WO1998035055A1 - Method for identifying selective inhibitors of nitric oxide synthase - Google Patents

Method for identifying selective inhibitors of nitric oxide synthase Download PDF

Info

Publication number
WO1998035055A1
WO1998035055A1 PCT/GB1998/000353 GB9800353W WO9835055A1 WO 1998035055 A1 WO1998035055 A1 WO 1998035055A1 GB 9800353 W GB9800353 W GB 9800353W WO 9835055 A1 WO9835055 A1 WO 9835055A1
Authority
WO
WIPO (PCT)
Prior art keywords
nos
compound
activity
concentration
determining
Prior art date
Application number
PCT/GB1998/000353
Other languages
French (fr)
Inventor
Ian George Charles
Kiran Bhagat
Patrick John Thompson Vallance
Aroon Dinesh Hingorani
Original Assignee
University College London
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 University College London filed Critical University College London
Priority to EP98903134A priority Critical patent/EP0961835A1/en
Priority to AU59962/98A priority patent/AU5996298A/en
Priority to JP53399198A priority patent/JP2001511009A/en
Publication of WO1998035055A1 publication Critical patent/WO1998035055A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/26Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase

Definitions

  • the present invention relates to methods for identifying compounds which selectively inhibit nitric oxide synthase (NOS). It also relates to the use of such compounds in methods of treatment.
  • NOS nitric oxide synthase
  • NO nitric oxide
  • iNOS inducible NOS
  • eNOS endothelial NOS
  • nNOS neuronal NOS
  • iNOS In rodent cells, expression of iNOS occurs readily in vascular smooth muscle cells in response to bacterial endotoxin or certain inflammatory cytokines and largely accounts for the hypotension seen in experimental septic shock. Despite the evidence for involvement of iNOS in rodent models, reproducible and consistent induction of functionally active iNOS in human vascular cells and tissues in vitro has not been achieved.
  • the present invention is based on the surprising finding that cytokine induced vasodilatation in humans caused by an increase in NO generation is due to activation of eNOS and not induction of iNOS. This phenomenon appears to be as a result of transcriptional regulation of GTP cyclohydrolase 1 leading to an increase in levels of the pterin tetrahydrobiopterin (BH 4 ), a co-factor for all three known isoforms of NOS. The increase in levels of BH 4 in turn leads to increased eNOS activity.
  • the present invention provides a method for identifying a compound which selectively inhibits a nitric oxide synthase (NOS) which method comprises: (a) determining the inhibition of the activity of the NOS in the presence of a candidate compound and a first concentration of BH 4 ; (b) determining the inhibition of the activity of the NOS in the presence of the candidate compound and a second concentration BH 4 ; (c) determining whether the candidate compound inhibits the activity of the NOS to a different extent in (a) than in (b).
  • NOS nitric oxide synthase
  • step (a) comprises: (i) determining the activity of said NOS in the absence of the candidate compound and in the presence of a first concentration of BH 4 ; (ii) determining the activity of said NOS in the presence of the candidate compound and the first concentration of BH 4 ; (iii) comparing the activity of said NOS in steps (i) and (ii).
  • step (b) comprises: (i) determining the activity of said NOS in the absence of the candidate compound and in the presence of a second concentration of BH 4 ; (ii) determining the activity of said NOS in the presence of the candidate compound and the second concentration of BH 4 ; (iii) comparing the activity of said NOS in steps (i) and (ii).
  • the second concentration of BF£ 4 used in step (b) is preferably lower than the first concentration of BF£ 4 used in step (a).
  • the NOS is preferably a human NOS, more preferably human eNOS.
  • nitric oxide synthase activity may be used to treat a variety of conditions in which overproduction of nitric oxide has been implicated, for example septic shock, asthma, arthritis, inflammatory bowel disease, heart failure and acute systemic inflammation.
  • this approach might be of value in the treatment of neurological disease states in which nNOS has been implicated (for example, strokes, dementia and Parkinsons disease).
  • the present invention also provides the use of a compound identified by the method of the invention in the manufacture of a medicament for use in selectively inhibiting a human nitric oxide synthase.
  • the present invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound identified by the method of the invention together with a pharmaceutically acceptable carrier or diluent.
  • Cytokine-induced stimulation of NO production by NOS appears to be mediated via an increase in the activity of GTP cyclohydrolase 1 which is the rate-limiting enzyme in the synthesis of BH 4 . Since BH 4 is required for NOS activity, and therefore increases in
  • BH 4 levels in response to cytokines result in increased NOS activity
  • inhibition of GTP cyclohydrolase activity may prevent or reduce NO production by any isoform of NOS.
  • the present invention further provides the use of a compound in the manufacture of a medicament for use in inhibiting a human nitric oxide synthase wherein said compound is capable of inhibiting or reducing synthesis of BH 4 by GTP cyclohydrolase 1.
  • the compound is identified by an assay method which method comprises:
  • NOS nitric oxide synthase
  • the phrase "nitric oxide synthase” is intended to include all naturally occuring forms of eNOS, iNOS and nNOS as well as variants which retain NOS activity, for example functional forms of iNOS encoded by additional copies of the iNOS gene or variants produced by mutagenesis techniques.
  • the NOS is preferably of mammalian origin, for example rodent (including rat and mouse) or primate (such as human).
  • the NOS is of human origin.
  • NOS used in the assays may be obtained from mammal cellular extracts or produced recombinantly from, for example, bacteria, yeast or higher eukaryotic cells including mammalian cell lines and insect cell lines.
  • NOS used in the assays is recombinant (see, for example, Charles et al, 1996).
  • Suitable candidate compounds may include analogues of arginine, aminoguanidine or analogues thereof, guanidine compounds, isothiourea, derivatives of L-N 6 - (l-iminoethyl)lysine (L-NMMA), 2-nitroaryl and 2-cyanoaryl compounds or ⁇ -(S-methylisothioureido)-L-norvaline.
  • the inventors have found that the sensitivity of NOS to different inhibitors may vary depending on the physiological pterin concentration. This may be due to an allosteric effect on the conformation of NOS resulting in a BH 4 -modif ⁇ ed isoform.
  • the inventors have found that the rise in physiological BH 4 levels caused by cytokine/endotoxin stimulation increases the sensitivity of eNOS to aminoguanidine. Therefore to provide maximum specificity and clinical efficacy it is very important to screen for potential inhibitors, for use in therapeutic methods, at a physiologically or pathophysiologically relevant pterin concentration.
  • the principle behind the assay methodologies is to screen for compounds which inhibit a particular NOS isoform at a particular physiologically or pathophysiologically relevant concentration or concentration range of BH 4 but which have a reduced inhibitory effect at other concentrations.
  • the human eNOS isoform which is present at normal BH 4 concentrations is essential for maintaining homeostatic pressure. Any compound which dramatically inhibits this isoform will have a potentially deleterious effect on the patient.
  • eNOS is converted into a form, possibly due to an allosteric conformational change, which is responsible for generating the NO which causes refractory hypotension. It is this latter form which needs to be targeted.
  • a suitable inhibitor would inhibit eNOS at the pathophysiological concentration of BH 4 present as a result of the action of, for example, inflammatory cytokines but would not inhibit eNOS at normal BH 4 concentrations.
  • Step (a) of the method of the invention comprises determining the inhibition of NOS activity by the candidate compound in the presence of a first concentration of BH 4 .
  • the amount of NOS used in the assay has a specific activity of from 10 to 600 pmol/min.
  • the concentration of BH 4 used in the assay is from 1 ⁇ M to 1000 ⁇ M, preferably from 10 ⁇ M to 1000 ⁇ M, more preferably from 100 ⁇ M to 1000 ⁇ M or 500 ⁇ M to 1000 ⁇ M, most preferably from 500 ⁇ M to 1000 ⁇ M.
  • the concentration of candidate compound used in the assay is from 1 ⁇ M to 1000 ⁇ M, preferably from 1 ⁇ M to 100 ⁇ M, more preferably from 1 ⁇ M to 10 ⁇ M.
  • inhibition, if any, of NOS activity is determined by firstly measuring NOS activity in the absence of the candidate compound but in the presence of the first concentration of BH 4 . This provides a control value for NOS activity under the assay conditions. Secondly, NOS activity is measured in the presence of both the candidate compound and the first concentration of BH 4 . The inhibition of NOS activity due to the candidate compound can be determined by comparing the control value with the value obtained in the presence of the candidate compound. These assay steps are conducted by contacting NOS with the candidate compound and/or BH 4 where appropriate. The extent and/or the rate of reaction can be measured quantitatively, semi-quantitatively or qualitatively. If, for example, a fast throughput screening assay format is required, the control reaction in the absence of the candidate compound need not be performed for every candidate compound tested.
  • NOS activity can be measured in a number of ways (see Packer, 1996). For example, NOS activity can be measured directly by conversion of radiolabelled arginine to citrulline or indirectly by assaying for NO production. Assays for NO include chemical assays or bioassays. A typical chemical assay for NO utilises the change in absorbance at 405 nm and 420 nm when NO oxidises oxyhaemoglobin to methemoglobin.
  • the reaction mixture comprises: 3 ⁇ M oxyhaemoglobin, 200 ⁇ M CaCl 2 (for eNOS and nNOS), 1 mM MgCl 2 , 1 ⁇ M FAD, 1 ⁇ M FMN, 100 ⁇ M NADPH, 0.1 ⁇ M calmodulin, 30 ⁇ M L-arginine (substrate for NOS), from 1 ⁇ M to 1000 ⁇ M BH 4 and 100 ⁇ M DTT; in 100 mM HEPES buffer, pH 7.4 and in a final reaction volume of 250 ⁇ l (Charles et al, 1996).
  • This assay is suitable for use in a microtitre plate format.
  • Step (b) of the method of the invention comprises determining the extent to which the activity of the NOS is inhibited by the candidate compound in the presence of a second concentration of BH 4 .
  • This step is similar to step (a) except that the concentration of BH 4 used is different to that used in step (a), preferably lower.
  • the amount of NOS used in the assay has a specific activity of from 10 to 600 pmol/min.
  • the second concentration of BH 4 used in the assay is from 1 ⁇ M to 1000 ⁇ M, preferably from 1 ⁇ M to 100 ⁇ M, more preferably from 1 ⁇ M to 10 ⁇ M.
  • the concentration of candidate compound used in the assay is from 1 ⁇ M to 1000 ⁇ M, preferably from 1 ⁇ M to 100 ⁇ M, more preferably from 1 ⁇ M to 10 ⁇ M.
  • the steady-state inhibition is determined after between 15 to 30 minutes incubation.
