EP0671941A1 - Bradykinin antagonists - Google Patents

Bradykinin antagonists

Info

Publication number
EP0671941A1
EP0671941A1 EP93924412A EP93924412A EP0671941A1 EP 0671941 A1 EP0671941 A1 EP 0671941A1 EP 93924412 A EP93924412 A EP 93924412A EP 93924412 A EP93924412 A EP 93924412A EP 0671941 A1 EP0671941 A1 EP 0671941A1
Authority
EP
European Patent Office
Prior art keywords
bkan
pharmacophore
heterodimer
peptide
heterodimer according
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP93924412A
Other languages
German (de)
French (fr)
Other versions
EP0671941A4 (en
Inventor
John C. Chronis
James K. Blodgett
Val Smith Goodfellow
Manoj V. Marathe
Lyle W. Spruce
Eric T. Whalley
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cortech Inc
Original Assignee
Cortech Inc
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 Cortech Inc filed Critical Cortech Inc
Publication of EP0671941A1 publication Critical patent/EP0671941A1/en
Publication of EP0671941A4 publication Critical patent/EP0671941A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/18Kallidins; Bradykinins; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to pharmaceutically effective heterodimers comprising a bradykinin antagonist (BKAn) component covalently linked to another different pharmacophore componen .
  • BKAn bradykinin antagonist
  • bradykinin antagonist dimers of the type: X(BKAn) 2 where BKAn represents a bradykinin antagonist peptide and X is a linking group which joins the two BKAn components at points intermediate to their ends.
  • the BKAn substituents may be the same or different.
  • certain heterodimers involving the linkage of a BKAn peptide and another peptide of different receptor activity through the linking group X, e.g. an N ⁇ or NK 2 antagonist peptide or a mu-opioid receptor agonist peptide. Such heterodimers are particularly useful where there is a close relationship between the activities of concern.
  • tissue and plasma derived mediators such as kinins acting at BK2 receptors
  • neuronally derived factors such as substance P (NK ⁇ _ receptors) and neurokinin A (NK2 receptors) .
  • neuronal receptors of the mu-opiate class that when stimulated can inhibit the release of the neurogenic peptides regardless of type (substance P, neurokinin A, neurokinin B, cholecystokinin, CGRP, etc.) .
  • heterodimers described in the above-noted applications are directed towards addressing these problems with single agents possessing dual selectivity. Other advantages of such heterodimers will also be appreciated by those in the art .
  • the present invention in its broadest aspect, is. concerned with heterodimers obtained by linking a BKAn peptide to another pharmacophore which is not a bradykinin antagonist, i.e. which may be either a peptide as described in WO 92/17201, or a non- peptide, effective against ' a different, non- bradykinin component responsible, for example, for pain and/or the inflammatory process, or other problems related to or occurring in concert with the activity of the kinins.
  • the resulting compounds are "dual action" compounds that are capable of interacting with two receptor populations or, alternatively, with a receptor and an enzyme.
  • the overall pharmacological effect of administering such a compound in an appropriate dose is at least the summation of the two types of activities.
  • the compounds can be designed to remain intact or they can be designed to be dissociated into two separate molecules each retaining its own identifiable activity.
  • heterodimers of the invention can be structurally represented as follows:
  • BKAn is a bradykinin antagonist peptide
  • X is a linking group
  • Y is a peptide or non-peptide pharmacophore which is not a bradykinin antagonist and demonstrates activity towards a different receptor or enzyme than the BKAn component, preferably one related to pain or the inflammatory process.
  • the present heterodimers offer the possibility of providing a wider spectrum of treatment for pain and inflammation. It is a generally held opinion that in inflammatory states, regardless of severity, the likelihood that a single agent or mediator is completely responsible for all of the clinical manifestations of the syndrome being addressed is extraordinarily small. A corollary to this is that, given the role of bradykinin in inflammatory pathophysiology, any combination therapy used in the treatment of inflammatory disorders should include bradykinin antagonism as part of its overall profile of action. Broad spectrum and potent non-specific therapies (such as the use of steroids in asthma) while perhaps efficacious, carry with them the burdens of undesired and potentially serious side effects and toxicities.
  • two discrete mediators are known to act synergistically and to account for an overwhelming proportion of the clinically important manifestations of the disease being treated.
  • substance-P acting at NK X receptors
  • bradykinin acting at BK 2 receptors
  • neutrophil elastase as one of the more important down stream effectors of inflammation
  • bradykinin as one of the more important initiating and sustaining inflammatory mediators also can be viewed as being synergistic in their actions.
  • bradykinin antagonist peptides are known in the art and any of these may be used for present purposes to provide the BKAn substituent of the present dimers .
  • One of the more potent bradykinin antagonists in vi tro is the peptide having the formula:
  • bradykinin antagonist peptide See Regoli et al, Trends in Pharmacological Science, ⁇ :156-161 (1990) .
  • This peptide is referred to herein for convenience as CP-0088. While CP-0088 is a convenient BKAn to use, those in the art will appreciate that other available or known bradykinin antagonist peptides can also be used for present purposes.
  • a wide variety of such bradykinin antagonist peptides have been disclosed in the recent patent literature and any of these can be used for present purposes. See, for example, EP-A-0334244 (Procter and Gamble) which ' discloses nona- and larger bradykinin antagonist peptides in which certain amino acid residues are modified.
  • EP-A-0370453 (Hoechst) and WO 89/01780 and WO 89/01781 (Stewart et al) also describe bradykinin antagonist peptides. None of these patent publications appears to show dimers as contemplated herein. However, as noted, the peptides of these publications can be used in the practice of the present invention. Any linking group X may be used for present purposes to chemically or covalently link together the BKAn and Y components provided this does not interfere with the activity of the components BKAn and Y.
  • the linking group may be inorganic (e.g.
  • the linking group may be such that the heterodimer remains intact when used.
  • the linking group X can include an -S- atom derived by reacting a sulfhydryl group on the BKAn peptide chain with the other pharmacophore component. This can be accomplished by reaction involving a cysteine (Cys) sulfhydryl group within the peptide chain, i.e. intermediate the ends of the peptide. This may require initially modifying the starting BKAn peptide so that it includes a Cys group in the appropriate position in the peptide chain.
  • Cys cysteine
  • CP-0088 may be modified by replacing the Ser in the 6-position with Cys (such modified CP-0088 being called CP-0126 hereinafter) to provide for convenient linking to the other pharmacophore through the Cys sulfhydryl .
  • CP-0126 can be structurally illustrated as follows :
  • the linking group X then includes the -S- of the cysteine sulfhydryl. This may be the entire linking group X (as in a disulfide based dimer) or only a part thereof.
  • the linking group may comprise a J issuccinimidoalkane such as bissuccinimidohexane joined at its end to the BKAn and Y components.
  • linking groups X can be derived from the six families of compounds listed below which can be generically categorized as amino acid analog linkers or maleimide-based linkers. These linkers are included as examples only and are not intended to be totally inclusive of all potential linking moieties:
  • ammo acid analog linkers (Class I) can be directly incorporated into the peptide chain of the BKAn and then used to form esterase stable or labile heterodimers with the geminal pharmacophore (component Y) .
  • the maleimide based linker can be reacted with the desired pharmacophore and then conjugated to a sulfhydryl containing peptide.
  • R 1 and R 2 can be varied so as to provide for completely unhindered or significantly hindered access to the carbonyl carbon of an ester based linking element so that the rate of in vivo hydrolysis of said ester can be controlled as desired.
  • Certain of the linker-modified BKAns or pharmacophores used herein to prepare the dimers of the invention are themselves novel and constitute a further aspect of the invention.
  • the component Y of the present heterodimers may be any peptide or non-peptide pharmacophore, other than a bradykinin antagonist, which demonstrates activity towards a different (non-bradykinin) component related to pain and/or the inflammatory process so as to provide dual action compounds that are capable of interacting independently with two different receptor populations or a receptor and an enzyme.
  • the component Y may be a non-peptide mu-opioid receptor agonist, e.g. morphine or one of its derivatives such as oxycodone or oxymorphone. Indomethacin is a useful choice for the Y component when cyclooxygenase inhibition (COI) is desired.
  • any of the other conventional non-steroidal anti-inflammatory agents such as aspirin, ibuprofen, napr ⁇ xyn or the like can be. used.
  • the BKAn/COI heterodimer may need to be hydrolyzed in order to obtain in vivo COI activity as cyclooxygenase is generally considered to be an intra-cellular enzyme.
  • neutrophil elastase inhibition is required, this may be an active ester, e.g. 2- phenyl-alkanoate ester.
  • a heteroaryl alkanoate esterase inhibitor A preferred neutrophil elastase inhibitor for use herein as component Y is identified below as CE- 1218. This is believed to be a new compound and constitutes a further feature of the present invention.
  • elastase inhibitors which may also be used as component Y, include fluoromethyl ketones, phosphonates, benzoxazoles, beta-lactams, etc.
  • component Y may comprise a peptide or non-peptide inhibitor having a desired activity other than bradykinin antagonist activity.
  • the Y component is preferably selected to provide activity against receptors or enzymes which have a common or close relationship to the activity of bradykinin, e.g. the treatment of pain or inflammation.
  • the rationale for using combinations of a BKAn with a mu-opioid receptor agonist, neutrophil elastase inhibitor, cyclooxygenase inhibitor or NKj or NK 2 receptor antagonist in various conditions is discussed below for purposes of illustration.
  • C-Fiber afferents are known to mediate both the sensation of pain as well as the neurogenic component of inflammation. These afferent neurons release a variety of neuropeptides in response to specific and non-specific stimuli in both the central nervous system (CNS) as well as in the peripherally innervated tissues. Some of these neuropeptides include: substance-P, neurokinin A, neurokinin B, calcitonin gene related peptide
  • CRP cholecystokinin
  • VIP vasoactive intestinal polypeptide
  • neuropeptide Y neuropeptide Y
  • C-fibers appear to contain different amounts and/or ratios of these neuropeptides depending on the tissue innervated. All of these peptides have been shown to play contributory roles in the various neurogenic processes that have been implicated in numerous diseases and clinical syndromes. In fact, specific antagonists to these peptides are being developed as potential therapeutics by a variety of pharmaceutical companies.and independent research laboratories. One apparently common feature among this otherwise diverse group of neurons is that they all have mu-opioid receptors that modulate the release of these neuropeptides.
  • Both the endogenous enkephalins as well as other exogenously administered small molecular weight compounds such as morphine, oxymorphone, fentanyl and their derivatives will inhibit the release of the neuropeptides from peripheral C-fibers by acting as mu-opioid receptor agonists locally (at terminal mu- opioid receptors in the periphery) and in the CNS. This inhibition is independent of both the constellation of peptides contained in the specific C-fiber as well as the stimulus causing their release.
  • BKAn/mu-opioid receptor agonist heterodimers are BKAn/mu-opioid receptor agonist heterodimers. These compounds would be expected to attenuate or block both the humoral component of the inflammatory process as represented by the kinins as well as the neurogenic aspects of inflammation produced by the release of the neuropeptides.
  • one of the limiting aspects of the use of existing mu opioid agonists is their propensity to produce sedation, confusion, and a depressed respiratory drive, not to mention their potential for the development of addiction and/or tolerance in the patients being treated with these agents.
  • mu-opioid receptor agonists are due to their ability to easily penetrate the CNS.
  • BKAn/mu-opoid receptor agonist heterodimers should not penetrate the CNS due to the highly cationic nature of the BKAn. Consequently, mu-opoid receptor agonist activity should be limited to the periphery and should result in a substantially reduced side effect/toxicity profile for these types of compounds.
