 and 75 μίτιοΙ rhC1 INH or pdC1 INH for 30 min at room temperature. Therefore, stock solutions of pdC1 INH and rhC1 INH were diluted in water to appropriate concentrations. Volumes corresponding with 15, 30 and 75 μίτιοΙ rhdlNH or pdCIINH were taken and adjusted to 15 μΙ with water. The buffer in which rhdlNH is dissolved (20 mM citrate, 0.19 M sucrose pH 6.8; 0.22 μπι filtered) was taken along in the same dilutions as rhdlNH to control for interference with the Wieslab Complement System. The positive controls (PC) and negative controls (NC) from both the classical and MBL pathway (provided with the kit), and both serum samples were diluted 1/101 in Diluent CP for the classical pathway and Diluent MP for the MBL pathway according to manufacturer’s instructions. Of these diluted sera, 127.5 μΙ was supplemented with 22.5 μΙ water, pdC1 INH, rhdlNH or buffer and incubated for 30 minutes at RT. Next, 100 μΙ/well of PC, NC, Diluent CP or MP (blanks) and samples were pipetted on the appropriate plate and incubated for 1 hour at 37 °C. After incubation the wells were washed 3 times with 300 μΙ/well washing solution and subsequently incubated for 30 minutes at room temperature with 100 μΙ/well of conjugate. After another wash, wells were incubated with 100 μΙ/well of substrate and again incubated for 30 minutes at roomtemperature. The reaction was stopped by the addition of 100 μΙ/well 5 mM EDTA and the absorbance was read at 405 nm.
[0066] For the calculation of the results, the absorbance of the blanks (Diluent CP or MP) was subtracted from the PC, NC and samples. The percentage complement activation was calculated per measurement with this formula: (Sam-ple-NC)/(PC-NC)x100. This means that the PC is always set at 100%. For each condition the mean, standard deviation and CV% was calculated.
Results
Effect of rhC1 INH and pd-C1 INH on the classical pathway as examined by Wielisa.
[0067] The inhibitory properties of both rhC1 INH and pdC1 INH on classical pathway activation were analyzed in two different serum samples. As shown in Figures 12, 13 and 16, both rhdlNH and pdCIINH dose-dependently reduced the classical pathway mediated C5b-9 deposition in both sera samples. Whereas rhdlNH - at a concentration of 75 μΜ - seems to inhibit the classical pathway activation in serum 1 slightly stronger than pdCIINH, such an effect was not seen in serum sample 2. At all other concentrations tested no differences in inhibitory properties were observed between rhC1 INH and pdC1 INH. Therefore, it was concluded that both rhC1 INH and pdC1 INH are equally effective in inhibiting classical pathway activation in human sera.
Effect of rhdlNH and pdCIINH on the MBL pathway as examined by Wielisa.
[0068] In the same set of experiments, also the inhibitory properties of both rhC1 INH and pdC1 INH on the MBL pathway activation were analyzed. As shown in Figures 14, 15 and 16, both rhdlNH and pdCIINH also dose-dependently reduced activation of the MBL pathway. However, in contrast to the classical pathway where no differences were seen, rhC1 INH appeared to be a more potent inhibitor of the MBL pathway when compared to pdC1 INH. At all 3 concentrations tested and in both serum samples, the rhC1 INH-mediated inhibition of the MBL pathway is - 20% higher as compared to pdC1 INH. Therefore, it was concluded that rhC1 INH is a more effective inhibitor of the MBL pathway than pdC1 INH.
Conclusion [0069] The results show that both rhC1 INH and pdC1 INH are equally effective in inhibiting the classical pathway, but rhC1 INH is a more potent inhibitor of the MBL pathway. At all concentrations tested, rhC1 INH mediated MBL pathway inhibition was -20% stronger as compared to pdC1 INH.
