AU2004294800A1 - Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour - Google Patents

Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour Download PDF

Info

Publication number
AU2004294800A1
AU2004294800A1 AU2004294800A AU2004294800A AU2004294800A1 AU 2004294800 A1 AU2004294800 A1 AU 2004294800A1 AU 2004294800 A AU2004294800 A AU 2004294800A AU 2004294800 A AU2004294800 A AU 2004294800A AU 2004294800 A1 AU2004294800 A1 AU 2004294800A1
Authority
AU
Australia
Prior art keywords
use according
prostaglandin
labour
female
cells
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.)
Abandoned
Application number
AU2004294800A
Inventor
Phillip Robert Bennett
Tamsin Lindstrom
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.)
Ip2ipo Innovations Ltd
Original Assignee
Imperial College Innovations Ltd
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
Priority claimed from GBGB0327975.9A external-priority patent/GB0327975D0/en
Application filed by Imperial College Innovations Ltd filed Critical Imperial College Innovations Ltd
Publication of AU2004294800A1 publication Critical patent/AU2004294800A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • A61K31/5575Eicosanoids, e.g. leukotrienes or prostaglandins having a cyclopentane, e.g. prostaglandin E2, prostaglandin F2-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/06Antiabortive agents; Labour repressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Reproductive Health (AREA)
  • Epidemiology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Endocrinology (AREA)
  • Virology (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Description

WO 2005/053706 PCT/GB2004/005087 USE OF A CYCLOPENTENONE PROSTAGLANDIN FOR DELAYING FOR THE ONSET AND/OR PREVENTING THE CONTINUATION OF LABOUR The present invention relates to agents for improving perinatal outcome in pre-term labour. In particular, the present invention relates to the use of 5 prostaglandins to prevent and/or reduce an inflammatory response in the reproductive system of a female, thereby delaying the onset of labour. Human pre-term labour, defined as spontaneous labour occurring prior to 37 weeks of gestation (with 39 weeks being term) continues to be a major 10 problem, particularly in developed countries. Preterm birth occurs in 5-10% of all pregnancies but is associated with 70% of all neonatal deaths and up to 75% of neonatal morbidity (Rush et al., 1976). Premature neonates are at high risk of cerebral palsy, developmental delay, visual and hearing impairment and chronic lung disease. 15 During pregnancy, the uterus is maintained in a state of non-contractile quiescence whilst the cervix remains firm and closed. With the onset of labour, the cervix needs to become softer and to offer low resistance to force applied and have fibres which move under tension. The uterus also 20 needs to begin contracting. Both at term and preterm, the biochemistry of labour resembles an inflammatory reaction and there is accumulating evidence pointing to a pivotal role for pro-inflammatory cytokines and prostaglandins (PGs) in the 25 labour process. Interleukin-1 IP (IL-1 IP) levels are elevated in amniotic fluid (Romero et al., 1990), gestational membranes (Keelan et al., 1999; Elliot et al., 2001) and the lower uterine segment (Maul et al., 2002) at term labour, and may contribute to labour onset by stimulating IL-8 and PG synthesis (Mitchell et al., 1990; Brown et al., 1998). PGs increase in maternal urine 30 and blood and in fetal membranes in association with labour (Satoh et al., 1979; Skinner and Challis, 1985). PGE 2 stimulates uterine contractions 1 WO 2005/053706 PCT/GB2004/005087 (Dyal and Crankshaw, 1985), indirectly increases fundamentally dominant myometrial contractility by upregulation of oxytocin receptors and synchronisation of contractions (Garfield et a[., 1990), and acts in concert with IL-8 to remodel the cervix (reviewed in Kelly, 2002). 5 The onset of labour is associated with activation of the Nuclear Factor Kappa B (NFicB) transcription factor system in the amnion which plays a role in the expression of pro-inflammatory genes such as interleukin-8 (IL 8), interleukin-6 (IL-6) and cyclo-oxygenases 1 and 2 (COX-1 and COX-2). 10 COX genes are also referred to as prostaglandin H synthase or PG synthase. The resulting inflammatory infiltrate (mediated by the cytokines) and increase in prostaglandin synthesis (mediated by the cyclo-oxygenases) leads to cervical ripening, fetal membrane rupture and myometrial contractions. 