  • Step (c) of the method of the invention comprises determining whether the candidate compound inhibits the NOS activity to a different extent in (a) than in (b).
  • a candidate compound may inhibit eNOS by 70% in the presence of a high concentration of BH 4 , but only by 5% in the presence of low concentrations of BH 4 .
  • Such a compound would be a useful therapeutic agent because it does not inhibit the normal physiological form of eNOS involved in maintaining homeostatic pressure, but does inhibit the isoform of eNOS which is generated in the presence of high levels of BH 4 caused by inflammatory cytokines/endotoxins.
  • NOS activity in the presence of an inhibitor is dependent on
  • the inhibitor is preferably selective with respect to different isoforms of NOS, for example eNOS, iNOS and nNOS, whether from the same organism or different organisms.
  • an inhibitor may inhibit human eNOS but not human iNOS.
  • inhibition of eNOS activity at a higher level of BH 4 by an inhibitor compound is typically at least 20%, preferably at least 30%, more preferably at least 40, 50, 75, 90 or 95% greater than the inhibition of eNOS activity by the inhibitor compound at a lower level of BH 4 .
  • concentrations typically envisaged for the higher and lower levels of BH 4 are discussed above.
  • assays may include in vivo assays such as those described in WO95/34534.
  • the inhibition of eNOS and iNOS in situ in rat aortic rings can assessed by measuring the increases in ring tension caused by NO synthase inhibition.
  • endothelium-denuded rings are typically exposed to LPS (0.1 ⁇ g/ml from S.
  • eNOS and iNOS in vivo can also be assessed by the effects of inhibitors on blood pressure in either normal or endotoxin shocked conscious mice. For example, one suitable method is as follows. Mice are anaesthetised briefly with isoflourane (2%).
  • Cannula lines are implanted in a femoral vein, tunnelled subcutaneously to exit at the top of the back and connected to a swivel tether system for continuous monitoring of blood pressure and for inhibitor administration respectively.
  • animals with mean blood pressures in the normal range 90-110 mm Hg are used to obtain cumulative concentration curves for inhibitors on blood pressure either without further treatment ("normal mice") or 7 h after administration of lipopolysaccharide (12.5 mg/kg of LPS from E. coli 026:B6 intravenously over 30 s) to induce shock (“shocked mice”).
  • lipopolysaccharide 12.5 mg/kg of LPS from E. coli 026:B6 intravenously over 30 s
  • cytokine-induced stimulation of NO production by NOS appears to be mediated via an increase in the activity of GTP cyclohydrolase 1 which is the rate-limiting enzyme in the synthesis of BH 4 . Since BH 4 is required for NOS activity, and therefore increases in BH 4 levels in response to cytokines result in increased NOS activity, inhibition of GTP cyclohydrolase activity may prevent or reduce NO production by NOS. Thus compounds which inhibit GTP cyclohydrolase 1 activity may also inhibit NOS activity as a result of the inhibition or reduction in BH 4 production.
  • inhibition, if any, of GTP cyclohydrolase 1 is determined by firstly measuring GTP cyclohydrolase 1 activity in the absence of the candidate compound. This provides a control value for GTP cyclohydrolase 1 activity under the assay conditions. Secondly, GTP cyclohydrolase 1 activity is measured in the presence of the candidate compound. The inhibition of GTP cyclohydrolase 1 activity due to the candidate compound can be determined by comparing the control value with the value obtained in the presence of the candidate compound. The assays are conducted by contacting GTP cyclohydrolase 1 with the candidate compound. The extent and/or the rate of reaction can be measured quantitatively, semi-quantitatively or qualitatively.
  • the control reaction in the absence of the candidate compound need not be performed for every candidate compound tested.
  • the amount of GTP cyclohydrolase 1 used in the assay is from 1 to 10 ng of pure protein.
  • the concentration of candidate compound used in the assay is from 1 ⁇ M to 1000 ⁇ M, preferably from 1 ⁇ M to 500 ⁇ M or 1 ⁇ M to 100 ⁇ M, more preferably from 1 ⁇ M to 10 ⁇ M.
  • GTP cyclohydrolase 1 activity can be measured, for example, by assessing the production of BH 4 or GTP consumption.
  • BH 4 production can be determined, for example, by oxidation to biopterin using iodine and measurement of biopterin using HPLC (mobile phase is 5% methanol run at 1.5 ml/min on a 10 micron ODS reverse phase column (4.6 mm x 25 cm) with excitation at 350 nm and emission at 410 nm).
  • nitric oxide synthases can lead to potentially fatal vasodilatation and hypotension.
  • Selective inhibition of nitric oxide synthases by a compound identified by the method of the invention may be useful in alleviating this potentially fatal response.
  • Compounds identified by the method of the invention may be used therapeutically to treat conditions in which over-production of NO is implicated, for example septic shock, asthma, rheumatoid arthritis, inflammatory bowel disease, heart failure or acute systemic inflammation. NOS is also implicated in various neurological disease states including strokes, dementia and Parkinsons disease.
  • compounds which inhibit or reduce synthesis of BH 4 by GTP cyclohydrolase 1 may also be used therapeutically to treat the conditions described above in which NOS is implicated.
  • the compound is used to treat diseases in which eNOS is implicated.
  • the formulation will depend upon the nature of the compound identified but typically the compounds may be formulated for clinical administration by mixing them with a pharmaceutically acceptable carrier or diluent.
  • a pharmaceutically acceptable carrier or diluent for example they can be formulated for topical, parenteral, intravenous, intramuscular, subcutaneous, intraocular or transdermal administration.
  • the compound is used in an injectable form. It may therefore be mixed with any vehicle which is pharmaceutically acceptable for an injectable formulation, preferably for a direct injection at the site to be treated.
  • the pharmaceutically carrier or diluent may be, for example, sterile or isotonic solutions. It is also preferred to formulate that compound in an orally active form.
  • the dose of compound used may be adjusted according to various parameters, especially according to the compound used, the age, weight and condition of the patient to be treated, the mode of administration used and the required clinical regimen.
  • the amount of compound administered by injection is suitably from 0.01 mg/kg to 30 mg/kg, preferably from 0.1 mg/kg to 10 mg/kg.
  • FIG. 2 Panel A - Two adjacent vessels were studied simultaneously and at a distension pressure of 40 mmHg.
  • the sympathetic nervous system was activated by asking subjects to take a deep breath. This manoeuvre produced transient constriction in both vessels. Incubation of one vessel (lower trace) with IL-l ⁇ virtually abolished sympathetically mediated constriction 6 h later. Sympathetic constriction was restored by infusion of L-NMMA. Congesting cuff deflation and re-inflation is marked by solid circle.
  • Panel B Traces showing effects of L-NMMA on basal vessel tone in control vessel (upper trace) and vein treated with IL- l ⁇ (lower trace). The effects of L-NMMA were reversed by L-arginine.
  • Panel B - Original trace showing effects of BH 4 infusion (upper trace) and reversal by aminoguanidine (lower trace). Congesting cuff deflation and re-inflation is marked by a solid circle.
  • Figure 4 - PCR reactions were carried out in 20 ⁇ l volumes, and 5 ⁇ l samples analysed by electrophoresis through 2% agarose gels.
  • Panel A shows the result with the iNOS primer pair BB52 and BB71.
  • Track 1 size markers tracks 2 & 3 niRNA from uninduced hand vein; track 4 and 5 mRNA from cytokine-induced hand vein; track 6 positive control (cytokine-induced CAPAN-1 mRNA).
  • a PCR product of 499 bp is only visible in track 6.
  • Panel B shows the result of PCR amplification with the eNOS specific primer pair
  • Panel C shows the ⁇ -actin specific PCR products from the primer set BB27 and BB28.
  • Traces 1-4 are the hand vein mRNA samples amplified with an initial reverse transcriptase step.
  • Tracks 5-8 are negative controls, amplified in the absence of a reverse transcriptase step.
  • Track 9 is a positive control using, placental mRNA.
  • Track 10 is size markers. Bands corresponding to ⁇ -actin can be detected in tracks 1-4 confirming that mRNA had been isolated from hand vein biopsies.
  • the volume of blood in the isolated vein is in the order of 1-2 ml and the calculated concentration of cytokine was in the order of 300-1000 pg/ml (TNF ⁇ and IL-l ⁇ ) and 30-100 pg/ml (IL-6).
  • RT-PCR Reverse transcriptase polymerase chain reaction
  • Poly A mRNA extracted from hand veins using Fast Track reagents (InvitrogenTM).
  • Positive control poly A + mRNA for eNOS was carried out on human placental preparations (ClontechTM).
  • Positive controls for iNOS and GTPCH-1 were carried out on poly A + mRNA isolated from the human pancreatic adenocarcinoma line CAPAN-1 stimulated with cytokines. The following primer sets were used:
  • GTPCH-1 igcl 5'-ttggttatcttcctaacaag-3' igc2 5'-gtgctggtcacagttttgct-3'
  • PCR was carried out in thin-walled tubes using a Gene AmpTM RT PCR kit (Perkin-ElmerTM) in a Gene Amp PCR system 9600 thermocycler (Perkin-Elmer). The following conditions were used: 95°C for lmin 45s, followed by 30 cycles of 95°C, 15s, 60°C, 30s. An elongation step of 72°C was carried out for 7 min at the end of the cycle.
  • tumour necrosis factor ⁇ TNF ⁇
  • IL-6 100 pg, in 1 ml of saline
  • co-instillation of these cytokines with IL-l ⁇ (1 ng/ml) for 1 h caused a prolonged (>24 h) attenuation of the response to noradrenaline ( Figure lb).
  • eNOS is sensitive to inhibition by an iNOS inhibitor, aminoguanidine. in the presence of
  • aminoguanidine was inhibited by aminoguanidine.
  • the ability of aminoguanidine to inhibit eNOS activity is dependent on the prevailing concentration of BH 4 .
  • IL-l ⁇ induces expression of GTP cyclohydrolase 1 leading to increased generation of BH,.
  • the rate limiting enzyme for BH 4 synthesis is GTP cyclohydrolase 1.
  • This enzyme is transcriptionally regulated in response to cytokines or endotoxin, and its induction can be prevented by glucocorticoids. It is clear, therefore that the pharmacological changes we observed in the hand vein in vivo might be explained equally well by activation of eNOS by BH 4 synthesised following induction of GTP cyclohydrolase 1, as by induction of iNOS.
  • To explore further the mechanism of the functional induction of NOS activity we excised a portion of hand vein. As expected, mRNA encoding eNOS was detected in all of the samples ( Figure 4).
  • eNOS is an endothelial enzyme that has not been detected in smooth muscle cells.
  • An implication of our findings is that the endothelium can generate sufficient NO to cause profound vasodilatation and that expression of NOS throughout the much larger smooth muscle cell layer is not necessary to cause septic hypotension.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Cardiology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