  • BKAn/Neutrophil Elastase Inhibitor NED
  • the control of both systemic and local inflammatory responses may require interventions in more than one inflammatory pathway.
  • the ability to block the activity of a primary mediator responsible for the initiation and maintenance of the inflammatory process such as bradykinin
  • a primary ' final pathway effector responsible for actual tissue degradation and injury such as neutrophil elastase
  • Heterodimers containing combined BKAn/NEI activities can be designed to remain intact or to be dissociable as both targets (bradykinin receptors and neutrophil elastase) are extracellular in nature.
  • targets bradykinin receptors and neutrophil elastase
  • linking moieties tethering the two active components of the heterodimer can be designed to be hydrolyzed by, for example, plasma hydrolases.
  • bradykinin A large proportion of the biological activity of bradykinin is interwoven with the generation of prostaglandins. For example, much of the hyperalgesia associated with inflammatory pain appears to be dependent on the generation of certain prostaglandins both by the injured tissues and by the C-fibers themselves. In the latter case, bradykinin and substance-P appear to be the primary stimuli for these "second messengers" . The local generation of prostaglandins by the injured tissues is bradykinin independent. This interaction of peptide pro-inflammatory mediators and prostaglandins occurs in other settings as well and can also be considered a target for dual action compounds.
  • Heterodimers containing combined BKAn/COI activities may need to undergo in vivo dissociation of the respective pharmacophores as cyclooxygenase is an intracellular enzyme and functional bradykinin receptors are limited to the external plasma membrane.
  • Bradykinin and substance-P are known to act synergistically in the initiation and maintenance of the inflammatory and neurogenic components of both asthma and a variety of painful conditions.
  • bradykinin is one of the more potent, if not the most potent, agents capable of stimulating C-fiber sensory afferents that mediate peripheral pain and/or the sensation of cough and dyspnea in asthma.
  • These neurons regardless of the primary stimulus, will release substance-P which amplifies and augments the activity of bradykinin and other stimuli at the sensory nerve endings where these stimuli are acting.
  • This "one/two punch" of initial stimulus followed by local amplification is well documented and has significant implications for the success or failure of any single intervention.
  • Bradykinin's ability to produce acute bronchial smooth muscle constriction is at least partially dependent on the release of neurokinin A by the same C-fibers that release substance-P.
  • Neurokinin A exerts its effect via NK 2 receptors on the bronchial smooth muscle.
  • more than just bradykinin can release neurokinin A from these neurons and, as a result, a dually-specific antagonist with combined BK 2 /NK 2 antagonist activity should provide better overall amelioration of bronchoconstriction in the asthmatic patient than any other single agent .
  • the heterodimers •of the invention may be prepared in generally the same manner as described in WO 92/17201.
  • linking group X to the BKAn component at an appropriate position along the peptide chain followed by joining the non-peptide pharmacophore to the BKAn through the linking group.
  • the linking group may be added to the non-peptide pharmacophore and the BKAn thereafter joined to the linker-modified pharmacophore. Representative procedures are described below although it will be recognized that various modifications may be used.
  • the thiol isolated was dissolved in a minimum quantity of DMF (N,N-dime hyl formamide, 2 ml) .
  • Compound X (approximately 0.3 equiv) was dissolved in tris buffer (0.5 M, 4.0 ml) and added to the DMF- solution and then stirred for 17h.
  • the crude mixture was purified on a reverse phase Vydac C-18 HPLC column using the gradient 15 - 40% CH 3 CN in water, 0.1% constant TFA, over 20 minutes. Retention time was 16.0 minutes. 26.4 mg of II was isolated as a white powder on lyophillization.
  • the mass spectra was run on a Finnigan Lasermat Mass Analyzer.
  • Oxymorphone hydrochloride (0.56g, 1.66 mmol) , S-benzyl cysteamine (0.69g, 4.15 mmol) , acetic acid (1.52 ml, 26.5 mmol) and sodium cyanoborohydride (O.llg, 1.66 mmol) were used according to the procedure for I .
  • the composition of the third eluant, EtOAc-MeOH-Et 3 N, used in the purification of the crude mixture was 9:1:0.3, v/v/v. On purification, 0.213g of compound VII (29.0%) was isolated as an oil.
  • VII (0.063g, 0.14 mmol) was deprotected following the procedure for II. The thiol was then treated with X (0.335g, 0.152 mmol) in tris buffer. The crude mixture was purified on a reverse phase Vydac C-18 HPLC column using the gradient 15 - 70% CH 3 CN in water, 0.1% constant TFA over 35 minutes. VIII had a retention time of 15.0 minutes. 119.0 mg (45.0%) of VII was isolated as a white powder on lyophillization.
  • mice In order to test the activity of these compounds in vivo, a model of inflammatory and neurogenic pain was used. This model measures the behavioral responses of mice injected in the hind limb foot pad with 50 ul of formalin. The data from these studies are summarized in Figures 2, 3 and 4. Control mice (open circles) show a characteristic bi-phasic response to the injected formalin wherein there is a short lasting initial response followed by a quiescent period which is then followed by a sustained period of hind limb licking. The licking behavior is interpreted to mean that the limb is irritated and painful. The greater the time spent licking, the more painful the stimulus.
  • Oxymorphone (Fig. 2 A and B) reduces both phases of the licking behavior but with significant behavioral obtundation resulting in catalepsy and frank respiratory depression at the highest doses (0.9 and 3.0 umoles/kg) .
  • the bradykinin antagonist CP-0127 (a potent BK 2 selective antagonist--Fig. 3 A and B) will reduce the time spent licking in both phases of the formalin test but at doses that are substantially greater than would be practical in a clinical setting.
  • CP-0494 (Fig.
  • BKAn/mu-opioid receptor agonist heterodimers are pharmacologically qualitatively superior to either of the parent pharmacophores as would be expected from the theoretical considerations outlined above.
  • the compounds described are representative of a wide variety of compounds in which each of the components of the heterodimer (BKAn, linker and/or mu-opioid receptor agonist) can be varied to produce the optimal effect desired.
  • the linking element used in this heterodimer was chosen so as to allow for unhindered' hydrolysis of the joining ester bond by serum esterases.
  • the linker can be modified so as to provide different rates of hydrolysis varying from rapid to practically zero by altering the steric accessibility of the ester carbonyl carbon or by changing the chemistry to an amide linkage.
  • Completely stable linker moieties can also be used which are free from potential hydrolytic degradation.
  • Trifluoroacetic acid 25 mL was added to a stirred solution of 4- (3' -carbo- tert-butoxy- propylmercapto)phenyl 4- tert-butylphenylisobutrate
  • the reaction mixture was injected on a Vydac 1" C-18 reverse phase column, and eluted at lOml/min, 15%-90% acetonitrile in H20 over 35 minutes (Constant .1% TFA) .
  • the appropriate fractions were lyophilized to yield 52 mg (35%) of a colorless white powder (CP-0502)
  • Laser desorption mass spectrometry M/Z 1890 (M+H) , calculated 1890.
  • BKAn activity was assessed using the rat uterus preparation and NEI (K ⁇ 8 ) activity was evaluated using purified human neutrophil elastase (HNE) and a synthetic soluble chromogenic substrate, methoxysuccinyl-alanyl-alanyl-prolyl-valyl- paranitroaniline (MOS-AAPV-pNA) .
  • HNE human neutrophil elastase
  • MOS-AAPV-pNA methoxysuccinyl-alanyl-alanyl-prolyl-valyl- paranitroaniline
  • the inhibitor was mixed with MOS-AAPV-pNA (0.5 mM) in 0.05 M sodium phosphate, 0.1 M NaCl, 0.005% Triton X-100, 5% DMSO, pH 7.5. HNE (10-20 nM) is then added. The production of nitroaniline was monitored spectrophotometrically at a wavelength of 400-410 nm at 25 C. An ENZFITTER program then automatically calculated standard enzyme kinetic parameters including K ⁇ 5 .
  • an in vi tro "surrogate" system was employed wherein the parent heterodimer (CP- 0502) was incubated with human plasma and the resulting metabolites analyzed by reverse phase HPLC.
  • CP-0502 - was added to freshly obtained normal human plasma and allowed to incubate at 37°C for varying amounts of time. At the designated time the samples were treated with acidified (0.1 N HCl) acetonitrile in order to precipitate the plasma proteins. Aliquots (75 ul) of the supernatents were then analyzed on a Vydac C-18 reverse phase HPLC column using 24% to 80% acetonitrile gradient in 0.1% TFA. The eluent was monitored at 214 nm.
  • Figures 5 a and b are representative reverse phase HPLC chromatograms illustrative of this type of analysis.
  • the parent compound appears to be readily hydrolyzed to the succinimidohexanol modified monomer, CP-0487 and its des-Arg 9 derivative (Plasma carboxypeptidase will cleave the terminal arginine residue from both the intact heterodimer, CP-0502, as well as CP-0487.)
  • the apparent T 1/2 of this hydrolysis reaction is approximately 113 minutes .
  • the NEI component of the heterodimer is an active ester and undergoes hydrolysis as well.
  • the intact NEI monomer as well as its hydrolysis products are obscured by the plasma derived peaks seen in the middle of this tracing and cannot be visualized using this system. Since the NEI is equally active as a component of the heterodimer as it is as a monomer, the dissociation of the heterodimer into its two component parts will have less of an effect on its activity than that for the BKAn component.
  • hydrolysis rate of the heterodimer can be influenced by the steric and electronic environment of the "linking" ester moiety and that the type of chemistry used is only a single example of the types of chemistry. that can be employed to adjust the rate of dissociation (or lack thereof) of the two components of the heterodimer.
  • indomethacin (1.90 g, 0.00532 mol), 25 mL of CH 2 C1 2 and DCC (0.55 g, 0.00266 mol) . After 2 hr, the mixture filtered, the
  • CP-0126 (100 mg, 0.08 mmol) was reacted with compound (9) (0.12 mmol, 1.5 eqv) in 2 mL 95% DMF/5% 0.IM ammonium bicarbonate containing 50 ul diisopropylethylamine, for 30 minutes, with occasional mixing.
  • the reaction mixture was purified in 1 injection on a Vydac 1" C-18 " reverse phase column at lOml/min, using a gradient running from 15% acetonitrile/0.1% TFA to 40% acetonitrile/0.1% TFA in 20 minutes. Appropriate fractions were lyophilized 'to yield 64 mg (45%) of a colorless powder (CP-0460) .
  • Laser desorption mass spectrometry: M/Z 1802 (M+H) , calculated 1802.
  • CP-0460 was exposed to rat lung parenchymal strips which were then challenged with arachidonic acid.
  • This tissue is known to contain both non-specific esterase activity as well as to convert arachidonic acid to thromboxane (via a cyclooxygenase dependent pathway) which is then ultimately responsible for the smooth muscle contraction observed in this assay.
  • Intact CP-0460 was also tested for BKAn activity using the standard rat uterus assay ,and the pA 2 of the CP-0460 was found to be approximately 7.8. Again, CP-0460 (similarly to CP-0502) did not behave as a classical competitive antagonist of bradykinin induced uterine contraction but rather as a type of "pseudo-non-competitive" antagonist, particularly at higher concentrations. This atypical behavior cannot be attributed to COI activity per se as free indomethacin has no effect on this assay at any concentration.
  • pharmacologically important BKAn/COI heterodimers can be ' made using a variety of appropriate linking moieties to provide a free hydroxyl and the carboxyl group (a common feature- of many COIs) of the COI monomer to form a hydrolyzable ester based heterodimer.
  • Compounds such as these may be used in the treatment of a variety of inflammatory or painful conditions as well as in the treatment of dysfunctional uterine smooth muscle activity.
  • this component may equally comprise in whole or part a peptide as exemplified in the afore-mentioned WO 92/17201, including the heterodimers therein described.
  • the dimers of the invention may be used in the form of conventional pharmaceutical compositions comprising the active component and a pharmaceutically acceptable carrier.
  • Such compositions may be adapted for topical, oral, aerosolized, intramuscular, subcutaneous or intravenous administration.
  • the amount of active component present in such compositions will range from, for example, about 0.001 to 90.0% by weight depending on the application and mode of administration although more or less of the active component may be used.
  • Conventional dosages will vary considerably on the basis of the intended application and mode of administration. Usually, however, an effective dose is in the order of 0.1 to 1000 micrograms per kg body weight.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A bradykinin antagonist of the formula: (BKAn) (X) (Y) where BKAn is a bradykinin antagonist peptide; Y is a pharmacophore; and X is a bridging link chemically joining the BKAn and Y components.