15 Five members of the NF-icB/Rel family have been identified in mammals: NF-1cB1 (p50 and its precursor p105), NF-1cB2 (p52 and its precursor p100), p65 (ReIA), c-rel, and Rel B. These proteins share a structurally conserved amino-terminal region termed the Rel homology domain (RHD). The RHD 20 is responsible for dimerisation, DNA binding, and interaction with the inhibitors of kappa B (IicB) proteins. It also contains a nuclear localisation signal (NLS). In its active DNA-binding form NF-1cB consists of heterogeneous dimers of various combinations of NF-KB subunits: each member of the NF-1cB family, except for Rel B, can fonn homodimers, as 25 well as heterodimers with one another. The p 6 5, c-rel and Rel B proteins contain a carboxy-teininal non-homologous transactivation domain, which activates transcription from icB sites in target genes; in contrast, p50 and p52 proteins lack a transactivation domain. The various NF-icB diners exhibit different binding affinities for specific xB sites (Kunsch et al., 1992, 2 WO 2005/053706 PCT/GB2004/005087 Phelps et al., 2000), and differentially stimulate transcription through distinct icB elements (Lin et al., 1995). In resting cells, NF-icB diners are normally sequestered in an inactive form S in the cytoplasm by association with the inhibitory IicB proteins, which include IicB a, IkB P and IB s. The IicB s are characterized by the presence of multiple ankyrin repeats which mediate binding to the RHD and mask the NIL S of NF-1cB. 10 The major NF-icB signalling pathway, which is activated by pro inflammnatory stimuli and LPS, targets IlcBa- and IxBp-bound NF-icB (for review see Li and Verma 2002). p5O/p65 dimers are the most abundant form of NF-icB in most cell types, and activation of IkB-bound p50/p65 dimers is the best characterised pathway. In this 'classical' pathway, 15 diverse stimuli trigger signal transduction cascades that ultimately converge on the activation of a specific IcB kinase (IKK). The IKK complex consists of several proteins, the main ones being IKKa. (IKKl), IKKO (IKK2), and NF-KB essential modulator (NEMO or IKKy). The activated IKK complex phosphorylates IBa at shrines 32 and 36, which results in the poly 20 ubiquitination of IKBa at lysines 21 and 22. This modification targets IlcBa for rapid degradation by the 26S proteasome. The degradation of the IlcB inhibitor exposes the NLS of NF-icB resulting in translocation of the p50/p65 dimer to the nucleus where it can bind to B sites in the promoter of target genes and promote transcription. 25 Most stimuli cause only the transient activation of NF--KB. The critical inhibitory step in NF-icB inactivation involves binding of newly synthesised IicBa to NF-4B in the nucleus. IxBa is quickly resynthesised following its degradation. The newly synthesised IcBa is localised in the nucleus and 30 displaces NFicB from its DNA binding sites. IKBa contains leucine-rich 3 WO 2005/053706 PCT/GB2004/005087 nuclear export sequences (NES) (Johnson et al 1999), which then enable it to transport NF-KB back to the cytoplasm, thereby completing an autoregulatory post-induction repression. 5 In many cells nearly half of the NF-ixB is sequestered by the other major IxB isoform, IcB (Whiteside et al., 1997). In contrast to IiBa, IiBp is not NF-kB inducible and does not exert a rapid post-induction repression of NF-xB activity. Rather, IkBp has been implicated in persistent NF-kB activation. Prolonged exposure to certain stimuli, such as LPS, leads to the 10 long-term induction of NF-KB activity despite high levels of newly synthesised IcBa. Following stimulus-induced degradation, the newly synthesised IiBp is un-phosphorylated and, in contrast to lIBa or the constitutively phosphorylated IKBp, can interact with NF-idB bound to target promoters without displacing it from the DNA (Suyang et al., 1996). 15 This interaction of un-phosphorylated IKB [ with DNA-bound NF-icB is thought to protect NF-KB from nuclear export, and thus inhibition, by IxB a, and the outcome is a sustained NF-KB response. PGs are a family of biologically active molecules having a diverse range of 20 actions depending on the prostaglandin type and cell target. There is considerable evidence to support a central role for PGs in human parturition. Labour is associated with increased PG synthesis within the uterus (Turnbull 1977) particularly from the fetal membranes (Skinner and Challis 1985). PGs act to mediate cervical ripening and to- stimulate uterine 25 contractions (Cranckshaw and Dyal 1994) and indirectly to increase fundamentally dominant myometrial contractility 'by up-regulation of oxytocin receptors and synchronisation of contractions (Garfield et al 1990). PG synthesis in amnion, chorion-decidua and myometrium increases with labour (for a review, see Bennett and Slater 1996). Chorion 30 prostaglandin dehydrogenases are thought to protect the uterus from basal 4 WO 2005/053706 PCT/GB2004/005087 prostaglandin synthesis during pregnancy but are down-regulated at term. Deficiency of prostaglandin dehydrogenase in chorion has been associated with pre-term labour (van Meir 1996, 1997). 