A method for identifying a compound which selectively inhibits a human nitric oxide synthase (NOS) comprises: (a) determining the inhibition of the activity of the NOS in the presence of a candidate compound and a first concentration of tetrahydrobiopterin (BH4); (b) determining the inhibition of the activity of the NOS in the presence of the candidate compound and a second concentration of the BH4; (c) determining whether the candidate compound inhibits the activity of the NOS to a different extent in (a) than in (b).

Description

METHOD FOR IDENTIFYING SELECTIVE INHIBITORS OF NITRIC OXIDE SYNTHASE
Field of the invention
The present invention relates to methods for identifying compounds which selectively inhibit nitric oxide synthase (NOS). It also relates to the use of such compounds in methods of treatment.
Background to the invention
Septic shock is associated with refractory hypotension and claims over 100,000 lives each year in the USA. Vasodilatation is a ubiquitous and conserved response of the cardiovascular system to acute systemic inflammation, and is the major cause of the hypotension associated with septic shock. Over-production of nitric oxide (NO) has been implicated in the pathophysiology of septic shock in both rodents and man. NO synthesis is catalysed by the enzyme nitric oxide synthase (NOS). There are three known types of mammalian NOS: inducible NOS (iNOS), endothelial NOS (eNOS) and neuronal NOS (nNOS). In rodent cells, expression of iNOS occurs readily in vascular smooth muscle cells in response to bacterial endotoxin or certain inflammatory cytokines and largely accounts for the hypotension seen in experimental septic shock. Despite the evidence for involvement of iNOS in rodent models, reproducible and consistent induction of functionally active iNOS in human vascular cells and tissues in vitro has not been achieved.
Although over-production of NO has been implicated in the pathophysiology of septic shock in man and iNOS is a target for drug therapy, there is no direct evidence that iNOS is expressed in the vasculature of patients with sepsis. Furthermore the plasma concentrations of NOx in patients with sepsis are elevated about 0.5 fold, compared with the 10-fold elevation seen in rodent models. These observations are supported by molecular evidence for important functional differences between iNOS in different species.
Summary of the invention
The present invention is based on the surprising finding that cytokine induced vasodilatation in humans caused by an increase in NO generation is due to activation of eNOS and not induction of iNOS. This phenomenon appears to be as a result of transcriptional regulation of GTP cyclohydrolase 1 leading to an increase in levels of the pterin tetrahydrobiopterin (BH4), a co-factor for all three known isoforms of NOS. The increase in levels of BH4in turn leads to increased eNOS activity.
The finding that the physiological pathway leading to NO generation by NOSs can be modulated by changes in pterin concentration has specific implications for the design of drugs to treat septic hypotension and inflammatory vasodilatation. It might also be more generally relevant to the route by which excess NO is generated in multiple cell types during systemic inflammation in humans, as well as in the wide range of inflammatory or neoplastic conditions in which pterin synthesis is altered.
Accordingly, the present invention provides a method for identifying a compound which selectively inhibits a nitric oxide synthase (NOS) which method comprises: (a) determining the inhibition of the activity of the NOS in the presence of a candidate compound and a first concentration of BH4; (b) determining the inhibition of the activity of the NOS in the presence of the candidate compound and a second concentration BH4; (c) determining whether the candidate compound inhibits the activity of the NOS to a different extent in (a) than in (b).
Preferably step (a) comprises: (i) determining the activity of said NOS in the absence of the candidate compound and in the presence of a first concentration of BH4; (ii) determining the activity of said NOS in the presence of the candidate compound and the first concentration of BH4; (iii) comparing the activity of said NOS in steps (i) and (ii).
Preferably step (b) comprises: (i) determining the activity of said NOS in the absence of the candidate compound and in the presence of a second concentration of BH4; (ii) determining the activity of said NOS in the presence of the candidate compound and the second concentration of BH4; (iii) comparing the activity of said NOS in steps (i) and (ii).
The second concentration of BF£4 used in step (b) is preferably lower than the first concentration of BF£4 used in step (a). The NOS is preferably a human NOS, more preferably human eNOS.
Compounds identified by the method of the invention which selectively inhibit nitric oxide synthase activity may be used to treat a variety of conditions in which overproduction of nitric oxide has been implicated, for example septic shock, asthma, arthritis, inflammatory bowel disease, heart failure and acute systemic inflammation. In addition, this approach might be of value in the treatment of neurological disease states in which nNOS has been implicated (for example, strokes, dementia and Parkinsons disease). Thus, the present invention also provides the use of a compound identified by the method of the invention in the manufacture of a medicament for use in selectively inhibiting a human nitric oxide synthase.
The present invention further provides a pharmaceutical composition comprising a compound identified by the method of the invention together with a pharmaceutically acceptable carrier or diluent.
Cytokine-induced stimulation of NO production by NOS appears to be mediated via an increase in the activity of GTP cyclohydrolase 1 which is the rate-limiting enzyme in the synthesis of BH4. Since BH4 is required for NOS activity, and therefore increases in
BH4 levels in response to cytokines result in increased NOS activity, inhibition of GTP cyclohydrolase activity may prevent or reduce NO production by any isoform of NOS.
Thus the present invention further provides the use of a compound in the manufacture of a medicament for use in inhibiting a human nitric oxide synthase wherein said compound is capable of inhibiting or reducing synthesis of BH4 by GTP cyclohydrolase 1. Preferably the compound is identified by an assay method which method comprises:
(i) contacting GTP cyclohydrolase 1 with a candidate compound; and (ii) determining the inhibition of GTP cyclohydrolase-mediated synthesis of BH4 by said compound.
Detailed description of the invention
The phrase "nitric oxide synthase" (NOS) is intended to include all naturally occuring forms of eNOS, iNOS and nNOS as well as variants which retain NOS activity, for example functional forms of iNOS encoded by additional copies of the iNOS gene or variants produced by mutagenesis techniques. The NOS is preferably of mammalian origin, for example rodent (including rat and mouse) or primate (such as human). Preferably the NOS is of human origin. NOS used in the assays may be obtained from mammal cellular extracts or produced recombinantly from, for example, bacteria, yeast or higher eukaryotic cells including mammalian cell lines and insect cell lines. Preferably, NOS used in the assays is recombinant (see, for example, Charles et al, 1996).
Suitable candidate compounds may include analogues of arginine, aminoguanidine or analogues thereof, guanidine compounds, isothiourea, derivatives of L-N6- (l-iminoethyl)lysine (L-NMMA), 2-nitroaryl and 2-cyanoaryl compounds or δ-(S-methylisothioureido)-L-norvaline.
Assay methodologies
The inventors have found that the sensitivity of NOS to different inhibitors may vary depending on the physiological pterin concentration. This may be due to an allosteric effect on the conformation of NOS resulting in a BH4-modifϊed isoform. In particular, as an example, the inventors have found that the rise in physiological BH4 levels caused by cytokine/endotoxin stimulation increases the sensitivity of eNOS to aminoguanidine. Therefore to provide maximum specificity and clinical efficacy it is very important to screen for potential inhibitors, for use in therapeutic methods, at a physiologically or pathophysiologically relevant pterin concentration. The principle behind the assay methodologies is to screen for compounds which inhibit a particular NOS isoform at a particular physiologically or pathophysiologically relevant concentration or concentration range of BH4 but which have a reduced inhibitory effect at other concentrations. By way of an example, the human eNOS isoform which is present at normal BH4 concentrations is essential for maintaining homeostatic pressure. Any compound which dramatically inhibits this isoform will have a potentially deleterious effect on the patient. However, at increased BH4 concentrations induced by, for example, inflammatory cytokines, eNOS is converted into a form, possibly due to an allosteric conformational change, which is responsible for generating the NO which causes refractory hypotension. It is this latter form which needs to be targeted. Thus a suitable inhibitor would inhibit eNOS at the pathophysiological concentration of BH4 present as a result of the action of, for example, inflammatory cytokines but would not inhibit eNOS at normal BH4 concentrations.