Description

BRADYKININ ANTAGONISTS
The present invention relates to pharmaceutically effective heterodimers comprising a bradykinin antagonist (BKAn) component covalently linked to another different pharmacophore componen .
Related Applications
In prior application WO 92/17201 published October 15, 1992, there is described bradykinin antagonist dimers of the type: X(BKAn)2 where BKAn represents a bradykinin antagonist peptide and X is a linking group which joins the two BKAn components at points intermediate to their ends. The BKAn substituents may be the same or different. However, also described are certain heterodimers involving the linkage of a BKAn peptide and another peptide of different receptor activity through the linking group X, e.g. an N^ or NK2 antagonist peptide or a mu-opioid receptor agonist peptide. Such heterodimers are particularly useful where there is a close relationship between the activities of concern. Thus, it is known that in a number of pathophysiologically important processes, there is an intimate interaction of inflammatory and neurogenic mediators. This occurs, for example, in both pain secondary to tissue trauma (accidental and post-operative) as well as in asthma. In both situations, there is a complex interplay of tissue and plasma derived mediators (such as kinins acting at BK2 receptors) and neuronally derived factors such as substance P (NK^_ receptors) and neurokinin A (NK2 receptors) . There are also locally acting neuronal receptors of the mu-opiate class that when stimulated can inhibit the release of the neurogenic peptides regardless of type (substance P, neurokinin A, neurokinin B, cholecystokinin, CGRP, etc.) .
Given the interaction of these as well as other inflammatory and neurogenic mediators, no one agent is likely to be universally efficacious in ameliorating the symptoms attendant to the pathophysiσlogy. The heterodimers described in the above-noted applications are directed towards addressing these problems with single agents possessing dual selectivity. Other advantages of such heterodimers will also be appreciated by those in the art .
Brief Description of the Invention The present invention, in its broadest aspect, is. concerned with heterodimers obtained by linking a BKAn peptide to another pharmacophore which is not a bradykinin antagonist, i.e. which may be either a peptide as described in WO 92/17201, or a non- peptide, effective against' a different, non- bradykinin component responsible, for example, for pain and/or the inflammatory process, or other problems related to or occurring in concert with the activity of the kinins. The resulting compounds are "dual action" compounds that are capable of interacting with two receptor populations or, alternatively, with a receptor and an enzyme. This is not intended to suggest that the single molecule will engage two receptors or a receptor and an enzyme simultaneously; only that the molecule is capable of interacting with either one of two receptor types or with a single class of receptors and/or an enzyme. The overall pharmacological effect of administering such a compound in an appropriate dose, however, is at least the summation of the two types of activities. The compounds can be designed to remain intact or they can be designed to be dissociated into two separate molecules each retaining its own identifiable activity.
The heterodimers of the invention can be structurally represented as follows:
(Y) (X) (BKAn) where BKAn is a bradykinin antagonist peptide; X is a linking group and Y is a peptide or non-peptide pharmacophore which is not a bradykinin antagonist and demonstrates activity towards a different receptor or enzyme than the BKAn component, preferably one related to pain or the inflammatory process.
The present heterodimers offer the possibility of providing a wider spectrum of treatment for pain and inflammation. It is a generally held opinion that in inflammatory states, regardless of severity, the likelihood that a single agent or mediator is completely responsible for all of the clinical manifestations of the syndrome being addressed is extraordinarily small. A corollary to this is that, given the role of bradykinin in inflammatory pathophysiology, any combination therapy used in the treatment of inflammatory disorders should include bradykinin antagonism as part of its overall profile of action. Broad spectrum and potent non-specific therapies (such as the use of steroids in asthma) while perhaps efficacious, carry with them the burdens of undesired and potentially serious side effects and toxicities.
In many cases, two discrete mediators are known to act synergistically and to account for an overwhelming proportion of the clinically important manifestations of the disease being treated. Such is the case, for example, with substance-P acting at NKX receptors and bradykinin acting at BK2 receptors in the contexts of asthma and post-traumatic or post-operative pain. Similarly, neutrophil elastase as one of the more important down stream effectors of inflammation and bradykinin as one of the more important initiating and sustaining inflammatory mediators also can be viewed as being synergistic in their actions.
The concept of providing homodimers of pharmaceutically active materials to improve such characteristics as metabolic stability, selectivity and receptor binding has previously been described for other systems. This prior work has included the dimerization of peptide agonists and antagonists in order to increase potency and/or duration of action. See, Caporale et al, Proc. 10th American Peptide Svmp. , Pierce Chemical Co., Rockford, IL 449-451 (1988) and Rosenblatt et al, European Patent
Application No. EP 293130A2. Thus, dimerization of peptide agonists has been disclosed for enkephalins/endorphins (Shimohigashi, Y., et al, BBRC. 146, 1109-1115, 1987) ; substance P (Higuchi, Y., et al, E.J.P.. 160, 413-416, 1989); bradykinin (Vavrek, R. and Stewart, J., J. Proc. 8th Amer. Pept . Svm .. 381-384, 1983) ; neurokinin A & B, (Kodama, H., et al, E.J.P. , 151, 317-320, 1988) ; insulin (Roth, R.A., et al, FEBS. 170, 360-364, 1984) and atrial natriuretic peptide (Chino, N., et al, BBRC, 141, 665-672, 1986) . Dimerization of antagonists has been shown for parathyroid hormone (Caproale, L.H., et al, Proc. 10th Amer. Pept. Svmp. , 449-451, 1987)) . However, the literature has not disclosed heterodimers comprised of a bradykinin antagonist and a different pharmacophore as contemplated herein.
Detailed Description of the Invention
Numerous bradykinin antagonist peptides are known in the art and any of these may be used for present purposes to provide the BKAn substituent of the present dimers . One of the more potent bradykinin antagonists in vi tro is the peptide having the formula:
D-ARG°-Arg1-Pro2-Hyp3-Gly -Phe5-Ser6-D-Phe7-Leu8- Arg9
See Regoli et al, Trends in Pharmacological Science, ϋ:156-161 (1990) . This peptide is referred to herein for convenience as CP-0088. While CP-0088 is a convenient BKAn to use, those in the art will appreciate that other available or known bradykinin antagonist peptides can also be used for present purposes. A wide variety of such bradykinin antagonist peptides have been disclosed in the recent patent literature and any of these can be used for present purposes. See, for example, EP-A-0334244 (Procter and Gamble) which 'discloses nona- and larger bradykinin antagonist peptides in which certain amino acid residues are modified. EP-A-0370453 (Hoechst) and WO 89/01780 and WO 89/01781 (Stewart et al) also describe bradykinin antagonist peptides. None of these patent publications appears to show dimers as contemplated herein. However, as noted, the peptides of these publications can be used in the practice of the present invention. Any linking group X may be used for present purposes to chemically or covalently link together the BKAn and Y components provided this does not interfere with the activity of the components BKAn and Y. The linking group may be inorganic (e.g. -S- ) or organic and may be selected so as to hydrolyze or otherwise dissociate in order to liberate the two active components BKAn and Y in vivo . Alternatively, the linking group may be such that the heterodimer remains intact when used.
Conveniently the linking group X can include an -S- atom derived by reacting a sulfhydryl group on the BKAn peptide chain with the other pharmacophore component. This can be accomplished by reaction involving a cysteine (Cys) sulfhydryl group within the peptide chain, i.e. intermediate the ends of the peptide. This may require initially modifying the starting BKAn peptide so that it includes a Cys group in the appropriate position in the peptide chain. For example, CP-0088 may be modified by replacing the Ser in the 6-position with Cys (such modified CP-0088 being called CP-0126 hereinafter) to provide for convenient linking to the other pharmacophore through the Cys sulfhydryl . CP-0126 can be structurally illustrated as follows :
In abbreviated fashion, the formula may be stated as :
DR-R-P-J-G-F-C-DF-L-R Using Cys as the position of attachment, the linking group X then includes the -S- of the cysteine sulfhydryl. This may be the entire linking group X (as in a disulfide based dimer) or only a part thereof. Thus, for example, the linking group may comprise a J issuccinimidoalkane such as bissuccinimidohexane joined at its end to the BKAn and Y components. These and other linking groups are disclosed in applicant's related application and any of these may be used for present purposes.
Other linking groups X, some of which do not require 'or contain an -S- atom, can be derived from the six families of compounds listed below which can be generically categorized as amino acid analog linkers or maleimide-based linkers. These linkers are included as examples only and are not intended to be totally inclusive of all potential linking moieties:
CLASS I (Amino Acid Analogs)
"D" or "L" Configur
D) NH2 O R, _
* C, 1H(CH2)y N r N (CH2)X — \ c / 2
CO2H O R3
CLASS II (Maleimide Based Linkers)