5 Accordingly, inhibition of prostaglandin synthesis is an effective method of preventing or arresting pre-term labour (Keirse, 1995). Conversely, prostaglandins have been administered to induce labour as a means to terminate pregnancy (Ganstrom et al., 1987). 10 Most PGs bind to prostanoid receptors localised on the cell surface and act through second messenger systems (Narumiya, 1995). However, PGD2 metabolites are actively incorporated into the nuclei of cells (Narumiya et al., 1987) and can exert their effects through direct interactions with nuclear receptors. Peroxisome proliferator-activated receptors (PPARs) are ligand 15 activated transcription factors belonging to the nuclear receptor superfamily. They exist in three distinct forms, PPAR-a, PPAR-S, and PPAR-y, which forn heterodimers with the retinoic X receptor (RXR) and bind to PPAR response elements (PPREs) in the promoter of target genes to induce transcription. PPAR-y can also repress gene transcription by 20 negatively interfering with the NF-iB, AP-1, STAT and C/EBP pathways (Zhou et al, 1999; Subbaranaiah et al., 2001; Takata et al., 2002; Suzawa et al., 2003). The aetiology of pre-term labour is multi-factorial but bacterial infection is 25 believed to play an important role, especially at earlier gestational ages (for review see Romero et al., 2002). A growing body of epidemiological data suggests that intrauterine infection is an important cause of brain injury in infants born before 32 weeks of gestation. During ascending intrauterine infection, micro-organisms can stimulate the production of pro 30 inflammatory cytokines, such as tumour necrosis factor a (TNFa) and IL 1p, as well as PGs and other inflammatory mediators, resulting in the WO 2005/053706 PCT/GB2004/005087 premature onset of labour. Intrauterine infection/inflammation has also been identified as a key contributor to the development of cerebral palsy (CP) and schizophrenia (Urakubo et al., 2001; Gibson et al., 2003), and, although CP does occur in term infants, the risk of CP is strongly associated 5 with prematurity (Dammann et al., 1999). In addition, inflammatory responses caused by mechanical stretching of the uterus may contribute to the onset of labour. Mechanical stretching of the uterus occurs to an extent as a normal part of pregnancy and may be 10 responsible for some of the biochemical changes which occur near to term and which cause the nonral onset of labour at term. In the context of preterm labour, mechanical stretch may occur where the uterus is overdistended by multiple pregnancy or by excess amniotic fluid (clinically termed hydramnios or polyhydrainios). There may also be more local 15 stretch of the lower segment of the uterus, the cervix and overlying fetal membranes -in cases where there is cervical weakness (clinically termed cervical incompetence). Stretch leads to an increase in the production of a series of 'labour-associated' proteins including COX-2 (which then increases prostaglandin synthesis), cytokines such as IL-8 and IL-lb and the 20 oxytocin receptor. Increased prostaglandin and cytokine productions causes cervical ripening or further cervical ripening (and may lead to neonatal brain injury). Prostaglandins and OTR receptor lead to uterine contractions. Obstetric management of pre-term labour is still largely reactive and centred 25 on the use of drugs intended to inhibit contractions to delay delivery. This was thought to be principally dependent upon gestational age leading to the concept that prolongation of the pregnancy will always improve outcome. However, there is now growing evidence that the mechanisms leading to pre-tenn birth also cause fetal cerebral damage. Characteristically, damage 30 is localised to the white matter, involving both a diffuse astrogliosis with subsequent loss of myelin-producing oligodendrocytes, as well as 6 WO 2005/053706 PCT/GB2004/005087 multifocal necroses resulting in cystic change (periventricular leucomalacia, PVL). Such lesions lead to cerebral palsy in 60-90% of affected infants (described in Vlope, 2001). 