Step (a) of the method of the invention comprises determining the inhibition of NOS activity by the candidate compound in the presence of a first concentration of BH4. Typically, the amount of NOS used in the assay has a specific activity of from 10 to 600 pmol/min. Typically, the concentration of BH4 used in the assay is from 1 μM to 1000 μM, preferably from 10 μM to 1000 μM, more preferably from 100 μM to 1000 μM or 500 μM to 1000 μM, most preferably from 500 μM to 1000 μM. Typically, the concentration of candidate compound used in the assay is from 1 μM to 1000 μM, preferably from 1 μM to 100 μM, more preferably from 1 μM to 10 μM.
Generally, inhibition, if any, of NOS activity, is determined by firstly measuring NOS activity in the absence of the candidate compound but in the presence of the first concentration of BH4. This provides a control value for NOS activity under the assay conditions. Secondly, NOS activity is measured in the presence of both the candidate compound and the first concentration of BH4. The inhibition of NOS activity due to the candidate compound can be determined by comparing the control value with the value obtained in the presence of the candidate compound. These assay steps are conducted by contacting NOS with the candidate compound and/or BH4 where appropriate. The extent and/or the rate of reaction can be measured quantitatively, semi-quantitatively or qualitatively. If, for example, a fast throughput screening assay format is required, the control reaction in the absence of the candidate compound need not be performed for every candidate compound tested.
NOS activity can be measured in a number of ways (see Packer, 1996). For example, NOS activity can be measured directly by conversion of radiolabelled arginine to citrulline or indirectly by assaying for NO production. Assays for NO include chemical assays or bioassays. A typical chemical assay for NO utilises the change in absorbance at 405 nm and 420 nm when NO oxidises oxyhaemoglobin to methemoglobin. Typically, the reaction mixture comprises: 3 μM oxyhaemoglobin, 200 μM CaCl2 (for eNOS and nNOS), 1 mM MgCl2, 1 μM FAD, 1 μM FMN, 100 μM NADPH, 0.1 μM calmodulin, 30 μM L-arginine (substrate for NOS), from 1 μM to 1000 μM BH4 and 100 μM DTT; in 100 mM HEPES buffer, pH 7.4 and in a final reaction volume of 250 μl (Charles et al, 1996). This assay is suitable for use in a microtitre plate format.
All three isoforms require L-arginine, NADPH, FMN, FAD, BH4 and calmodulin for activity. eNOS and nNOS also require calcium (Charles et al, 1996). Step (b) of the method of the invention comprises determining the extent to which the activity of the NOS is inhibited by the candidate compound in the presence of a second concentration of BH4. This step is similar to step (a) except that the concentration of BH4 used is different to that used in step (a), preferably lower. Typically, the amount of NOS used in the assay has a specific activity of from 10 to 600 pmol/min. Typically the second concentration of BH4 used in the assay is from 1 μM to 1000 μM, preferably from 1 μM to 100 μM, more preferably from 1 μM to 10 μM. Typically, the concentration of candidate compound used in the assay is from 1 μM to 1000 μM, preferably from 1 μM to 100 μM, more preferably from 1 μM to 10 μM. Generally the steady-state inhibition is determined after between 15 to 30 minutes incubation. Step (c) of the method of the invention comprises determining whether the candidate compound inhibits the NOS activity to a different extent in (a) than in (b). This can be accomplished by simply comparing the inhibition in step (a), as indicated by comparing the control value with the second value, and the extent of inhibition in step (b) similarly indicated by comparing the control value with the second value. Thus, for example, a candidate compound may inhibit eNOS by 70% in the presence of a high concentration of BH4, but only by 5% in the presence of low concentrations of BH4. Such a compound would be a useful therapeutic agent because it does not inhibit the normal physiological form of eNOS involved in maintaining homeostatic pressure, but does inhibit the isoform of eNOS which is generated in the presence of high levels of BH4 caused by inflammatory cytokines/endotoxins. In other words, NOS activity in the presence of an inhibitor is dependent on
(inversely proportional to) the concentration of BH4. The inhibitor is preferably selective with respect to different isoforms of NOS, for example eNOS, iNOS and nNOS, whether from the same organism or different organisms. For example, an inhibitor may inhibit human eNOS but not human iNOS. By way of example in numerical form, inhibition of eNOS activity at a higher level of BH4 by an inhibitor compound is typically at least 20%, preferably at least 30%, more preferably at least 40, 50, 75, 90 or 95% greater than the inhibition of eNOS activity by the inhibitor compound at a lower level of BH4. The actual concentrations typically envisaged for the higher and lower levels of BH4 are discussed above. When screening candidate compounds for use in inhibiting NOS in the treatment of cardiovascular disorders it will be preferable to use eNOS in the assay. Conversely, when screening candidate compounds for use in inhibiting NOS in the treatment of neurological disorders it will be preferable to use nNOS in the assay.
Other types of assay may include in vivo assays such as those described in WO95/34534. For example, the inhibition of eNOS and iNOS in situ in rat aortic rings can assessed by measuring the increases in ring tension caused by NO synthase inhibition. For studies of basal tone, rings of thoracic aorta with intact endothelium are typically prepared as described in Rees et al. (1989) and cumulative concentration curves obtained for the inhibitors in the presence of a threshold concentration of phenylephrine (ED10=10nM). For studies of induced smooth muscle tone, endothelium-denuded rings are typically exposed to LPS (0.1 μg/ml from S. thphosa) in the presence of phenylephrine at approximately ED90 for 6 h as described in Rees et al. (1990). During this time a progressive loss of tone usually occurs because of iNOS induction. Generally, cumulative concentration curves are then obtained for the inhibitors. The inhibition of eNOS and iNOS in vivo can also be assessed by the effects of inhibitors on blood pressure in either normal or endotoxin shocked conscious mice. For example, one suitable method is as follows. Mice are anaesthetised briefly with isoflourane (2%). Cannula lines are implanted in a femoral vein, tunnelled subcutaneously to exit at the top of the back and connected to a swivel tether system for continuous monitoring of blood pressure and for inhibitor administration respectively. Following recovery from surgery, animals with mean blood pressures in the normal range (90-110 mm Hg) are used to obtain cumulative concentration curves for inhibitors on blood pressure either without further treatment ("normal mice") or 7 h after administration of lipopolysaccharide (12.5 mg/kg of LPS from E. coli 026:B6 intravenously over 30 s) to induce shock ("shocked mice"). It will be appreciated that other suitable assays may be used to determine inhibitors of NOS activity in vivo (or indeed in vitro) such as those described in the examples and that the above mentioned assays are by way of example.
The inventors have also found that cytokine-induced stimulation of NO production by NOS appears to be mediated via an increase in the activity of GTP cyclohydrolase 1 which is the rate-limiting enzyme in the synthesis of BH4. Since BH4 is required for NOS activity, and therefore increases in BH4 levels in response to cytokines result in increased NOS activity, inhibition of GTP cyclohydrolase activity may prevent or reduce NO production by NOS. Thus compounds which inhibit GTP cyclohydrolase 1 activity may also inhibit NOS activity as a result of the inhibition or reduction in BH4 production.
Generally, inhibition, if any, of GTP cyclohydrolase 1, is determined by firstly measuring GTP cyclohydrolase 1 activity in the absence of the candidate compound. This provides a control value for GTP cyclohydrolase 1 activity under the assay conditions. Secondly, GTP cyclohydrolase 1 activity is measured in the presence of the candidate compound. The inhibition of GTP cyclohydrolase 1 activity due to the candidate compound can be determined by comparing the control value with the value obtained in the presence of the candidate compound. The assays are conducted by contacting GTP cyclohydrolase 1 with the candidate compound. The extent and/or the rate of reaction can be measured quantitatively, semi-quantitatively or qualitatively. If, for example, a fast throughput screening assay format is required, the control reaction in the absence of the candidate compound need not be performed for every candidate compound tested. Typically, the amount of GTP cyclohydrolase 1 used in the assay is from 1 to 10 ng of pure protein. Typically, the concentration of candidate compound used in the assay is from 1 μM to 1000 μM, preferably from 1 μM to 500 μM or 1 μM to 100 μM, more preferably from 1 μM to 10 μM.