The ammo acid analog linkers (Class I) can be directly incorporated into the peptide chain of the BKAn and then used to form esterase stable or labile heterodimers with the geminal pharmacophore (component Y) . Alternatively, the maleimide based linker can be reacted with the desired pharmacophore and then conjugated to a sulfhydryl containing peptide. Finally, linkers from any of these families of compounds which contain -C02H as the R3 moiety can be reacted with another linker from these classes of compounds to form esterase labile (R3= -OH containing linkers) or esterase stable (R3= -NH2 containing linkers) which can then be used to form the desired peptide/non-peptide heterodimer. R1 and R2 can be varied so as to provide for completely unhindered or significantly hindered access to the carbonyl carbon of an ester based linking element so that the rate of in vivo hydrolysis of said ester can be controlled as desired. Certain of the linker-modified BKAns or pharmacophores used herein to prepare the dimers of the invention are themselves novel and constitute a further aspect of the invention.
The component Y of the present heterodimers may be any peptide or non-peptide pharmacophore, other than a bradykinin antagonist, which demonstrates activity towards a different (non-bradykinin) component related to pain and/or the inflammatory process so as to provide dual action compounds that are capable of interacting independently with two different receptor populations or a receptor and an enzyme. Thus, for example, the component Y may be a non-peptide mu-opioid receptor agonist, e.g. morphine or one of its derivatives such as oxycodone or oxymorphone. Indomethacin is a useful choice for the Y component when cyclooxygenase inhibition (COI) is desired. However, any of the other conventional non-steroidal anti-inflammatory agents, such as aspirin, ibuprofen, naprσxyn or the like can be. used. In this case, the BKAn/COI heterodimer may need to be hydrolyzed in order to obtain in vivo COI activity as cyclooxygenase is generally considered to be an intra-cellular enzyme. Where neutrophil elastase inhibition is required, this may be an active ester, e.g. 2- phenyl-alkanoate ester. There may also be used a heteroaryl alkanoate esterase inhibitor. A preferred neutrophil elastase inhibitor for use herein as component Y is identified below as CE- 1218. This is believed to be a new compound and constitutes a further feature of the present invention.
Other types of elastase inhibitors which may also be used as component Y, include fluoromethyl ketones, phosphonates, benzoxazoles, beta-lactams, etc.
As noted earlier, component Y may comprise a peptide or non-peptide inhibitor having a desired activity other than bradykinin antagonist activity. However, the Y component is preferably selected to provide activity against receptors or enzymes which have a common or close relationship to the activity of bradykinin, e.g. the treatment of pain or inflammation. The rationale for using combinations of a BKAn with a mu-opioid receptor agonist, neutrophil elastase inhibitor, cyclooxygenase inhibitor or NKj or NK2 receptor antagonist in various conditions is discussed below for purposes of illustration. BKAn/mu-opioid receptor agonists
C-Fiber afferents are known to mediate both the sensation of pain as well as the neurogenic component of inflammation. These afferent neurons release a variety of neuropeptides in response to specific and non-specific stimuli in both the central nervous system (CNS) as well as in the peripherally innervated tissues. Some of these neuropeptides include: substance-P, neurokinin A, neurokinin B, calcitonin gene related peptide
(CGRP) , cholecystokinin (CCK) , vasoactive intestinal polypeptide (VIP) , and neuropeptide Y, among other neurotransmitters. To add to this complexity, different C-fibers appear to contain different amounts and/or ratios of these neuropeptides depending on the tissue innervated. All of these peptides have been shown to play contributory roles in the various neurogenic processes that have been implicated in numerous diseases and clinical syndromes. In fact, specific antagonists to these peptides are being developed as potential therapeutics by a variety of pharmaceutical companies.and independent research laboratories. One apparently common feature among this otherwise diverse group of neurons is that they all have mu-opioid receptors that modulate the release of these neuropeptides. Both the endogenous enkephalins as well as other exogenously administered small molecular weight compounds such as morphine, oxymorphone, fentanyl and their derivatives will inhibit the release of the neuropeptides from peripheral C-fibers by acting as mu-opioid receptor agonists locally (at terminal mu- opioid receptors in the periphery) and in the CNS. This inhibition is independent of both the constellation of peptides contained in the specific C-fiber as well as the stimulus causing their release.
As a result, one important class of compounds considered to have a particularly good profile of activities for the treatment of conditions that are produced by combined humoral and neurogenic processes are BKAn/mu-opioid receptor agonist heterodimers. These compounds would be expected to attenuate or block both the humoral component of the inflammatory process as represented by the kinins as well as the neurogenic aspects of inflammation produced by the release of the neuropeptides. In addition, one of the limiting aspects of the use of existing mu opioid agonists is their propensity to produce sedation, confusion, and a depressed respiratory drive, not to mention their potential for the development of addiction and/or tolerance in the patients being treated with these agents. These undesirable aspects of mu-opioid receptor agonists are due to their ability to easily penetrate the CNS. BKAn/mu-opoid receptor agonist heterodimers, however, should not penetrate the CNS due to the highly cationic nature of the BKAn. Consequently, mu-opoid receptor agonist activity should be limited to the periphery and should result in a substantially reduced side effect/toxicity profile for these types of compounds.
BKAn/Neutrophil Elastase Inhibitor (NED As previously mentioned the control of both systemic and local inflammatory responses may require interventions in more than one inflammatory pathway. In particular, the ability to block the activity of a primary mediator responsible for the initiation and maintenance of the inflammatory process (such as bradykinin) and a primary' final pathway effector responsible for actual tissue degradation and injury (such as neutrophil elastase) may be a key to "single drug therapy" of sepsis or other severe inflammatory conditions requiring parenteral therapy or for the treatment of inflammatory dermatologic or dental/periodontal conditions. Heterodimers containing combined BKAn/NEI activities can be designed to remain intact or to be dissociable as both targets (bradykinin receptors and neutrophil elastase) are extracellular in nature. However, should dissociation of the two active pharmacophores be desired, linking moieties tethering the two active components of the heterodimer can be designed to be hydrolyzed by, for example, plasma hydrolases. These types of dissociable or hydrolyzable heterodimers are discussed herein.
BKAn/Cycloox genase Inhibitor (COI)
A large proportion of the biological activity of bradykinin is interwoven with the generation of prostaglandins. For example, much of the hyperalgesia associated with inflammatory pain appears to be dependent on the generation of certain prostaglandins both by the injured tissues and by the C-fibers themselves. In the latter case, bradykinin and substance-P appear to be the primary stimuli for these "second messengers" . The local generation of prostaglandins by the injured tissues is bradykinin independent. This interaction of peptide pro-inflammatory mediators and prostaglandins occurs in other settings as well and can also be considered a target for dual action compounds. Heterodimers containing combined BKAn/COI activities may need to undergo in vivo dissociation of the respective pharmacophores as cyclooxygenase is an intracellular enzyme and functional bradykinin receptors are limited to the external plasma membrane.
BKAn/NK1-Receptor Antagonist (NKlAn)
Bradykinin and substance-P are known to act synergistically in the initiation and maintenance of the inflammatory and neurogenic components of both asthma and a variety of painful conditions. In both of these situations, bradykinin is one of the more potent, if not the most potent, agents capable of stimulating C-fiber sensory afferents that mediate peripheral pain and/or the sensation of cough and dyspnea in asthma. These neurons, regardless of the primary stimulus, will release substance-P which amplifies and augments the activity of bradykinin and other stimuli at the sensory nerve endings where these stimuli are acting. This "one/two punch" of initial stimulus followed by local amplification is well documented and has significant implications for the success or failure of any single intervention. By targeting both components of these processes with a single compound, it is possible to provide a dually-specific agent which is superior than mono- specific agents used alone and both easier and cheaper to use than combination therapies.
BKAn/NK, Antagonist (NK2An)
Bradykinin's ability to produce acute bronchial smooth muscle constriction is at least partially dependent on the release of neurokinin A by the same C-fibers that release substance-P. Neurokinin A exerts its effect via NK2 receptors on the bronchial smooth muscle. However, more than just bradykinin can release neurokinin A from these neurons and, as a result, a dually-specific antagonist with combined BK2/NK2 antagonist activity should provide better overall amelioration of bronchoconstriction in the asthmatic patient than any other single agent . The heterodimers •of the invention may be prepared in generally the same manner as described in WO 92/17201. Normally this involves adding the linking group X to the BKAn component at an appropriate position along the peptide chain followed by joining the non-peptide pharmacophore to the BKAn through the linking group. Alternatively, the linking group may be added to the non-peptide pharmacophore and the BKAn thereafter joined to the linker-modified pharmacophore. Representative procedures are described below although it will be recognized that various modifications may be used.