REFERENCES
[0070] 1. De Simoni, M. G. et al. Neuroprotection by complement (C1) inhibitor in mouse transientbrain ischemia. J Cereb Blood Flow Met 23, 232-239 (2003). 2. De Simoni, M. G. et al. The powerful neuroprotective action of C1-inhibitor on brainischemia-reperfusion injury does not require C1q. Am JPathol 164, 1857-63 (2004). 3. Storini, C. et al. C1 inhibitor protects against brain ischemia-reperfusion injury via inhibitionof cell recruitment and inflammation. Neurobiol Disease 19,10-17 (2005). 4. Schmued, L. C. &amp; Hopkins, K. J. Fluoro-Jade B: a high affinity fluorescent marker for thelocalization of neuronal degeneration. Brain Res 874, 123-30. (2000). 5. Caliezi, C. et al. C1 esterase inhibitor: an anti-inflammatory agent and its potential use in thetreatment of diseases other than hereditary angioedema. Pharmacol Rev 52,91-112 (2000). 6. Cai, S. et al. A direct role for C1 inhibitor in regulation of leukocyte adhesion. J Immunol174, 6462-6 (2005). 7. Walsh, M. C. et al. Mannose-binding lectin is a regulator of inflammation that accompaniesmyocardial ischemia and reperfusion injury. J Immunol 175, 541-6 (2005). 8. Moller-Kristensen, M. etal. Mannan-binding lectin recognizes structures on ischaemicreperfused mouse kidneys and is implicated in tissue injury. Scand Jlmmunol 61,426-34 (2005). 9. Hart, M. L. et al. Gastrointestinal ischemia-reperfusion injury is lectin complement pathwaydependent without involving Clq. J Immunol 174, 6373-80 (2005).
SEQUENCE LISTING
[0071] <110> Pharming Intellectual Property B.V. <120> Use of C1 inhibitor for the prevention of ischemia-reperfusion injury <130> P6005723EP1 <150> 05112630.8 <151> 2005-12-21 <150> 60/760,944 <151> 2006-01-23 <160> 1 <170> Patentln version 3.3 <210> 1 <211 > 478
<212> PRT <213> Homo sapiens <400> 1
Asn Pro Asn Ala Thr Ser Ser Ser Ser Gin Asp Pro Glu Ser Leu Gin 15 10 15
Asp Arg Gly Glu Gly Lys Val Ala Thr Thr Val Ile Ser Lys Met Leu 20 25 30
Phe Val Glu Pro Ile Leu Glu Val Ser Ser Leu Pro Thr Thr Asn Ser 35 40 45
Thr Thr Asn Ser Ala Thr Lys Ile Thr Ala Asn Thr Thr Asp Glu Pro 50 55 60
Thr Thr Gin Pro Thr Thr Glu Pro Thr Thr Gin Pro Thr Ile Gin Pro 65 70 75 80
Thr Gin Pro Thr Thr Gin Leu Pro Thr Asp Ser Pro Thr Gin Pro Thr 85 90 95
Thr Gly Ser Phe Cys Pro Gly Pro Val Thr Leu Cys Ser Asp Leu Glu 100 105 110
Ser His Ser Thr Glu Ala Val Leu Gly Asp Ala Leu Val Asp Phe Ser 115 120 125
Leu Lys Leu Tyr His Ala Phe Ser Ala Met Lys Lys Val Glu Thr Asn 130 135 140
Met Ala Phe Ser Pro Phe Ser Ile Ala Ser Leu Leu Thr Gin Val Leu 145 150 155 160
Leu Gly Ala Gly Glu Asn Thr Lys Thr Asn Leu Glu Ser Ile Leu Ser 165 170 175
Tyr Pro Lys Asp Phe Thr Cys Val His Gin Ala Leu Lys Gly Phe Thr 180 185 190
Thr Lys Gly Val Thr Ser Val Ser Gin Ile Phe His Ser Pro Asp Leu 195 200 205
Ala Ile Arg Asp Thr Phe Val Asn Ala Ser Arg Thr Leu Tyr Ser Ser 210 215 220
Ser Pro Arg Val Leu Ser Asn Asn Ser Asp Ala Asn Leu Glu Leu Ile 225 230 235 240
Asn Thr Trp Val Ala Lys Asn Thr Asn Asn Lys Ile Ser Arg Leu Leu 245 250 255
Asp Ser Leu Pro Ser Asp Thr Arg Leu Val Leu Leu Asn Ala Ile Tyr 260 265 270
Leu Ser Ala Lys Trp Lys Thr Thr Phe Asp Pro Lys Lys Thr Arg Met 275 280 285
Glu Pro Phe His Phe Lys Asn Ser Val Ile Lys Val Pro Met Met Asn 290 295 300
Ser Lys Lys Tyr Pro Val Ala His Phe Ile Asp Gin Thr Leu Lys Ala 305 310 315 320
Lys Val Gly Gin Leu Gin Leu Ser His Asn Leu Ser Leu Val Ile Leu 325 330 335
Val Pro Gin Asn Leu Lys His Arg Leu Glu Asp Met Glu Gin Ala Leu 340 345 350