GTP cyclohydrolase 1 activity can be measured, for example, by assessing the production of BH4 or GTP consumption. BH4 production can be determined, for example, by oxidation to biopterin using iodine and measurement of biopterin using HPLC (mobile phase is 5% methanol run at 1.5 ml/min on a 10 micron ODS reverse phase column (4.6 mm x 25 cm) with excitation at 350 nm and emission at 410 nm).
Therapeutic uses Over-production of nitric oxide as a result of, for example, the effects of bacterial endotoxins, can lead to potentially fatal vasodilatation and hypotension. Selective inhibition of nitric oxide synthases by a compound identified by the method of the invention may be useful in alleviating this potentially fatal response. Compounds identified by the method of the invention may be used therapeutically to treat conditions in which over-production of NO is implicated, for example septic shock, asthma, rheumatoid arthritis, inflammatory bowel disease, heart failure or acute systemic inflammation. NOS is also implicated in various neurological disease states including strokes, dementia and Parkinsons disease.
Similarly, compounds which inhibit or reduce synthesis of BH4 by GTP cyclohydrolase 1 may also be used therapeutically to treat the conditions described above in which NOS is implicated. Preferably, the compound is used to treat diseases in which eNOS is implicated.
The formulation will depend upon the nature of the compound identified but typically the compounds may be formulated for clinical administration by mixing them with a pharmaceutically acceptable carrier or diluent. For example they can be formulated for topical, parenteral, intravenous, intramuscular, subcutaneous, intraocular or transdermal administration. Preferably, the compound is used in an injectable form. It may therefore be mixed with any vehicle which is pharmaceutically acceptable for an injectable formulation, preferably for a direct injection at the site to be treated. The pharmaceutically carrier or diluent may be, for example, sterile or isotonic solutions. It is also preferred to formulate that compound in an orally active form. The dose of compound used may be adjusted according to various parameters, especially according to the compound used, the age, weight and condition of the patient to be treated, the mode of administration used and the required clinical regimen. As a guide, the amount of compound administered by injection is suitably from 0.01 mg/kg to 30 mg/kg, preferably from 0.1 mg/kg to 10 mg/kg.
The routes of administration and dosages described are intended only as a guide since a skilled practitioner will be able to determine readily the optimum route of administration and dosage for any particular patient and condition.
The invention will be described with reference to the following examples which are intended to be illustrative only and not limiting. In the accompanying drawings:
Figure 1 - Dose response curves to noradrenaline before and after (a) IL-lβ (n=5) (b) IL-lβ and TNFα (n=5) or (c) IL-lβ, TNFα and IL-6 (n=5)
(a) Dose response curves were constructed to noradrenaline before (o) and 1(D), 6 (D) and 24 h (A) after instillation of IL-lβ (1 ng).
(b) Dose response curves were constructed to noradrenaline before and 1, 6, and 24 h after instillation of IL-lβ and TNFα (1 ng) and IL-6 (100 pg).
(c) Dose response curves were constructed to noradrenaline before and 1, 6, 24 and
48h () after instillation of IL-lβ (1 ng), TNFα (1 ng) and IL-6 (100 pg). indicates p<0.05
(d) Dose response curves were constructed to noradrenaline before (o) and 1(D) and 6h (D) after instillation of IL-lβ. Six hours after instillation of IL-lβ a repeat dose-response curve was constructed with L-NMMA (1 μmol/min; - A) co-infused with noradrenaline this was followed by a saline washout period of 15 min and a repeat dose response curve to noradrenaline was constructed with L-arginine (1 μmol min - τ) co-infused with noradrenaline.
(e) Dose response curves were constructed to noradrenaline before (o) and 1 (D) and 6h (D) after instillation of IL-lβ. Six hours after instillation of IL-lβ a repeat dose- response curve was constructed with a aminoguanidine (1 μmol/min - x) co-infused with noradrenaline this was followed by a saline washout period of 15 min and a repeat dose response curve to noradrenaline was constructed with L-arginine (1 μmol/min - ▼) co-infused with noradrenaline.
Figure 2 - Panel A - Two adjacent vessels were studied simultaneously and at a distension pressure of 40 mmHg. The sympathetic nervous system was activated by asking subjects to take a deep breath. This manoeuvre produced transient constriction in both vessels. Incubation of one vessel (lower trace) with IL-lβ virtually abolished sympathetically mediated constriction 6 h later. Sympathetic constriction was restored by infusion of L-NMMA. Congesting cuff deflation and re-inflation is marked by solid circle. Panel B - Traces showing effects of L-NMMA on basal vessel tone in control vessel (upper trace) and vein treated with IL- lβ (lower trace). The effects of L-NMMA were reversed by L-arginine.
Figure 3 - Panel A - Infusion of BH4 for 20 min into a control vein causes sustained vasodilatation (hatched line; n=6). Co-infusion of aminoguanidine reverses the dilatation
(solid line; n=6).
Panel B - Original trace showing effects of BH4 infusion (upper trace) and reversal by aminoguanidine (lower trace). Congesting cuff deflation and re-inflation is marked by a solid circle.
Figure 4 - PCR reactions were carried out in 20 μl volumes, and 5 μl samples analysed by electrophoresis through 2% agarose gels.
Panel A shows the result with the iNOS primer pair BB52 and BB71. Track 1 size markers; tracks 2 & 3 niRNA from uninduced hand vein; track 4 and 5 mRNA from cytokine-induced hand vein; track 6 positive control (cytokine-induced CAPAN-1 mRNA). A PCR product of 499 bp is only visible in track 6.
Panel B shows the result of PCR amplification with the eNOS specific primer pair
BB158 and BB159. Tracks 1 & 2 mRNA from uninduced vein; tracks 3 & 4 mRNA from cytokine-induced vein; track 5 positive control (human placenta); track 6 size markers. Bands corresponding to eNOS can be detected in tracks 1-5. Panel C shows the β-actin specific PCR products from the primer set BB27 and BB28. Traces 1-4 are the hand vein mRNA samples amplified with an initial reverse transcriptase step. Tracks 5-8 are negative controls, amplified in the absence of a reverse transcriptase step. Track 9 is a positive control using, placental mRNA. Track 10 is size markers. Bands corresponding to β-actin can be detected in tracks 1-4 confirming that mRNA had been isolated from hand vein biopsies.
EXAMPLE
Materials and methods
In vivo pharmacology
Studies were approved by the local ethics committee and were performed on male and female subjects aged 19-40. Subjects were included who stated that they were healthy and on no medication, and who gave their informed, written consent. Subjects lay supine in a temperature-controlled laboratory (28-30°C). A contesting cuff was placed around the upper arm and inflated to 40 mmHg. Drugs or physiological saline were infused through a 23 gauge needle placed in a dorsal hand vein. The diameter of the vein was measured 5-10 mm downstream from the tip of the infusion needle by recording the linear displacement of a light-weight probe placed on the skin overlying the summit of the vessel when the pressure in the contesting cuff was lowered from 40 to 0 mmHg. In all studies saline was infused for at least 15 min until a stable baseline vein diameter was recorded. To instil cytokines, a length of the vein under study was isolated from the circulation by means of two wedges placed 2-3cm apart on the skin overlying the vessel as described previously, instilled for 1 h, either individually or together. At the end of the period of instillation, the wedges were removed and the vein was reconnected with the circulation for assessment of reactivity. This method of instillation produces local changes in the study vein but adjacent vessels remain unaffected. The volume of blood in the isolated vein is in the order of 1-2 ml and the calculated concentration of cytokine was in the order of 300-1000 pg/ml (TNFα and IL-lβ) and 30-100 pg/ml (IL-6). Biopsy
Surgical removal of the vein was done under local anaesthesia (1% lignocaine™). All samples were handled minimally and immediately frozen to -80°C.
PCR
Reverse transcriptase polymerase chain reaction (RT-PCR) was carried out on poly A" mRNA extracted from hand veins using Fast Track reagents (Invitrogen™). Positive control poly A+ mRNA for eNOS was carried out on human placental preparations (Clontech™). Positive controls for iNOS and GTPCH-1 were carried out on poly A+ mRNA isolated from the human pancreatic adenocarcinoma line CAPAN-1 stimulated with cytokines. The following primer sets were used:
β actin:
BB27 5'- gattgtgggtatgggtcagaagta-3' BB28 5'- gctcattgccgatagtgatgacct-3'
GTPCH-1 igcl 5'-ttggttatcttcctaacaag-3' igc2 5'-gtgctggtcacagttttgct-3'
iNOS
BB52 5' -tccatgcagacaacctt-3'
BB71 5' -gcctcgctctggaaaga-3'
eNOS
BB158 5' -cagtgtccaacatgctgctggaaatt-3' BB159 5' -taaaggtcttcttcctggtgatgcc-3'
PCR was carried out in thin-walled tubes using a Gene Amp™ RT PCR kit (Perkin-Elmer™) in a Gene Amp PCR system 9600 thermocycler (Perkin-Elmer). The following conditions were used: 95°C for lmin 45s, followed by 30 cycles of 95°C, 15s, 60°C, 30s. An elongation step of 72°C was carried out for 7 min at the end of the cycle.
Results Effects of inflammatory cytokines on the vasoconstrictor responses to noradrenaline
We have developed an in vivo model to explore mechanisms of septic vasodilatation in man (Bhagat et al, 1996). In this model, drugs or mediators are given into a single superficial vein in very low doses sufficient only to produce changes in the study vessel. This makes it possible to expose a vessel in situ to individual cytokines without initiating a systemic cytokine cascade.
First we sought to explore the effects of inflammatory cytokines on the vasoconstrictor responses to noradrenaline. Three cytokines implicated in the systemic cardiovascular response to sepsis were studied. Instillation of IL-lβ (1 ng in 1 ml of saline for 1 h) into a single superficial blood vessel caused a rightward shift in the dose response curve to noradrenaline and suppressed the maximal constriction achieved (Figure la). This effect developed over several hours and reached a maximum 6 h after exposure of the vessel to IL-l β. By 24 h the potency of noradrenaline was fully restored. Instillation of either tumour necrosis factor α (TNFα); 1 ng in 1 ml of saline), or IL-6 (100 pg, in 1 ml of saline) alone produced no significant change in the response to noradrenaline (data not shown), but co-instillation of these cytokines with IL-lβ (1 ng/ml) for 1 h caused a prolonged (>24 h) attenuation of the response to noradrenaline (Figure lb). The slowly developing hyporesponsiveness to noradrenaline was prevented by prior treatment of subjects with oral hydrocortisone (100 mg, given 2 h before instillation of cytokine; n=6, data not shown). These studies identify IL-lβ as a key cytokine inducing vascular changes in man and indicate that a therapeutically relevant dose of a glucocorticoid prevents the development of the vasorelaxation. Effect of NOS inhibitors on IL-lβ-induced hyporesponsiveness to noradrenaline.
To determine whether increased generation of NO contributed to the hyporesponsiveness to noradrenaline, we tested the effects of N0 monomethyl-L-arginine (L-NMMA; a standard NOS inhibitor) , and aminoguanidine (a compound that has been widely used as a selective inhibitor of iNOS). Both L-NMMA (1 μmol/min) and aminoguanidine (1 pmol/min) reversed the IL-lβ-induced attenuation in the dose response curve to noradrenaline (Figure 1). Infusion of the substrate for NOS (L-arginine; 1 μmol/min) reversed the effect of L-NMMA or aminoguanidine (Figure 1). In control veins (not treated with IL-lβ) L-NMMA (1 μmol/min) and aminoguanidine (1 μmol/min) were devoid of constrictor action (Figure 2B) and did not alter the response to noradrenaline (data not shown), confirming previous observations that these vessels do not generate NO basally. As expected, in control veins not exposed to cytokines, L-NMMA (1 μmol/min) inhibited the dilatation produced by the endothelium-dependent dilator bradykinin by 88%, whereas aminoguanidine (1 μmol/min) did not. Together these studies demonstrate that the hyporesponsiveness induced by IL-lβ is due to generation of NO. The time course and pharmacological profile of the response (prevention by glucocorticoids, reversal by L-NMMA and aminoguanidine) are consistent with expression of iNOS, and have been taken as indicative of such in many previous studies.
The potential physiological significance of the increase in NO synthesis induced by IL-lβ is illustrated in Figure 2 A. Instillation of cytokines virtually abolished endogenous venoconstriction due to transient activation of the sympathetic nervous system (achieved by asking subjects to take and hold a deep breath), sympathetic nervous system activity is a major determinant of venous tone in humans and sympathetic blockers cause profound venodilatation and postural hypotension. Our findings indicate that the profound venodilatation that accompanies sepsis or a systemic inflammatory response is likely to be mediated by an induction of basal generation of NO within the vessel wall that blunts the ability of the sympathetic nervous system to alter venous tone. Local infusion of L-NMMA did not alter control responses to sympathetic activation, but restored the ability of the sympathetic nervous system to cause venoconstriction in vessels exposed to IL-lβ (Figure 2 A). This explains the observation that systemic administration of L-NMMA does not alter venous pressure in healthy volunteers but increases it in patients with septic shock.
eNOS is sensitive to inhibition by an iNOS inhibitor, aminoguanidine. in the presence of
In a separate series of studies we found that infusion of BH4 (a co-factor for all 3 isoforms of NOS) into preconstricted healthy control veins caused rapid vasodilatation
(Figure 3). In contrast to bradykinin-induced vasodilatation, this dilator response to BH4 was fully reversed by infusion of aminoguanidine (1 μmol/min; Figure 3). Since control vessels would be expected to express eNOS, but not iNOS, and the vasodilatation produced by BH4 was rapid and consistent with activation of a constitutive enzyme, this finding apparently presents a conundrum. However, whilst aminoguanidine had little effect on the activity of purified human eNOS in the absence of BH4, as BH4 was added to the enzyme, aminoguanidine became an effective inhibitor (data not shown).
Furthermore, addition of BH4 to human endothelial cells in culture increased generation of
NO and this was inhibited by aminoguanidine. Thus, the ability of aminoguanidine to inhibit eNOS activity is dependent on the prevailing concentration of BH4.
IL-lβ induces expression of GTP cyclohydrolase 1 leading to increased generation of BH,.
The rate limiting enzyme for BH4 synthesis is GTP cyclohydrolase 1. This enzyme is transcriptionally regulated in response to cytokines or endotoxin, and its induction can be prevented by glucocorticoids. It is clear, therefore that the pharmacological changes we observed in the hand vein in vivo might be explained equally well by activation of eNOS by BH4 synthesised following induction of GTP cyclohydrolase 1, as by induction of iNOS. To explore further the mechanism of the functional induction of NOS activity we excised a portion of hand vein. As expected, mRNA encoding eNOS was detected in all of the samples (Figure 4). Messenger RNA encoding GTP cyclohydrolase 1 was detected in veins exposed to cytokines (Figure 4). In contrast, under identical conditions, no mRNA encoding iNOS was detected (Figure 4). Taken together, the simplest explanation of the pharmacological and molecular studies is that IL-lβ induces expression of GTP cyclohydrolase 1 which leads to increased generation of BH4 and consequent activation of eNOS. This interpretation is further strengthened by the finding that treatment of human cultured endothelial cells with IL-lβ leads to appearance of mRNA for GTPCH-1 but not iNOS and to induction of NO generation as assessed by measurement of nitrite in the culture medium. FACS analysis showed that the cells expressed eNOS protein but iNOS protein was not detected. It is not clear whether the mRNA for GTP cyclohydrolase 1 we detected in the hand veins was present in the vascular endothelium, the smooth muscle, or throughout the vessel wall, but it has been shown that BH4 generated in one cell can enter neighbouring cells to activate NOS. Neopterin levels are greatly increased in the plasma of patients with infections and activated human macrophages make significant amounts of neopterin and biopterin. Our study clearly identifies IL-lβ as a key cytokine causing physiologically significant vasodilatation in humans by increasing NO Generation. We suggest that the phenomenon is explained by transcriptional regulation of GTP cyclohydrolase 1 and the consequent activation of eNOS by BH4. eNOS is an endothelial enzyme that has not been detected in smooth muscle cells. An implication of our findings is that the endothelium can generate sufficient NO to cause profound vasodilatation and that expression of NOS throughout the much larger smooth muscle cell layer is not necessary to cause septic hypotension.
References
1. Charles, I. G. et al, (1996), Expression of human nitric oxide synthase isozymes. Methods in Enzymology 268, p 449-460.
2. Bhagat, K., Collier, J. and Vallance, P. (1996), Local venous responses to endotoxin in humans. Circulation 94, p490-497
3. Nitric Oxide - Part A - Sources and Detection of NO; NO Synthase (1996). Methods in Enzymology 268, L. Packer (Ed.) Academic Press.
4. Rees et al. (1989) Br. J. Pharmacol 96, p418-424)
5. Rees et al. (1990) Biochem. Biophys. Res. Commun. 173, p541-547