5 There are currently no drugs available which will safely and effectively inhibit pre-term contractions. The most commonly used agents, f-sympathomimetics such as Ritodrine, Salbutamol and Terbutaline, cause significant maternal cardiovascular, respiratory and metabolic side effects and may lead to pulmonary oedema, cardiac failure and maternal death. 10 Furthermore they are subject to tachyphylaxis and become ineffective after 24 to 48 hours. Meta-analysis of randomised controlled trials has shown that the value of p-sympathomimetics is only in the temporary delay of labour to allow in utero transfer or administration of steroid to improve fetal lung surfactant production. 15 Other than the antenatal administration of corticosteroids, no obstetric interventions affect neonatal outcome although improvements in neonatal intensive care have dramatically increased survival rates. Commonly used agents are dexamethasone or betamethasone. Antenatal administration of 20 corticosteroids improves the outcome for the pre-tenn neonate since it reduces the incidence and severity of respiratory distress syndrome, intracranial haemorrhage and necrotising enterocolitis. One function of corticosteroids is to mature the fetal lung, which leads to an increase in surfactant production and therefore prevents or reduces the severity of 25 neonatal respiratory problems. Such agents are known to those skilled in the art. Current obstetric management of pre-term labour (or threatened pre-term labour or pre-tern premature rupture of membranes) is to attempt to delay 30 delivery using 'tocolytic' drugs to allow time for steroid administration. 7 WO 2005/053706 PCT/GB2004/005087 Typically, effective tocolytic drugs are oxytocin receptor antagonists, calcium channel blockers, sympathomimetics and nitric oxide donors. A commonly used oxytocin receptor antagonist is Atosiban, that functions 5 by blocking the oxytocin receptor, thereby preventing activation of the receptor by endogenous oxytocin that stimulates uterine contractions. A comnmonly used calcium channel blocker is Nifedipine, that functions to block the influx of calcium into the myometrial cells, which is a requirement for contractions to take place. A comnmonly used 10 sympathomimetic is Ritodrine, that functions by activating adrenergic receptors on the myocyte cell membrane leading to phosphorylation and down-regulation of the activity of myosin light chain kinase, an enzyme essential for contractions. A commonly used nitric oxide donor is glyceryl trinitrate, that functions by increasing myocyte cGMP thereby down-regulating 15 the activity of myosin light chain kinase, an enzyme essential for contractions. Indomethacin, a cyclo-oxygenase inhibitor, is effective in preventing the contractions of pre-term labour. It is more effective in short term 20 prolongation of pregnancy than the p-sympathomimetics and, unlike P-sympathomimetics, it can reduce the risk of delivery pre-term (Keirse 1995). The use of indomethacin is limited by fetal side effects. Indomethacin reduces fetal urine output and constriction of the ductus arteriosus (Moise et al 1995). Clinically significant ductal constriction 25 occurs only in a proportion, increasing with gestational age from 10% at 26 weeks to 50% at 32 weeks. Accordingly the use of indomethacin is limited in clinical practice to use < 32 weeks, and to short courses (5 48 hours) after which any effects on the constriction of the ductus have been shown to be reversible (Tulzer et al 1991; Moise et al 1993; Respondek et al 1995). 30 8 WO 2005/053706 PCT/GB2004/005087 Because of these side effects some obstetricians now use Sulindac, which appears to be equally good as a tocolytic (Carlon et at 1992) in place of indomethacin. Sulindac produces a smaller reduction in fetal urine output and minimal effect on ductal patency (Carlon et al 1992; Rasanen and 5 Jouppila 1995). However, Sulindac is far from an ideal choice of tocolytic agent. Accordingly, new agents or regimens capable of reducing and/or preventing an inflammatory response in the reproductive system of a female are highly 10 desired. Such medicaments or approaches would allow the treatment of pathogenic infection within the reproductive system of a female and/or delay pre-term delivery without causing injury to the fetus/neonate. In light of the above, the present inventors have surprisingly discovered that 15 prostaglandins can be used to delay the onset and/or prevent the continuation of labour in a female. Thus, in a first aspect, the present invention provides the use of a cyclopentenone prostaglandin in the manufacture of a medicament for 20 delaying the onset and/or preventing the continuation of labour in a female. Preferably, this is achieved by preventing and/or reducing an inflammatory response in the reproductive system of a female. 25 The invention sterns from the unexpected finding that the cyclopentenone prostaglandins, such as 15-deoxy-A'1 4 prostaglandin J 2 (15-dPGJ 2 ) and prostaglandin A 1