Ser Pro Ser Val Phe Lys Ala Ile Met Glu Lys Leu Glu Met Ser Lys 355 360 365
Phe Gin Pro Thr Leu Leu Thr Leu Pro Arg Ile Lys Val Thr Thr Ser 370 375 380
Gin Asp Met Leu Ser Ile Met Glu Lys Leu Glu Phe Phe Asp Phe Ser 385 390 395 400
Tyr Asp Leu Asn Leu Cys Gly Leu Thr Glu Asp Pro Asp Leu Gin Val 405 410 415
Ser Ala Met Gin His Gin Thr Val Leu Glu Leu Thr Glu Thr Gly Val 420 425 430
Glu Ala Ala Ala Ala Ser Ala Ile Ser Val Ala Arg Thr Leu Leu Val 435 440 445
Phe Glu Val Gin Gin Pro Phe Leu Phe Val Leu Trp Asp Gin Gin His 450 455 460
Lys Phe Pro Val Phe Met Gly Arg Val Tyr Asp Pro Arg Ala 465 470 475
Claims 1. Use of a C1 inhibitor that has a reduced level of terminal sialic acid residues as compared to plasma derived C1 inhibitor, wherein the reduced level of terminal sialic acid residues results in a plasma half life of less than 6 hours, in the preparation of a pharmaceutical composition for the prevention, reduction or treatment of an ischemia/reperfusion injury, whereby the C1 inhibitor is administered at most 3 hours, 2 hours, 1 hour, or 30 minutes before at least one foreseen occurrence of ischemia and reperfusion injury. 2. Use according to claim 1, wherein said foreseen occurrence of ischemia and reperfusion injury is organ transplantation. 3. Use according to any of claims 1 or 2, wherein said C1 inhibitor is administered continuously to a subject in need thereof. 4. Use according to claim 1 or 2, wherein said C1 inhibitor is administered to the organ to be transplanted.
Patentansprüche 1. Verwendung eines C1-Inhibitors, der im Vergleich zu von Plasma abgeleitetem C1-Inhibitor einen verringerten Anteil an terminalen Sialinsäureresten aufweist, wobei der verringerte Anteil an terminalen Sialinsäureresten zu einer Plasma-Halbwertszeit von weniger als 6 Stunden führt, bei der Herstellung einer pharmazeutischen Zusammensetzung zur Verhinderung, Verringerung oder Behandlung eines Ischämie/Reperfusionsschadens führt, wobei der C1-Inhibitor mindestens 3 Stunden, 2 Stunden, 1 Stunde oder 30 Minuten vor mindestens einem vorherzusehenden Auftreten von Ischämie und Reperfusionsschaden verabreicht wird. 2. Verwendung nach Anspruch 1, wobei es sich beim vorherzusehenden Auftreten von Ischämie und Reperfusionsschaden um eine Organtransplantation handelt. 3. Verwendung nach Anspruch 1 oder 2, wobei der C1-Inhibitor einem behandlungsbedürftigen Subjekt kontinuierlich verabreicht wird. 4. Verwendung nach Anspruch 1 oder 2, wobei der C1 -Inhibitor dem zu transplantierenden Organ verabreicht wird.
Revendications 1. Utilisation d’un inhibiteur de C1 qui a un niveau réduit de résidus d’acide sialique terminal par comparaison à l’inhibiteur de C1 dérivé de plasma, dans laquelle le niveau réduit des résidus d’acide sialique terminal entraîne une demi-vie plasmatique de moins de 6 heures, dans la préparation d’une composition pharmaceutique pour la prévention, la réduction ou le traitement d’une lésion d’ischémie/reperfusion, de telle sorte que l’inhibiteur de C1 est administré au plus 3 heures, 2 heures, 1 heure ou 30 minutes avant au moins une occurrence prévue de lésion d’ischémie et de reperfusion. 2. Utilisation selon la revendication 1, dans laquelle ladite occurrence prévue d’une lésion d’ischémie et de reperfusion est une transplantation d’organe. 3. Utilisation selon l’une quelconque des revendications 1 ou 2, dans laquelle ledit inhibiteur de C1 est administré en continu à un sujet en ayant besoin. 4. Utilisation selon la revendication 1 ou 2, dans laquelle ledit inhibiteur de C1 est administré à l’organe devant être transplanté.