The invention is illustrated but not limited by the following examples :
Examples 1-4 (BKAn/mu-opioid agonist)
Four different peptide/opiate heterodimers '(designated CP-0477, CP-0488, CP-0494 and CP-0499) were made in order to illustrate the invention. Three of these compounds were made using CP-0126 (DR-R-P-J-G-F-C-DF-L-R) and the fourth used CP-0347 (DR-R-P-J-G-Thi-C-DTic-Oic-R) . Similarly, two different opiates (oxycodone and oxymorphone) and two different linker chemistries were used to provide the respective heterodimers as follows:
The heterodimers CP-0477, CP-0488, CP-0494 and CP-0499 were prepared as detailed hereinafter with reference to the accompanying Figure 1 :
Preparation of Compound I: Oxycodone hydrochloride (0.182g, 0.52 mmol) , acetic acid (0.475 ml, 8.3 mmol) , S-benzyl cysteamine (0.174g, 1.04 mmol) and methanol (5 ml) were combined and stirred at room temperature for an . hour. Sodium cyanoborohydride (95%, 0.033g, 0.52 mmol) was added, and the reaction stirred at room temperature for 24 h. The mixture was concentrated in vacuo. The resulting oil was dissolved in ethyl acetate and the ethyl acetate fraction was washed with saturated sodium bicarbonate solution, dried over magnesium sulfate and evaporated in vacuo. The crude material was chromatographed on a silica column and eluted with EtOAc, EtOAc-MeOH (9:1, v/v) and EtOAcMeOH-Et3N (9:1:0.2, v/v/v) successively Compound I was isolated as an oil in 25.0% (59.0 mg) yield.
Preparation of Compound II (CP-0477) :
I (0.059g, 0.127 mmol) was dissolved in 2 mL dry tetrahydrofuran, and was transferred to a oven dried three-necked 100 ml flask. The flask was ' fitted with a dewar condensor, a nitrogen source and an ammonia inlet. Approximately 10 ml of ammonia was condensed into the flask maintained at -78C. Small pieces of sodium were added until the intense blue color was maintained and then quenched after 40 seconds with solid ammonium chloride. The reaction mixture was allowed to warm to room temperature and the ammonia boiled off through a bubbler, methanol (25 ml x 3) was added and evaporated in vacuo. The thiol isolated was dissolved in a minimum quantity of DMF (N,N-dime hyl formamide, 2 ml) . Compound X (approximately 0.3 equiv) was dissolved in tris buffer (0.5 M, 4.0 ml) and added to the DMF- solution and then stirred for 17h. The crude mixture was purified on a reverse phase Vydac C-18 HPLC column using the gradient 15 - 40% CH3CN in water, 0.1% constant TFA, over 20 minutes. Retention time was 16.0 minutes. 26.4 mg of II was isolated as a white powder on lyophillization.
Analysis :
The mass spectra was run on a Finnigan Lasermat Mass Analyzer.
calulated molecular weight--1916 observed molecular weight--1918
Amino Acid Analysis:
Gly 1.02 (1) , Arg 3.14 (3) , Pro 1.01 (1) , Leu 0.97 (1) , Phe 1.92 (2) and Hyp 0.94 (1) .
Preparation of Compound III:
To the mixture of oxycodone hydrochloride (1.0 g, 2.84 mmol) and ammonium acetate (2.2 g, 28.4 mmol) dissolved in methanol (10.0 ml) was added a methanolic (4.0 ml) solution of NaCNBH3 (0 18 g, 2.84 mmol) . The resulting solution was adjusted to pH 7.0 with concentrated hydrochloric acid, stirred for 17h, and acidified to pH 1.0 with concentrated hydrochloric acid. The solvent was removed in vacuo and the remaining material was dissolved in water. The aqueous layer was extracted with chloroform, adjusted to pH 9.0 with 10% sodium carbonate solution, saturated with NaCl and extracted with chloroform. The chloroform layer was dried over magnesium sulfate and evaporated in vacuo. The crude oil was purified by silica gel chromatography and eluted with EtOAc, EtOAc-MeOH (9:1, v/v) , EtOAc-
MeOH-Et3N (9:1:0.3, v/v/v) successively. Compound III was isolated as an oil in 47 0% (0.42g) yield.
Preparation of Compound IV:
BOC-Glycine (0.16g, 0.91 mmol), HOBt (0.125g, 0.91 mmol) and 1- (3-dimethylaminopropyl)
-3-ethylcarbodiimide hydrochloride (EDO ( 98 0%, 0.18g, 0.91 mmol) were dissolved in DMF (2.0 ml) and stirred at 0°C for an hour. The amine III (0.24g, 0.76 mmol) dissolved in DMF (3.0 ml) was added to the reaction mixture, the reaction mixture was warmed to room temperature and stirred for 17h. DMF was removed in vacuo and the resulting material was dissolved in ethyl acetate. The ethyl acetate layer was washed with saturated sodium bicarbonate solution, brine and dried over magnesium sulfate. The organic layer was evaporated in vacuo and the crude mixture was flash chromatograhed on a silica gel column and eluted with EtOAc-MeOH-Et3N (9.5:0.5:0.3, v/v/v) . Compound IV was isolated as an oil in 82.0% (0.29g) yield.
Preparation of Compound V:
The BOC protecting group was removed off the compound IV with TFA (5.0 ml) in methylene chloride (5.0 ml) . Methylene chloride was removed in vacuo and the residue was stripped with methylene chloride (20 ml x 3) and then with triethyl amine (3 ml x 3) . 3-S-benzyl mercapto propionic acid (0.15g, 0.75 mmol) , EDC (0.15g, 0.75 mmol) , HOBt (0.103g, 0.75 mmol) and Et3N (0.35 ml, 2.48 mmol)were dissolved in DMF (5.0 ml) and stirred at 0°C for an hour. The solution of the amine (0.23g, 0.62 mmol) in DMF (3.0- ml) was added to the reaction mixture. The reaction mixture was warmed to room temperature and stirred for 17h. DMF was evaporated in vacuo and the residue was dissolved in EtOAc. The EtOAc layer was washed with 10% Na2C03, brine, dried (over MgSO and evaporated in vacuo. The crude material was purified on a flash silica gel column and eluted with
EtOAc-MeOH-Et3N (9:1:0.3, v/v/v) . Compound V was isolated as an oil in 60.0% (0.205g) yield.
Preparation of Compound VI (CP-0488) :
32.0 mg (0.057 mmol) of V was deprotected using the procedure described for II and the thiol isolated was reacted with compound X (0.073g, 0.048 mmol) in tris buffer. The crude mixture was purified using the procedure for II. Retention time of the product was 16.82 minutes. 9.5 mg (10%) of VI was obtained as a white powder on lyophillization.
Mass spectral Data: calulated molecular weight 2002 observed molecular weight 2004
Amino Acid Analysis : Gly 1.76 (2) , Arg 3.19 (3) , Pro 1.06 (1) , Leu 0.99 (1) , Phe 2.06 (2) , Hyp 0.95 (1) . Preparation of Compound VII:
Oxymorphone hydrochloride (0.56g, 1.66 mmol) , S-benzyl cysteamine (0.69g, 4.15 mmol) , acetic acid (1.52 ml, 26.5 mmol) and sodium cyanoborohydride (O.llg, 1.66 mmol) were used according to the procedure for I . The composition of the third eluant, EtOAc-MeOH-Et3N, used in the purification of the crude mixture was 9:1:0.3, v/v/v. On purification, 0.213g of compound VII (29.0%) was isolated as an oil.
Preparation of Compound VIII (CP-0494) :
VII (0.063g, 0.14 mmol) was deprotected following the procedure for II. The thiol was then treated with X (0.335g, 0.152 mmol) in tris buffer. The crude mixture was purified on a reverse phase Vydac C-18 HPLC column using the gradient 15 - 70% CH3CN in water, 0.1% constant TFA over 35 minutes. VIII had a retention time of 15.0 minutes. 119.0 mg (45.0%) of VII was isolated as a white powder on lyophillization.
Mass Spectral Data: calculated molecular weight 1902 observed molecular weight 1904
Amino Acid Analysis: Gly 0.81 (1) , Arg 3.12 (3), Pro 1.07 (1), Leu 0.99 (1) , Phe 2.04 (2), Hyp 0.98 (1) .
Preparation of Compound IX (CP-0499) :
VII (0.009g, 0.02 mmol) was deprotected following the procedure for II and the thiol was then reacted with XI (0.026g, 0.016 mmol) in tris buffer. Crude mixture was purified on a Vydac C-18 reverse phase HPLC using the gradient 15 - 70% CH3CN in water, 0.1% constant TFA, flow rate of 8.0 ml/min over 40 minutes. Retention time of IX was 14.22 minutes. IX (6.4 mg, 20.0%) was isolated as white powder on lyophillization.
Mass Spectral Data: Calculatedmolecularweight 1957 Observed molecular weight 1958
In Vi tro Testing The BKAn/mu-opioid receptor agonist heterodimers were evaluated in vi tro using the rat uterus (BK2-receptor activity) and the electrically stimulated guinea pig ileum (mu-opiate receptor activity) assays. These assays are well known in the art. The results obtained are shown in Table I
TABLE I Compound pA2-Rat Uterus IC50 Guinea Pig Ileum (nmolar) inactive inactive inactive
21.7 inactive inactive
24.0 17.0
It should be noted that neither oxycodone nor the heterodimers derived from oxycodone (CP-0477 and CP-0488) showed any activity' in the in vi tro guinea pig ileum assay of mu-opiate receptor agonist activity. This is probably due to the fact that for complete activity, oxycodone apparently needs to be demethylated in vivo . As a result, oxycodone and oxycodone-based compounds would not be expected to show activity in an assay wherein the appropriate demethylating enzymes were missing.
More important, however, are the data regarding the activity of the BKAn component of these heterodimers on the rat uterus and the data regarding the activity of the oxymorphone containing compounds. As can be seen from the data outlined in Table I, full BKAn activity was retained in all of these heterodimers and in those compounds utilizing oxymorphone as the opiate, full mu-opiate receptor agonist activity was also retained. From these data, it is evident that BKAn/mu-opioid receptor agonist heterodimers can interact with their respective receptor populations in in vi tro systems.
In Vivo Testing
In order to test the activity of these compounds in vivo, a model of inflammatory and neurogenic pain was used. This model measures the behavioral responses of mice injected in the hind limb foot pad with 50 ul of formalin. The data from these studies are summarized in Figures 2, 3 and 4. Control mice (open circles) show a characteristic bi-phasic response to the injected formalin wherein there is a short lasting initial response followed by a quiescent period which is then followed by a sustained period of hind limb licking. The licking behavior is interpreted to mean that the limb is irritated and painful. The greater the time spent licking, the more painful the stimulus.