Claims

1. A method for identifying a compound which selectively inhibits a nitric oxide synthase (NOS) which method comprises:
(a) determining the inhibition of the activity of the NOS in the presence of a candidate compound and a first concentration of tetrahydrobiopterin (BH4);
(b) determining the inhibition of the activity of the NOS in the presence of the candidate compound and a second concentration of the BH4;
(c) determining whether the candidate compound inhibits the activity of the NOS to a different extent in (a) than in (b).
2. A method according to claim 1 wherein step (a) comprises
(i) determining the activity of said NOS in the absence of the candidate compound and in the presence of a first concentration of BH4; (ii) determining the activity of said NOS in the presence of the candidate compound and the first concentration of BH4; and (iii) comparing the activity of said NOS in steps (i) and (ii).
3. A method according to claim 1 or 2 wherein step (b) comprises
(i) determining the activity of said NOS in the absence of the candidate compound and in the presence of a second concentration of BH4; (ii) determining the activity of said NOS in the presence of the candidate compound and the second concentration of BH4; (iii) comparing the activity of said NOS in steps (i) and (ii).
4. A method according to claim 1, 2 or 3 wherein said second concentration is lower than said first concentration.
5. A method according to any one of the preceding claims wherein said NOS is human eNOS.
6. A method according to any one of claims 1 to 5 wherein said NOS is human iNOS.
7. A method according to any one of claims 1 to 5 wherein said NOS is human nNOS.
8. Use of a compound identified by the method of any one of the preceding claims in the manufacture of a medicament for use in selectively inhibiting a human nitric oxide synthase.
9. Use according to claim 8 wherein said NOS is eNOS.
10. Use according to claim 8 wherein said NOS is iNOS.
11. Use according to claim 8 wherein said NOS is nNOS.
12. Use of a compound identified by the method of any one of claims 1 to 7 in the manufacture of a medicament for use in treating toxic shock syndrome.
13. A method for identifying a compound which is an inhibitor of GTP cyclohydrolase 1- mediated synthesis of BH4 which method comprises:
(i) contacting GTP cyclohydrolase 1 with a candidate compound; and (ii) determining the inhibition of GTP cyclohydrolase 1 -mediated synthesis of BH4 by said compound.
14. Use of a compound in the manufacture of a medicament for use in inhibiting a human nitric oxide synthase wherein said compound is capable of inhibiting or reducing synthesis of BH4 by GTP cyclohydrolase 1.
15. Use according to claim 14 wherein said compound is identified by an assay method which method comprises:
(i) contacting GTP cyclohydrolase 1 with a candidate compound; and (ii) determining the inhibition of GTP cyclohydrolase 1 -mediated synthesis of BH4 by said compound.
16. Use according to claim 14 or 15 wherein said NOS is eNOS.
17. A compound identified by the method of any one of claims 1 to 7 or 13.
18. A compound according to claim 17 for use in therapy.
19. A compound according to claim 18 for use in treating toxic shock syndrome.
20. A compound according to claim 17 for use in the manufacture of a medicament for prophylaxis or treatment of a clinical condition caused by excessive NO production.
21. A pharmaceutical composition comprising a compound according to any one of claims 17 to 20 together with a pharmaceutically acceptable carrier or diluent.
22. A method for preventing or treating a clinical condition in a patient caused by excessive NO production which method comprises administering an effective amount of a compound according to any one of claims 17 to 20 or a pharmaceutical composition according to claim 21, to said patient.
PCT/GB1998/000353 1997-02-05 1998-02-05 Method for identifying selective inhibitors of nitric oxide synthase WO1998035055A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP98903134A EP0961835A1 (en) 1997-02-05 1998-02-05 Method for identifying selective inhibitors of nitric oxide synthase
AU59962/98A AU5996298A (en) 1997-02-05 1998-02-05 Method for identifying selective inhibitors of nitric oxide synthase
JP53399198A JP2001511009A (en) 1997-02-05 1998-02-05 Methods for identifying selective inhibitors of nitric oxide synthase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB9702312.1A GB9702312D0 (en) 1997-02-05 1997-02-05 Method for identifying selective inhibitors of nitric oxide synthase
GB9702312.1 1997-02-05