REFERENCES CITED IN THE DESCRIPTION
This list of references cited by the applicant is for the reader’s convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.
Patent documents cited in the description • WO 2004100982 A [0003] · WO 05112630 A [0071] • US 6846649 B [0020] · WO 60760944 A [0071] • WO 0157079 A [0039]
Non-patent literature cited in the description • DE SIMONI et al. J Cereb Blood Flow Metab., 2003, · D.L., February 1992, vol. 40 (116), 18 [0054] vol. 23, 232-9 [0003] · EEC Council Directive 86/609. OJ L, 12 December • AKITA et al. Neurosurgery, 2003, vol. 52, 395-400 1987, vol. 358, 1 [0054] [0003] · NIH Guide for the Care and Use of Laboratory Ani- • DE SIMONI et al. Am J Pathol., 2004, vol. 164, mais. U.S. National Research Council, 1996 [0054] 1857-63 [0003] · DE SIMONI, M. G. et al. Neuroprotection by comple- • STORINI et al. Neurobiol Dis., 2005, vol. 19, 10-7 ment (C1) inhibitor in mouse transient brain ischemia.
[0003] J Cereb Blood Flow Met, 2003, vol. 23, 232-239 • SCHAPIRA et al. Complement, 1985, vol. 2, 111 [0070] [0007] · DE SIMONI, M. G. et al. The powerful neuroprotec- • DAVIS. Ann. Rev. Immunol., 1988, vol. 6, 595 [0007] tive action of C1-inhibitor on brain ischemia-reper- • CARTER et al. Euro. J. Biochem., 1988, vol. 173, fusion injurydoes notrequire C1q. Am JPathol, 2004, 163 [0007] vol. 164, 1857-63 [0070] • PERKINS et al. J. Mol. Biol., 1990, vol. 214, 751 · STORINI, C. étal. C1 inhibitor protects against brain [0007] ischemia-reperfusion injury via inhibition of cell re- • ALTSCHUL et al. J. Mol. Biol., 1990, vol. 215, cruitment and inflammation. Neurobiol Disease, 403-410 [0008] 2005, vol. 19, 10-17 [0070] • HENIKOFF ;HENIKOFF.Proc. Natl.Acad. Sei. USA, · SCHMUED, L. C. ; HOPKINS, K. J. Fluoro-Jade B: 1989, vol. 89, 10915 [0008] a high affinity fluorescent marker for the localization • HIGGINS D. et al. Nucleic Acids Res., 1994, vol. 22, of neuronal degeneration. Brain Res, 2000, vol. 874, 4673-4680 [0008] 123-30 [0070] • DROUET et al. Clin Chim Acta., 1988, vol. 174, · CALIEZI, C. étal. C1 esterase inhibitor: an anti-in- 121-30 [0009] flammatory agent and its potential use in the treat- • CHOU et al. J Biol Chem., 1996, vol. 271 (32), ment of diseases other than hereditary angioedema. 19219-24 [0015] Pharmacol Rev, 2000, vol. 52, 91-112 [0070] • J Biol Chem., 1994, vol. 269 (29), 18821-6 [0015] · CAI, S. et al. A direct role for C1 inhibitor in regulation • KAWASAKI et al. J. Biochem, 1983, vol. 94, 937-47 of leukocyte adhesion. J Immunol, 2005, vol. 174, [0020] 6462-6 [0070] • TAYLOR et al. Biochem. J., 1989, vol. 262 (3), · WALSH, M.C. et al. Mannose-binding lectin is a reg- 763-771 [0020] ulatorof inflammation that accompanies myocardial • WEIS et al. Nature, 1992, vol. 360, 127-34 [0020] ischemia and reperfusion injury. J Immunol, 2005, • J Immunology, 2005, vol. 175, 541-546 [0023] vol. 175, 541-6 [0070] • Am J Pathol., 2004, vol. 165 (5), 1677-88 [0023] · MOLLER-KRISTENSEN, M. et al. Mannan-binding • J Immunol., 2005, vol. 174 (10), 6373-80 [0023] lectin recognizes structures on ischaemic reperfused • DESIMONI et al. Am J. Pathol., 2004, vol. 164, mouse kidneys and is implicated in tissue injury. 1857-63 [0023] Scand Jlmmunol, 2005, vol. 61,426-34 [0070] • Remington’s Pharmaceutical Science. Mack Publish- · HART, M. L. et al. Gastrointestinal ischemia-reper- ing, 1980 [0029] fusion injury is lectin complement pathway dependent • NUIJENS et al. J. Clin. Invest., 1989, vol. 84 , 443 without involving Clq. J Immunol, 2005, vol. 174, [0031] 6373-80 [0070]