Oxymorphone (Fig. 2 A and B) reduces both phases of the licking behavior but with significant behavioral obtundation resulting in catalepsy and frank respiratory depression at the highest doses (0.9 and 3.0 umoles/kg) . The bradykinin antagonist CP-0127 (a potent BK2 selective antagonist--Fig. 3 A and B) will reduce the time spent licking in both phases of the formalin test but at doses that are substantially greater than would be practical in a clinical setting. CP-0494 (Fig. 4 A and B) , however, not only blocks both phases of the pain response, but does so at doses substantially lower (0.1 umoles/kg) than for either oxymorphone (0.9 umoles/kg) or CP-0127 (12.6 umoles/kg) alone and, of equal or greater importance, with no observable narcotic effects over several hours. These data indicate that BKAn/mu-opioid receptor agonist heterodimers are pharmacologically qualitatively superior to either of the parent pharmacophores as would be expected from the theoretical considerations outlined above. One skilled in the art will appreciate that the compounds described are representative of a wide variety of compounds in which each of the components of the heterodimer (BKAn, linker and/or mu-opioid receptor agonist) can be varied to produce the optimal effect desired.
Example 5 (BKAn/NEI)
A BKAn/NEI type of compound (CP-0502) of the structure shown in Synthetic Scheme 3, was synthesized to illustrate that this class of compounds can be used as a potent topical and/or systemic anti-inflammatory agent. This compound is derived from CP-0126 and the prototype elastase inhibitor, CE-1218 (see Compound (6) , Synthetic Scheme 1 below) . SYNTHETIC SCHEME 1
THF. NaH Mel
H2θ2, AcOH
The linking element used in this heterodimer was chosen so as to allow for unhindered' hydrolysis of the joining ester bond by serum esterases. Those skilled in the art will recognize that the linker can be modified so as to provide different rates of hydrolysis varying from rapid to practically zero by altering the steric accessibility of the ester carbonyl carbon or by changing the chemistry to an amide linkage. Completely stable linker moieties can also be used which are free from potential hydrolytic degradation.
Synthesis and analysis of BKAn/NEI heterodimer(s)
The synthesis of these compounds is illustrated by reference to Synthetic Schemes 2 and 3 and the following detailed synthesis description which includes the preparation of the elastase inhibitor CE-1218 according to Synthetic Scheme 1:
SYNTHETIC SCHEME 1:
Synthesis of 4-tert-Butylacetophenone (1) To a dry 1-L flask was added CS2 (250 mL) and A1C13 (133.34 g, 0.56 mol) with .stirring. The suspension was cooled in an ice bath and a solution of tert-butylbenzene (50.00 g, 0.37 mol) and acetyl chloride (78.50 g, 0.41 mol) was added dropwise over 2 hr (not allowing the temperature to rise above 25°C) . The reaction was allowed to stir at room temperature overnight and then poured into a 2 L beaker filled with ice. After quenching with 200 mL of 6 N HCl the solution was saturated with NaCl and separated. The aqueous layer was washed with ether (2 x 100 mL) and combined with previous organics . This new organic solution was washed with water (100 mL) , dried (MgS04) and evaporated to give an oil which was distilled to give 52.1 g (79.3%) of 4- ϋert-acetophenone as a clear colorless oil (bp005 mm 70-76°C) . "H NMR (CDC13) δ 1.35 (s, 9 H) , 2.58 (s, 3 H) , 7.48 (d, J = 8.5 Hz, 2 H) , 7.91 (d, J = 8.5 Hz, 2 H) . 13C NMR (CDC13) <5 26.42, 30.96, 34.95, 125.36, 128.16, 134.49, 156.64, 197.61.
Synthesis of Methyl 4-tert-butylphenylacetate (2)
A dry 1 L flask equipped with a mechanical stirrer containing Pb(OAc)4 (132.06 g, 0.298 mol) and 250 mL of benzene was purged with nitrogen and cooled in an ice bath. To this cooled slurry was added dropwise a solution of BF30Et2 (137.8 mL, 1.12 mol) , 4-tert-butylacetophenone (50.00 g, 0.284 mol) in 70 mL of methanol over 1 hr. This mixture was allowed to stir overnight, quench with water (500 mL) , diluted with 250 mL ether and the layers separated. The organic layer was washed with water, diluted NaHC03 (carefully) and dried over MgSO„ . The mixture was filtered, evaporated and distilled to give 31.2 g (53.4%) of methyl 4-tert- butylphenylacetate as clear colorless oil (bp004 mm 75-80°C) .. :H NMR (CDCl3) 1.32 (s, 9 H) , 3.62 (s, 2 H) , 3.71 (s, 3 H) , 7.23 (d, J = 8.4 Hz, 2 H) , 7.37 (d, J = 8.4 Hz, 2 H) . 13C (CDC13) 31.33, 34.46, 40.67, 52.04, 125.53, 128.88, 130.91, 149.94, 172.26.
Synthesis of Methyl 4- tert-butylphenylisobutyrate
121
A solution of methyl 4- tert-butylphenylacetate (30.00 g, 0.145 mol) and iodomethane (45.41 g, 0.320 mol) in 125 mL of dry THF was added to a slurry of NaH (8.72 g, 0.363 mol) in 200 mL of THF dropwise over 30 minutes. After completion of the addition, the reaction mixture was heated at reflux for 1.5 hr. The reaction was allowed to cool to room temperature, filtered through Celite and concentrated. The residue was diluted in ether, washed with H20, and dried over MgS04. Evaporation of the solvent afforded the desired product as an oil. A mixture of the crude methyl 4-tert- butylphenylisobutyrate and 4:1 EtOH/H20 containing KOH (10.07 g, 0.179 mol) were heated to reflux for 4 hr. The EtOH was evaporated in vacuo, the residual solution was acidified to pH 2 with 2 N HCl, and the precipitated solid filtered. The white solid was the dried (60°C, 1 mm Hg, 24 hr) to give the desired product (23.45 g, 73.2% from methyl 4-tert- butylphenylacetate).. *H NMR (CDC13) 1.34 (s, 9 H) , 1.62 (s, 6 H) , 7.37 (s, 4 H) , 11.4-12.4 (brs, 1 H) .
13 C. NMR (CDC13) 26.16, 31.30, 34.35, 45.81, 125.31,
125.48, 140.64, 149.66, 183.57.
Synthesis of 4- (3' -carbo-tert-butoxy-propyl mercapto)phenyl 4 -tert-butylphenylisobutyrate (4) : A mixture of 4-tert-butylphenylisobutyric acid (2.00g, 0.0091mol) and thionyl chloride (1.62 g, 0.0136 mol) in 16 ml of CH2C12 was allowed to stir overnight under Argon. The volatiles were removed under vacuum and the resulting solid was dissolved into THF (15 mL) and a solution of tert-butyl-4- (4' - hydroxyphenyDmercaptobutyrate (2.44 g, 0.0091 mol) , TEA (2.5 mL) in THF (15 L) was added dropwise over 10 min. The mixture was stirred for 3 days, diluted with Et20 and extracted with 5% NaHC03. The organics were washed with H20, brine and dried (MgS04) . After evaporization the colored oil was separated (HPLC, silica gel 70:30 CH2Cl2/hexane to CH2C12 linear graduent) to give the desired product as an oil (2.20 g, 51.5%) . lU NMR (CDC13) δ 1.33 (s, 9 H) , 1.43 (s, 9 H) , 1.70 (s, 6 H) , 1.88 (tt, J = 7.2 Hz, 2 H) , 2.35 (t, J = 7.2 Hz, 2 H) , 2.90 (t, J = 7.2, 2 H) , 6.92 (d, J = 8.6 Hz, 2 H) , 7.32 (d, J = 8.6 Hz, 2 H) , 7.34 - 7.40 (m, 4 H) . 13C NMR (CDC13) δ 24.50,
26.42, 28.08, 31.31, 33.70, 34.11, 34.39, 46.40, 80.40, 121.92, 125.23, 125.46, 130.82, 133.03, 140.86, 149.58, 149.69, 172.21, 175.34.
Synthesis of 4- (3 ' -carboxy-propylmercapto)phenyl 4-tert-butylphenylisobutyrate (5)
Trifluoroacetic acid (25 mL) was added to a stirred solution of 4- (3' -carbo- tert-butoxy- propylmercapto)phenyl 4- tert-butylphenylisobutrate
(2.40 g, 0.00510 mol) in 20 mL of CH2C12 over 15 min. After an additional 1.5 min the volatiles were removed and the oil crystallized (hexane) to give 1.94 g (91.8%) of desired product as a white solid, m.p. 86.0-87.0°C, 1H(CDC13) 1.33 (s, 9 H) , 1.70 (s, 6 H) , 1.92 (tt, J = 7.0 Hz, 2 H) , 2.50 (C, J = 7.0 Hz, 2 H) , 2.93 (t, J = 7.0 Hz, 2 H), 6.93 (d, J = 8.7
Hz, 2 H) , 7.33 (d, J = 8.7 Hz, 2 H) , 7.35-7.39 (m, 4 H) . 13C NMR (CDC13) 23.89, 26.45, 31.32, 32.34, 33.63, 34.42, 46.42, 122.03, 125.24, 125.48, 131.12, 132.63, 140.85, 149.74, 175.40, 178.65.
Synthesis of 4- (3 ' -carboxy-propylsulfonyl)phenyl
4-tert-butylphenylisobutyrate (6)
To a 50 mL flask was added 4- (3 ' -carboxy- propylsulfonyl)phenyl 4- tert-butylphenylisobutyrate (1.64 g, 0.00396 mol), HOAc (25 mL) and 15 mL of 30% H202. The reaction was allowed to stir overnight, diluted with H20 (50 mL) and the resulting solid filtered. After drying (12 hr, 1 mmHg) the solid was recrystallized (CH2Cl2/hexane) to give 1.54 g (87.1%) of the desired product as a white powder, p 107-108.5°C. *H (CDC13) 1.33 (s, 9 H) , .172. (s, 6 H) , 2.02 (p, J = 7.0 Hz, 2 H) , 2.52 (t, J = 7.0 Hz, 2 H) , 3.17 (t, J = 7.0 Hz, 2 H) , 7.20 (d, J = 8.7 Hz, 2 H) , 7.36 (d, J = 8.7 Hz, 2 H) , 7.41 (d, J = 8.6 Hz, 2 H) , 7.90 (d, J = 8.6 Hz, 2 H) . 13C NMR (CDC13) 17.88, 26.29, 31.28, 31.79, 34.42, 46.53, 55.04, 122.56, 125.16, 125.61, 129.70, 135.81, 140.22, 150.02, 155.29, 174.73, 177.71.
Synthesis of 6-Maleimidohexanol (7) :
The synthesis of 6-maleimidohexanol to be used for linking is illustrated in Synthetic Scheme 2 below:
SYNTHETICSCHEME2
To a 100 mL flask was added 6-aminohexanol (0.76 g, 0.0064 mol) and 25 mL of saturated NaHC03. The homogenous was allowed to stir a RT and N- methoxycarbonylmaleimide (1.00 g, 0.0064 mol) was added as a solid. The mixture cleared shortly after the addition and was allowed to stir for 1 hr. The mixture was extracted by EtOAc, dried (MgS04) and evaporated. The resulting mixture was separated on silica gel (CH2C12 to EtOAC) . To give the product as a white solid 0.32 g (25.2%) , used without further purification. XH (CDC13) δ 1.25 - 1.45 (m, 4 H) , 1.45 - 1.70 (m, 4 H) , 3.53 (t, J = 7.3 Hz,' 2 H) , 3.63 (t, J = 6.0 Hz) , 6.71 (s, 2 H) .
Synthesis of CP-0502:
Compound (6) (CE-1218) was esterified with compound (7) to form compound (8) which was then conjugated to CP-0126 to form the dimer CP-0502. These latter reactions are illustrated in Synthetic Scheme 3 :
SYNTHETIC SCHEME 3
CP-0502 Referring more specifically to Synthetic Scheme 3, compound (6) (200 mg, 0.448 mmol) , triethylamine (0.124 ml, 2 eqv) , 6-maleimidohexanol (7) (97 mg. 1.1 eqv) were dissolved in 2 ml methylene chloride. Bis(2-oxo-3- oxazolidinyl)phosphinic chloride (122 mg, 1.0 eqv) was added to the stirred solution. The resulting suspension was stirred at room temperature for four hours. The reaction mixture was diluted with 25 ml methylene chloride and washed with saturated NaHC03. The organic solution was dried over anhydrous MgS04 and the solvent was removed in vacuo. Silica gel chromatography (2X18 cm column) ; 35/65 :acetone/hexane (Rf=0.4) as eluent provided the compound (8) as a colorless oil. Compound (8) (50 mg, O.Oδmmol) was dissolved in 10 ml DMF containing 100 ul diisopropylethylamine. 100 mg (O.Oδmmols) of CP-0126 was added and reaction proceeded for 30 minutes, with occasional mixing. The reaction mixture was injected on a Vydac 1" C-18 reverse phase column, and eluted at lOml/min, 15%-90% acetonitrile in H20 over 35 minutes (Constant .1% TFA) . The appropriate fractions were lyophilized to yield 52 mg (35%) of a colorless white powder (CP-0502) Laser desorption mass spectrometry M/Z =1890 (M+H) , calculated 1890.