(PGA
1 ), inhibit and/or reduce NFicB activity within uterine cells of the female reproductive system. Thus, cyclopentenone prostaglandins provide a means for the inhibition and/or reduction of NFicB 30 activity in the reproductive system of a female. Medicaments of the invention are believed to inhibit cytokine synthesis and inhibit the 9 WO 2005/053706 PCT/GB2004/005087 biochemical processes of labour, thereby safely prolonging pregnancy. Accordingly, the present invention will improve obstetric management of pre-term labour as the onset of labour may be delayed without injuring the fetus/neonate. The cyclopentenone prostaglandins are naturally-occurring substances that contain a cyclopentenone ring structure. The cyclopentenone ring is characterised by the presence of a chemically-reactive oa,p-unsaturated carbonyl and is formed by dehydration of the cyclopentane ring of a 10 precursor prostaglandin. Generally, the first step in the biosynthesis of prostaglandins involves the intracellular release of arachidonic acid from plasma membrane phospholipids via the action of phospholipase A 2 . Arachidonic acid is then 15 converted sequentially to PGG 2 and PGH 2 by the cyclo-oxygenase and peroxidase activities of the PGH synthases, PGH 1 and 2. The prostaglandins PGE 2 , PGD 2 and PGF 2 , are subsequently synthesised from PGH2 via the action of the PGE2, PGD 2 and PGF 2 , synthase, respectively. The cyclopentenone prostaglandins, prostaglandin A 2
(PGA
2 ), prostaglandin 20 A 1
(PGA
1 ) and prostaglandin J 2
(PGJ
2 ) are formed by dehydration of prostaglandin E 2
(PGE
2 ), prostaglandin E 1 (PGE1) and prostaglandin D 2
(PGD
2 ), respectively. PGJ 2 is metabolised further to A1 2 -prostaglanding J 2
(A
1
-PGJ
2 ), and 15-deoxy-A 2 4 prostaglandin J 2 (15-dPGJ 2 ). 25 Other unnatural or synthetic prostaglandins can be made by chemical synthesis. Total synthesis of prostaglandins was first accomplished by Corey in the 1960s (reviewed in Corey, 1991), and subsequently simplified by Suzuki et al. (1990). This latter scheme uses a C8 organometallic reagent for one side chain and a C7 acetylenic halide for the other side chain 30 which are added to the desired chemical head-group. This synthesis is versatile and allows the synthesis of a variety of natural and unnatural 10 WO 2005/053706 PCT/GB2004/005087 prostaglandins including the cyclopentenone prostaglandins. A general pathway for natural and chemical synthesis of prostaglandins and cyclopentenone prostaglandins is described in Straus and Glass (2001), the disclosure of which is incorporated herein. Chemical modification of cyclopentenone prostaglandins using techniques known in the art of chemistry may alter the clinical effectiveness of the molecule. Such alterations may, for example, increase or decrease the stability or another characteristic of the cyclopentenone prostaglandin, to 10 give a desired change in activity. For example, modification of the 15C residue of cyclopentenone prostaglandins will reduce the metabolism of the compound, thereby increasing its half-life in vivo. Such modifications will be appreciated by those skilled in the art. 15 Thus, by "cyclopentenone prostaglandin", we include any natural, unnatural or chemically-modified prostaglandin which has a cyclopentenone ring. Cyclopentenone prostaglandin is often abbreviated to "cyPG". Especially preferred cyclopentenone prostaglandins include prostaglandin D 2
(PGD
2 ) and its metabolite 15-deoxy-A' 2
"
4 prostaglandin J 2 (15-dPGJ2). Also 20 preferred is prostaglandin A 1 (PGA1). 15-dPGJ 2 may be obtained from Cayman Chemical, 1180 East Ellsworth Road, Ann Harbour, MI 48108 USA (catalogue number 18570); 9,10-di hydro-15-deoxy-A 12
"
4 -Prostaglandin J 2 may be obtained from Alexis 25 Biochemicals Ltd, PO Box 6757, Bingham, Nottingham, NG13 8LS, UK (catalogue number CAY-18590-MOO1). PGA 1 may be obtained from Alexis Biochemicals Ltd (address as above; catalogue number 340-045 M005). 30 By "onset of labour" and/or "continuation of labour" we include the biochemical and/or physiological changes associated with preparation of the 11 WO 2005/053706 PCT/GB2004/005087 tissues of the female reproductive system for delivery. For example, the uterus increases in contractility and undergoes contractions. The cervix also ripens in readiness for delivery. Such changes are well known in the arts of obstetrics, gynaecology and midwifery and, for example, the Bishop's score 5 indicates the degree of cervical ripening (described in Herman et aL., 1993). By "delaying the onset of labour in a female and/or preventing the continuation of labour in a female" we include the meaning that at least one of these biochemical and/or physiological changes are delayed or prevented. 