Publications (1)

Publication Number Publication Date
WO1998035055A1 true WO1998035055A1 (en) 1998-08-13

Family

ID=10807100

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1998/000353 WO1998035055A1 (en) 1997-02-05 1998-02-05 Method for identifying selective inhibitors of nitric oxide synthase

Country Status (5)

Country Link
EP (1) EP0961835A1 (en)
JP (1) JP2001511009A (en)
AU (1) AU5996298A (en)
GB (1) GB9702312D0 (en)
WO (1) WO1998035055A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999059566A1 (en) * 1998-05-15 1999-11-25 Glaxo Group Limited Use of nitric oxide synthase inhibitors in the manufacture of a medicament for the prophylaxis or treatment of bacterial infection
US7906520B2 (en) 2003-11-13 2011-03-15 The General Hospital Corporation Methods for treating pain

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995013803A1 (en) * 1993-11-15 1995-05-26 Cornell Research Foundation, Inc. Blocking induction of tetrahydrobiopterin to block induction of nitric oxide synthesis

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995013803A1 (en) * 1993-11-15 1995-05-26 Cornell Research Foundation, Inc. Blocking induction of tetrahydrobiopterin to block induction of nitric oxide synthesis

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
A.C.F. GORREN, B.M. LIST, A. SCHRAMMEL, E. PITTERS, B. HEMMENS, E.R. WERNER, K. SCHMIDT, B. MAYER: "Tetrahydrobiopterin-Free Neuronal Nitric Oxide Synthase: Evidence for Two Indentical Highly Anticooperative Pteridine Binding Sites", BIOCHEMISTRY, vol. 35, no. 51, 1996, pages 16735 - 16745, XP002066036 *
B. M. LIST, P. KLATT, E. R. WERNER, K. SCHMIDT, B. MAYER: "Overexpression of neuronal nitric oxide synthase in insect cells reveals requirement of haem for tetrahydrobiopterin binding", BIOCHEMICAL JOURNAL, vol. 315, no. 1, 1 April 1996 (1996-04-01), pages 57 - 63, XP002066037 *
G. WERNER-FELMAYER, E.R. WERNER, D. FUCHS, A. HAUSEN, G. REIBNEGGER, K. SCHMIDT, G. WEISS, H. WACHTER: "Pteridine Biosynthesis in Human Endothelial Cells", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 268, no. 3, 25 January 1993 (1993-01-25), pages 1842 - 1846, XP002066039 *
P. KLATT, K. SCHMIDT, G. URAY, B. MAYER: "Multiple Catalytic Functions of Brain Nitric Oxide Synthase", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 268, no. 20, 20 July 1993 (1993-07-20), pages 14781 - 14787, XP002066038 *
P. KLATT, M. SCHMID, E. LEOPOLD, K. SCHMIDT, E. R. WERNER, B. MAYER: "The Pteridine Binding Site of Brain Nitric Oxide Synthase", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 269, no. 19, 13 May 1994 (1994-05-13), pages 13861 - 13866, XP002066035 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999059566A1 (en) * 1998-05-15 1999-11-25 Glaxo Group Limited Use of nitric oxide synthase inhibitors in the manufacture of a medicament for the prophylaxis or treatment of bacterial infection
US7906520B2 (en) 2003-11-13 2011-03-15 The General Hospital Corporation Methods for treating pain

Also Published As

Publication number Publication date
EP0961835A1 (en) 1999-12-08
AU5996298A (en) 1998-08-26
JP2001511009A (en) 2001-08-07
GB9702312D0 (en) 1997-03-26

Similar Documents

Publication Publication Date Title
Inoue et al. Lipophilic HMG-CoA reductase inhibitor has an anti-inflammatory effect: reduction of mRNA levels for interleukin-1β, interleukin-6, cyclooxygenase-2, and p22phox by regulation of peroxisome proliferator-activated receptor α (PPARα) in primary endothelial cells
Bhagat et al. Cytokine-induced venodilatation in humans in vivo: eNOS masquerading as iNOS
Daujat et al. [33] Induction protocols for cytochromes P450IIIA in Vivo and in primary cultures of animal and human hepatocytes
Greenblatt et al. Antipyrine kinetics in the elderly: prediction of age-related changes in benzodiazepine oxidizing capacity.
Ishii et al. Induction of matrix metalloproteinase gene transcription by nitric oxide and mechanisms of MMP‐1 gene induction in human melanoma cell lines
Berger et al. Cardiac prostaglandin release during myocardial ischemia induced by atrial pacing in patients with coronary artery disease
Lorthioir et al. Polycystin deficiency induces dopamine-reversible alterations in flow-mediated dilatation and vascular nitric oxide release in humans
Wu et al. IL-1β upregulates Muc5ac expression via NF-κB-induced HIF-1α in asthma
JP7018470B2 (en) Biomarkers useful for treating subjects with eye diseases
Stratman et al. Ibuprofen: effect on inducible nitric oxide synthase
Richer et al. Captopril: pharmacokinetics, antihypertensive and biological effects in hypertensive patients.
McClarty et al. Elevated expression of M1 and M2 components and drug-induced posttranscriptional modulation of ribonucleotide reductase in a hydroxyurea-resistant mouse cell line
Dinc et al. Methylene blue inhibits the inflammatory process of the acetic acid-induced colitis in rat colonic mucosa
Chan et al. Reactive oxygen species mediate endotoxin-induced human dermal endothelial NF-κB activation
Coutinho et al. A role for spinal nitric oxide in mediating visceral hyperalgesia in the rat
Jones et al. Hydrogen peroxide—an intracellular signal in the pulmonary circulation: involvement in hypoxic pulmonary vasoconstriction
WO1998035055A1 (en) Method for identifying selective inhibitors of nitric oxide synthase
WO2000057871A2 (en) Methods for identifying agents that inhibit serum aging factors and uses and compositions thereof
US20210100780A1 (en) Methods for treating wild type isocitrate dehydrogenase 1 cancers
Naureckiene et al. Use of a murine cell line for identification of human nitric oxide synthase inhibitors
US7588885B2 (en) Use of biocistronic DNA constructs for identifying compounds that inhibit IRES-dependent translation
Cuzzocrea et al. The tyrosine kinase inhibitor tyrphostin AG 126 reduces the development of colitis in the rat
Noguchi et al. Evaluation of a rat model of functional urinary bladder outlet obstruction produced by chronic inhibition of nitric oxide synthase
Kang et al. Role of endogenous renin-angiotensin system in c-fos activation and PKC-epsilon translocation in adult rat hearts
Sarafoff et al. Clozapine induced increase of human plasma norepinephrine

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

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

Ref country code: JP

Ref document number: 1998 533991

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1998903134

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1998903134

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 1998903134

Country of ref document: EP