Automated amino acid sequence results confirmed the correct peptide sequence with no altered amino acidsi
In vi tro activity of BKAn/NEI heterodimer(s) In vi tro evaluation of BKAn and NEI activity of the following compounds was carried out according to standard protocols well known to those in the art. BKAn activity (pA2) was assessed using the rat uterus preparation and NEI (K^8) activity was evaluated using purified human neutrophil elastase (HNE) and a synthetic soluble chromogenic substrate, methoxysuccinyl-alanyl-alanyl-prolyl-valyl- paranitroaniline (MOS-AAPV-pNA) . The inhibitor was mixed with MOS-AAPV-pNA (0.5 mM) in 0.05 M sodium phosphate, 0.1 M NaCl, 0.005% Triton X-100, 5% DMSO, pH 7.5. HNE (10-20 nM) is then added. The production of nitroaniline was monitored spectrophotometrically at a wavelength of 400-410 nm at 25 C. An ENZFITTER program then automatically calculated standard enzyme kinetic parameters including K^5.
The following results were obtained:
The data in Table II indicate that for NEI activity there is little difference between the intact heterodimer (CP-0502) and the free monomeric NEI moiety (CE-1218) as far as their respective Kx's are concerned. This is not true for the activity of the BKAn portion of the intact heterodimer relative to its hydrolysis product, CP-0487 (the succinimidohexanol derivative of CP-0126) wherein the intact compound is almost a full log less potent than the monomeric BKAn. Interestingly, the activity of the intact compound displayed a type of irreversible bradykinin antagonism and an apparently enhanced antagonist activity of bradykinin induced uterine contractions at longer incubation times. These types of receptor interactions are not 'well measured by standard pA2 analyses so the differences in activity observed between CP-0487 and CP-0502 with respect to BKAn activity may be more apparent than real. Regardless of the molecular pharmacologic mechanisms underlying these data it is clear that combined BKAn and NEI activity can be incorporated into a single molecule. The above data suggest that allowing for in vivo hydrolysis of the intact compound may alter the behavior of the two moieties so as to enhance the overall in vivo activity of the primary compound. Unfortunately, there are no established animal models that can be employed to assess combined BKAn and NEI activity in vivo . Therefore, in order to assess the potential for in vivo hydrolysis of the intact heterodimer, an in vi tro "surrogate" system was employed wherein the parent heterodimer (CP- 0502) was incubated with human plasma and the resulting metabolites analyzed by reverse phase HPLC.
CP-0502 -was added to freshly obtained normal human plasma and allowed to incubate at 37°C for varying amounts of time. At the designated time the samples were treated with acidified (0.1 N HCl) acetonitrile in order to precipitate the plasma proteins. Aliquots (75 ul) of the supernatents were then analyzed on a Vydac C-18 reverse phase HPLC column using 24% to 80% acetonitrile gradient in 0.1% TFA. The eluent was monitored at 214 nm.
Figures 5 a and b are representative reverse phase HPLC chromatograms illustrative of this type of analysis. As can be seen from these chromatograms, the parent compound appears to be readily hydrolyzed to the succinimidohexanol modified monomer, CP-0487 and its des-Arg9 derivative (Plasma carboxypeptidase will cleave the terminal arginine residue from both the intact heterodimer, CP-0502, as well as CP-0487.) The apparent T1/2 of this hydrolysis reaction is approximately 113 minutes . The NEI component of the heterodimer is an active ester and undergoes hydrolysis as well. However, the intact NEI monomer as well as its hydrolysis products are obscured by the plasma derived peaks seen in the middle of this tracing and cannot be visualized using this system. Since the NEI is equally active as a component of the heterodimer as it is as a monomer, the dissociation of the heterodimer into its two component parts will have less of an effect on its activity than that for the BKAn component.
Those skilled in the art will appreciate that the hydrolysis rate of the heterodimer can be influenced by the steric and electronic environment of the "linking" ester moiety and that the type of chemistry used is only a single example of the types of chemistry. that can be employed to adjust the rate of dissociation (or lack thereof) of the two components of the heterodimer.
Example 6 (BKAn/COI)
Synthesis and Analysis of BKAn/COI Heterodimers
A representative BKAn/COI heterodimer (CP-0460) was synthesized according to Synthetic Schemes 4 and 5 below:
SYNTHETIC SCHEME 4
Synthesis of 6-Maleimidohexanyl 1- (4-chlorobenzoyl) -5-methoxy-2-methyl-3-indohylacetate (9)
To a 100 mL flask was added indomethacin (1.90 g, 0.00532 mol), 25 mL of CH2C12 and DCC (0.55 g, 0.00266 mol) . After 2 hr, the mixture filtered, the
DCU washed with 15 mL of CH2C12 and to this new solution was added 6-maleimidohexanol (0.50 g,
0.00253 mol) as a solid followed by anhydrous Na2C03
(0.32 g, 0.00304 mol) . After 4 days the mixture was filtered diluted with Et20 and washed with 5% NaHC03,
H20 and dried (MgS04) . The resulting yellow oil was purified in a HPLC (silica gel; CH2C12 to 80:20
CH2Cl2/Et0Ac, linear gradient 60 min.) to give 0.79 g
(58.0%) of the desired product as a yellow oil. 1H (CDC13) 1.20 - 1.35 (m, 4 H) , 2.38 (s, 3 H) , 3.47 (t,
J = 7.3 Hz, 2 H) , 3.66 (s, 2 H) , 3.83 (s, 3 H) , 4.08
(t, J = 6.6 Hz, 2 H) , 6.66 (J = 9.0 Hz, J = 2.5 Hz,
1 H), 6.68 (s, 1 H) , 6.87 (d, J = 9:0 Hz, 1 H) , 6.96
(d, J = 2.5 Hz, 1 H) , 7.47 (d, J = 8.5 Hz, 2 H) , 7.66 (d, J = 8.5 Hz, 2 H) .
Conjugation of compound (9) with CP-0126 to form CP-0460 is illustrated in Synthetic Scheme 5 and described thereafter:
SYNTHETIC SCHEME 5
H,N A
CP-0126 (100 mg, 0.08 mmol) was reacted with compound (9) (0.12 mmol, 1.5 eqv) in 2 mL 95% DMF/5% 0.IM ammonium bicarbonate containing 50 ul diisopropylethylamine, for 30 minutes, with occasional mixing. The reaction mixture was purified in 1 injection on a Vydac 1" C-18" reverse phase column at lOml/min, using a gradient running from 15% acetonitrile/0.1% TFA to 40% acetonitrile/0.1% TFA in 20 minutes. Appropriate fractions were lyophilized 'to yield 64 mg (45%) of a colorless powder (CP-0460) . Laser desorption mass spectrometry: M/Z= 1802 (M+H) , calculated 1802.
As mentioned previously, for the COI to work it may need to be dissociated from the BKAn so as to allow for its intracellular penetration.
Therefore, in order to evaluate the functional activity of a BKAn/COI heterodimer, CP-0460 was exposed to rat lung parenchymal strips which were then challenged with arachidonic acid. This tissue is known to contain both non-specific esterase activity as well as to convert arachidonic acid to thromboxane (via a cyclooxygenase dependent pathway) which is then ultimately responsible for the smooth muscle contraction observed in this assay. Using this system, the log dose ratio shifts for indomethacin and CP-0460 were found to be 0.998 +/- 0.425 and 1.029 +/- 0.042 respectively indicating that both indomethacin alone and CP-0460 will prevent the contraction produced in response to exogenously applied arachidonic acid with equal potency. BKAn's have no effect on this system in and of themselves. These data indicate that the COI component of BKAn/COI heterodimer is functionally active in a tissue containing both esterolytic and cyclooxygenase activities. Intact CP-0460 was also tested for BKAn activity using the standard rat uterus assay ,and the pA2 of the CP-0460 was found to be approximately 7.8. Again, CP-0460 (similarly to CP-0502) did not behave as a classical competitive antagonist of bradykinin induced uterine contraction but rather as a type of "pseudo-non-competitive" antagonist, particularly at higher concentrations. This atypical behavior cannot be attributed to COI activity per se as free indomethacin has no effect on this assay at any concentration.
Regardless of the explanation for the observed data, one skilled in the art will appreciate that, as in the other two classes of compounds illustrated herein, pharmacologically important BKAn/COI heterodimers can be' made using a variety of appropriate linking moieties to provide a free hydroxyl and the carboxyl group (a common feature- of many COIs) of the COI monomer to form a hydrolyzable ester based heterodimer. Compounds such as these may be used in the treatment of a variety of inflammatory or painful conditions as well as in the treatment of dysfunctional uterine smooth muscle activity. While the invention has been exemplified above by the use of Y components which are non-peptides, this component may equally comprise in whole or part a peptide as exemplified in the afore-mentioned WO 92/17201, including the heterodimers therein described.
The dimers of the invention may be used in the form of conventional pharmaceutical compositions comprising the active component and a pharmaceutically acceptable carrier. Such compositions may be adapted for topical, oral, aerosolized, intramuscular, subcutaneous or intravenous administration. The amount of active component present in such compositions will range from, for example, about 0.001 to 90.0% by weight depending on the application and mode of administration although more or less of the active component may be used. Conventional dosages will vary considerably on the basis of the intended application and mode of administration. Usually, however, an effective dose is in the order of 0.1 to 1000 micrograms per kg body weight.
The scope of the invention is defined in the following claims wherein:

Claims

CLAIMS What is claimed is:
1. A heterodimer of the formula:
(BKAn) (X) (Y)
where BKAn is a bradykinin antagonist peptide; Y is a different pharmacophore other than BKAn; and X is a linking moiety chemically joining the BKAn and Y components.
2. A heterodimer according to claim 1 wherein Y is a non-peptide pharmacophore with activity against a non-kinin component of the inflammatory process.
3. A heterodimer according to claim 1 wherein
Y is a peptide pharmacophore with activity against a non-kinin component of the inflammatory process.
4. A heterodimer according to claim 1 wherein
Y is a mu-opioid receptor agonist.
5. A heterodimer according to claim 1 wherein
Y is a neutrophil elastase inhibitor.
6. A heterodimer according to claim 1 wherein
Y is a cyclooxygenase inhibitor.
7. A heterodimer according to claim 1 wherein
Y is a NK1 receptor antagonist or NK2 receptor antagonist.
8. A heterodimer according to claim 1 wherein X is hydrolyzable.
9. A heterodimer according to claim 1 wherein X is non-hydrolyzable.
10. A heterodimer according to claim 1 wherein X comprises an amino acid or an amino acid analog incorporated into the BKAn.
11. A heterodimer according to claim 1 wherein X comprises a maleimide/succinimide-based linkage.
12. A heterodimer according to claim 1 wherein X comprises a bissuccinimidoalkane.
13. A heterodimer according to claim 1 wherein X comprises the S atom of a BKAn peptide sulfhydryl group.
14. A heterodimer according to claim 1 wherein
Y is selected from the group consisting of oxycodone or oxymorphone.
15. A heterodimer according to claim 1 wherein
Y is indomethacin.
16. A heterodimer according to claim 1 wherein
Y is a neutrophil elastase inhibitor and X includes a succinimide group attached to BKAn through the sulfur atom of a sulphydryl group of the BKAn
peptide chain.
17. A heterodimer according to claim 16
wherein the neutrophil elastase inhibitor is 4-(3'- carboxy-propylsulfonyl)phenyl-4-tert-butylphenyl isobutyrate.
18. A pharmacophore including an added linker group for linkage to a BKAn peptide.
19. A pharmacophore according to claim 18 wherein the linker group comprises an amine, a carboxylic acid, a hydroxyl, a sulfhydryl or a maleimide group or an amino acid capable of
incorporation into a peptide.
20. A BKAn peptide including a linker group for attachment to a non-peptide pharmacore.
21. A BKAn peptide according to claim 20 wherein the linker group comprises a sulfhydryl group, a maleimide group or an alkane chain
terminating in a hydroxyl, amine or carboxylic acid group. .
22. A pharmacophore according to claim 18 which is a mu-opioid receptor agonist.
23. A pharmacophore according to claim 18 which is a neutrophil elastase inhibitor.
24. A pharmacophore according to claim 18 which is a cyclooxygenase inhibitor.
25. A pharmacophore according to claim 18 which is a NK1 receptor antagonist or NK2 receptor antagonist.
26. A heterdimer according to claim 1 wherein BKAn is DR-R-P-J-G-F-C-DF-L-R or DR-R-P-J-G-Thi-G-
DTic-Oic-R.
27. A heterodimer according to claim 26 wherein the pharmacophore is a mu-opioid receptor agonist.
28. A heterodimer according to claim 26 wherein the pharmacophore is a neutrophil elastase inhibitor.
29. A heterodimer according to claim 26 wherein the pharmacophore is a cyclooxygenase inhibitor.
30. A heterodimer according to claim 26 wherein the pharmacophore is NK1 antagonist or NK2 antagonist.
31. A heterodimer according to claim 28 wherein the pharmacophore is 4-(3'-carboxypropylsulfonyl)phenyl-4-tert-butylphenyl isobutyrate
32. A heterodimer according to claim 31 wherein X comprises a bissuccinimidoalkane group joined by -S- to the BKAn component.
33. The compound 4-(3'-carboxypropylsulphonyl)-phenyl-4-tert-butylphenyl
isobutyrate.
34. A pharmaceutical composition comprising a heterodimer according to claim 1 and a
pharmaceutically acceptable carrier.
35. In a method of treating pain or
inflammation using a bradykinin antagonist, the improvement which comprises using a heterodimer according to claim 1.
EP93924412A 1992-11-10 1993-10-29 Bradykinin antagonists. Withdrawn EP0671941A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US97400092A 1992-11-10 1992-11-10
US974000 1992-11-10
PCT/US1993/010222 WO1994011021A1 (en) 1992-11-10 1993-10-29 Bradykinin antagonists

Publications (2)

Publication Number Publication Date
EP0671941A1 true EP0671941A1 (en) 1995-09-20
EP0671941A4 EP0671941A4 (en) 1996-05-29

Family

ID=25521455

Family Applications (1)

Application Number Title Priority Date Filing Date
EP93924412A Withdrawn EP0671941A4 (en) 1992-11-10 1993-10-29 Bradykinin antagonists.

Country Status (11)

Country Link
EP (1) EP0671941A4 (en)
JP (1) JPH08503460A (en)
CN (1) CN1094058A (en)
AU (1) AU5410994A (en)
CA (1) CA2147869A1 (en)
IL (1) IL107400A0 (en)
MX (1) MX9306988A (en)
NZ (1) NZ257477A (en)
PL (1) PL304654A1 (en)
WO (1) WO1994011021A1 (en)
ZA (1) ZA938014B (en)

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995003308A1 (en) * 1993-07-23 1995-02-02 Toray Industries, Inc. Morphinan derivative and medicinal use
AU6044496A (en) * 1995-06-05 1996-12-24 Cortech, Inc. Compounds having bradykinin antagonistic activity and mu-opi oid agonistic activity
CA2221865C (en) * 1995-06-09 2007-12-11 The Regents Of The University Of Michigan Bradykinin analogs as selective thrombin inhibitors
FR2739553B1 (en) * 1995-10-06 1998-01-02 Oreal USE OF BRADYKININE ANTAGONISTS TO STIMULATE OR INDUCE HAIR GROWTH AND / OR STOP THE HAIR LOSS
DE19612067A1 (en) * 1996-03-27 1997-10-02 Hoechst Ag Use of bradykinin antagonists for the manufacture of medicaments for the treatment of chronic fibrogenetic liver diseases and acute liver diseases
DE19642289A1 (en) 1996-10-14 1998-04-16 Hoechst Ag Use of bradykinin antagonists for the manufacture of medicaments for the treatment and prevention of Alzheimer's disease
US6982249B1 (en) 1997-04-23 2006-01-03 The Regents Of The University Of Michigan Bradykinin analogs as selective inhibitors of cell activation
US6359111B1 (en) * 1998-05-28 2002-03-19 Neorx Corporation Opioid receptor targeting
US6716452B1 (en) 2000-08-22 2004-04-06 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US7060708B2 (en) 1999-03-10 2006-06-13 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
WO2003020200A2 (en) 2000-11-16 2003-03-13 New River Pharmaceuticals Inc. A novel pharmaceutical compound and methods of making and using same
EP2080511A3 (en) * 2000-08-22 2009-09-30 Shire LLC Active agent delivery systems and methods for protecting and administering active agents
AU2001297565B2 (en) * 2000-11-16 2008-06-05 Takeda Pharmaceutical Company Limited A novel pharmaceutical compound and methods of making and using same
ES2373641T3 (en) * 2000-11-16 2012-02-07 Shire Llc A NEW PHARMACEUTICAL COMPOUND AND METHODS TO MANUFACTURE AND USE.
US7375082B2 (en) 2002-02-22 2008-05-20 Shire Llc Abuse-resistant hydrocodone compounds
US7338939B2 (en) 2003-09-30 2008-03-04 New River Pharmaceuticals Inc. Abuse-resistant hydrocodone compounds
US7169752B2 (en) 2003-09-30 2007-01-30 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US7700561B2 (en) 2002-02-22 2010-04-20 Shire Llc Abuse-resistant amphetamine prodrugs
IL163667A0 (en) 2002-02-22 2005-12-18 New River Pharmaceuticals Inc Novel sustained release pharmaceutical compounds to preventabuse of controlled substances
AR040956A1 (en) 2002-07-31 2005-04-27 Schering Ag NEW CONJUGATES OF EFFECTORS, PROCEDURES FOR THEIR PREPARATION AND PHARMACEUTICAL USE
EP1524979A2 (en) * 2002-07-31 2005-04-27 Schering AG New effector conjugates, process for their production and their pharmaceutical use
SG147485A1 (en) 2003-09-30 2008-11-28 Shire Llc Pharmaceutical compositions for prevention of overdose or abuse
GB0624880D0 (en) * 2006-12-14 2007-01-24 Johnson Matthey Plc Improved method for making analgesics

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992017201A1 (en) * 1991-04-01 1992-10-15 Cortech, Inc. Bradykinin antagonists
EP0514361A1 (en) * 1991-05-15 1992-11-19 Aktiebolaget Astra Galanin antagonist

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4894443A (en) * 1984-02-08 1990-01-16 Cetus Corporation Toxin conjugates
IE63490B1 (en) * 1988-11-24 1995-05-03 Hoechst Ag Peptides having bradykinin antagonist action

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992017201A1 (en) * 1991-04-01 1992-10-15 Cortech, Inc. Bradykinin antagonists
EP0514361A1 (en) * 1991-05-15 1992-11-19 Aktiebolaget Astra Galanin antagonist

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
12th American Peptide Symposium, Cambridge, June 16-21, 1991; Program and Abstracts *
JOURNAL OF MEDICINAL CHEMISTRY, vol. 35, no. 9, 1 May 1992 WASHINGTON US, pages 1563-1572, J.C. CHERONIS ET AL. 'A New Class of Bradykinin Antagonists: Synthesis and in Vitro Activity of Bissuccinimidoalkane Peptide Dimers' *
See also references of WO9411021A1 *

Also Published As

Publication number Publication date
CN1094058A (en) 1994-10-26
ZA938014B (en) 1994-07-11
CA2147869A1 (en) 1994-05-26
IL107400A0 (en) 1994-01-25
AU5410994A (en) 1994-06-08
WO1994011021A1 (en) 1994-05-26
PL304654A1 (en) 1995-01-09
MX9306988A (en) 1995-01-31
EP0671941A4 (en) 1996-05-29
JPH08503460A (en) 1996-04-16
NZ257477A (en) 1996-07-26

Similar Documents

Publication Publication Date Title
US6075120A (en) Bradykinin antagonist
EP0671941A1 (en) Bradykinin antagonists
Sandberg et al. Substance P
US5635593A (en) Bradykinin antagonists
CA1340948C (en) Peptidylaminodiols
EP0618810B1 (en) Bradykinin antagonist peptides
US5416191A (en) Bradykinin antagonists
AU696429B2 (en) Bradykinin antagonist peptides incorporating n-substituted glycines
US5610140A (en) Bradykinin receptor antagonists with neurokinin receptor blocking activity
PT90659B (en) PROCESS FOR THE PREPARATION OF RENIN INHIBITORS CONTAINING DIOIS
KR101631726B1 (en) Aminophosphinic derivatives that can be used in the treatment of pain
Bolis et al. Renin inhibitors. Dipeptide analogs of angiotensinogen incorporating transition-state, nonpeptidic replacements at the scissile bond
CA2049743A1 (en) Bradykinin antagonists
JPH0222294A (en) Bradykinin antagonistic peptides
US5750506A (en) Bradykinin antagonists with extended hydrophobic side chains
IE61034B1 (en) 2,3-disubstituted isoxazolidines, a process for their preparation, agents containing them, and their use
WO1993008211A1 (en) Tripeptide derivative anti-inflammatory agents
JP3465000B2 (en) Bradykinin-type peptide
US4631270A (en) Therapeutically useful pseudopeptides, compositions containing the same and methods of preparation and use
JPH05509331A (en) N-(mercaptoalkyl)amide
EP0832106A2 (en) Compounds having bradykinin antagonistic activity and mu-opioid agonistic activity
PL167322B1 (en) Method of obtaining polypeptidic compounds
US4829051A (en) N-substituted derivatives of 1-desaminovasopressin
US6770741B1 (en) Bradykinin antagonist peptides
CA1180006A (en) Therapeutically useful pseudopeptides, compositions containing the same and methods of preparation and use

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19950520

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): CH DE ES FR GB IT LI SE

RHK1 Main classification (correction)

Ipc: C07K 7/18

A4 Supplementary search report drawn up and despatched
AK Designated contracting states

Kind code of ref document: A4

Designated state(s): CH DE ES FR GB IT LI SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Withdrawal date: 19971015