Claims (4)

•Szabadalmi. igénypontok• Patents. claims 1,. Ci Inhibitor» .amely a plazma «redetô: bamfe Cl tnlhbilwC összehasonlítva csökkent terminális szialissav-ínaradék szintet nmssh amely csökkent terminális 8ziaimsav..}x«»8.dék ssint § óránál tövidabh plasma felezési időt eredményez, alkalmazása feehémWr^erttMŐs Wilds megelőzésére. csökkentésére vagy kezelésé* realkalmas gyógyszerkészítmény előáílkásbs, ahol a Cl ittblbitött az isehémia és reperfúziós sérülés legalább egy elÖrelálfeató alö&amp;rdalása előtt hgíMjebh 1 órával.1 ,. Ci Inhibitor », which is the plasma« downer: bamfe Cl tnlhbilwC compared to reduced terminal sialissic acid-residual levels nmssh which reduced terminal 8ziaimsic acid.} X «» 8.i ssint § hour at work for plasma half-life, use for the prevention of wild-type wilds. for reducing or treating * a realistic drug formulation wherein Cl is present before at least one of the predisposing disorder of the ischemia and reperfusion injury is 1 hour. 2. órával, 1 órával vagy’ 30 perccel adjnk be, X Az 1. Igánypoth ázérinti alkalmazás, áhöl az isebémb' ás ragarlsAás sétttlés' említett etíkeláíkaíó előfordulása szervátülmtós,2 hours, 1 hour, or '30 minutes, X Applying the 1st Raspberry Echinacea, the etiquette of the echinaceae and scabies, is transverse, 3 . Áa. Avagy X igénypont szerkői alblteaa&amp;s, áhöl az emitted Cl ialdbitörí fölvsmatossn adjak ha egy ezt igénylő egyednsk.3. AA. Or, editor alblteale &amp; s of the X claim, and then the emitted Cl ialdbitöri will be provided if it is an individual requiring it. 4. Az. I:,. vagy X igéstypont szerinti^alkalmazás« ahol az említett Cl Inhibitort az álttltetendő szervbe adjak ha.4. The I:,. or wherein X is to be administered to the organ to be administered.
HUE11172880A 2005-12-21 2006-12-19 Use of C1 inhibitor for the prevention of ischemia-reperfusion injury HUE035692T2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP05112630 2005-12-21
US76094406P 2006-01-23 2006-01-23

Publications (1)

Publication Number Publication Date
HUE035692T2 true HUE035692T2 (en) 2018-05-28

Family

ID=36168534

Family Applications (1)

Application Number Title Priority Date Filing Date
HUE11172880A HUE035692T2 (en) 2005-12-21 2006-12-19 Use of C1 inhibitor for the prevention of ischemia-reperfusion injury

Country Status (7)

Country Link
CN (2) CN101365478B (en)
DK (1) DK1965831T3 (en)
ES (2) ES2369522T3 (en)
HU (1) HUE035692T2 (en)
LT (1) LT2380587T (en)
PT (2) PT1965831E (en)
SI (1) SI1965831T1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2793935T3 (en) * 2011-12-22 2016-09-05 Csl Behring Gmbh Use of C1 inhibitor in the treatment of secondary edema in the central nervous system.
HUE041837T2 (en) * 2013-03-15 2019-05-28 Shire Viropharma Inc C1-inh compositions and methods for the prevention and treatment of disorders associated with c1 esterase inhibitor deficency
CN111818938A (en) * 2018-02-28 2020-10-23 法明知识产权股份有限公司 Treatment and prevention of preeclampsia

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH07285866A (en) * 1994-02-22 1995-10-31 Eisai Co Ltd Agent for preventing, treating and improving organopathy
EP0679396A1 (en) * 1994-03-02 1995-11-02 Pfizer Inc. Use of 3-substituted-2-oxidole-1-carboxamides for the manufacture of a medicament in the treatment and prevention of ischemia induced myocardial injury and cytokine mediated myocardial injury
US20060142220A1 (en) * 2002-10-17 2006-06-29 Berkel Patrick V Protein modification
JP2006518749A (en) * 2003-02-21 2006-08-17 タノックス インコーポレーテッド Method for preventing and treating tissue damage associated with ischemia-reperfusion injury
ES2830499T3 (en) * 2003-05-16 2021-06-03 Pharming Intellectual Property B V C1 inhibitor with a short half-life for transient treatment

Also Published As

Publication number Publication date
LT2380587T (en) 2018-01-25
PT2380587T (en) 2018-01-19
CN101365478A (en) 2009-02-11
PT1965831E (en) 2011-10-19
CN101365478B (en) 2016-06-22
CN105641688A (en) 2016-06-08
SI1965831T1 (en) 2011-11-30
ES2654816T3 (en) 2018-02-15
DK1965831T3 (en) 2011-10-24
ES2369522T3 (en) 2011-12-01
CN105641688B (en) 2020-06-09

Similar Documents

Publication Publication Date Title
EP2380587B1 (en) Use of C1 inhibitor for the prevention of ischemia-reperfusion injury
TWI677348B (en) A way to improve heart disease
JP2023156429A (en) Method and drug for regulating and controlling glp-1/glp-1r
JP5400006B2 (en) Preventive or therapeutic agent for ischemic disease
TW202123955A (en) Method for preventing and treating tissue and organ fibrosis
IL275414B1 (en) Compounds for treating and preventing extracellular histone mediated pathologies
HUE035692T2 (en) Use of C1 inhibitor for the prevention of ischemia-reperfusion injury
EP0914830A1 (en) Neovascularization inhibitor containing tissue factor pathway inhibitor
JP7073486B2 (en) Peptide composition for the treatment of damage associated with excitatory neurotoxicity
JP6818871B2 (en) Therapeutic peptides for injuries associated with excitatory neurotoxicity
JP7237085B2 (en) Medicine for treatment and/or improvement of sepsis accompanied by coagulation abnormality
US20220372111A1 (en) Polypeptide inhibitors of neutrophil elastase activity and uses thereof
JP2019522675A (en) Methods of treating injury associated with excitatory neurotoxicity