10 By "female" we include any female mammal such as human, or a domesticated mammal, preferably of agricultural significance including a horse, pig, cow, sheep, dog and cat. It is preferred if the female is a human female. 15 In a second aspect, the, present invention provides the use of a cyclopentenone prostaglandin in the manufacture of a medicament for preventing and/or reducing an inflamnmatory response in the reproductive system of a female. Such medicaments are able to inhibit and/or reduce NFicB activity in uterine cells. 20 By "NFicB" we include homo- and heterodimers of RelA (p65), RelB, NFicB1 (p50), NF 1 B2 (p52) and cRel. The ReIA (p65), RelB, NFil (p50), NFiB2 (p52). and cRel genes and the sequence of the polypeptide products are described in Li et al. (2002). 25 By "NFxB activity" we include the activities of NFrB associated with the expression of genes controlled by any homo- or heterodimer of RelA (p65), RelB, NFKB 1 (p50), NFKB2 (p52) or cRel of the NFicB transcription factor family. In particular, we include: nuclear translocation of NFicB which can 30 be measured, for example, by Western blotting analysis of nuclear and cytosolic cellular fractions for the protein of interest (described in 12 WO 2005/053706 PCT/GB2004/005087 Sambrook et al., 1989; Lee et al., 2003); binding of NFxB to target nucleic acid sequences (such as specific regions and sequences of DNA), which can be measured, for example, by Electro-Mobility Shift Assay (EMSA, as described in Dignam et al., 1983; Lee et al., 2003); and NFlB-mediated 5 expression of target genes which can be measured, for example, by northern blotting and/or Western blotting (Sambrook et al., 1989; Lee et al., 2003). Methods for measuring these activities of NFkB are well known by those skilled in the art of biochemistry and molecular biology. 10 By "uterine cells" we include any cells within the uterus of a female, or cells derived from the uterus of a female, particularly placental cells, amnion cells, myocytes, uterine and cervical fibroblasts, and maintained as a primary or transformed cell culture or line. These cell types are typically referred to as "gestational tissues". 15 Cultures of amnion cells may be prepared from tissue by separating the entire anirion, except for the part overlying the placenta, from the chorion, followed by separating ainion epithelial cells from fibroblasts and maintaining the epithelial cells using mammalian cell culture techniques 20 (Lee et al., 2003). Myometrial cell culture may be prepared from tissue from the lower uterine segment, separating cells by incubation with Dispase and collagenase/elastase/DNAase solution and maintaining the myometrial cells using mammalian cell culture techniques (Pieber et al., 2001). Techniques for the generation and maintenance of primary and transformed 25 mamnmalian cell cultures will be well known to those skilled in the relevant art. By "reproductive system of a female", we include any cells and/or tissues and/or organs of a female directly or indirectly involved in the fonnation, 30 nourislunent, maintenance and development of a neonate, embryo or fetus at any gestational stage during pregnancy. In particular we include the cells 13 WO 2005/053706 PCT/GB2004/005087 and/or tissues of the uterus, placenta, ainion, chorion, decidua, cervix and vagina. Preferably, the medicament is for preventing and/or reducing an 5 inflammnatory response in the reproductive system of a female that is pregnant. By "inflammatory response" we include biochemical and physiological changes associated with inflammation mediated by cells of the host's 10 immune system. Such changes are known in the arts of human and veterinary medicine, immunology, molecular biology and biological science. If a patient is detected clinically at being at high risk of preterm delivery, 15 because of detection of fibronectin in the vagina, identification of cervical shortening on ultrasound, the identification on clinical examination of cervical dilatation, or the onset of contractions then there is a high risk that there may be inflammation within the uterus. Other clinical measures of inflamnation within the uterus are maternal temperature, white blood cell 20 count, serum c-reactive protein concentrations and amniotic cytokine concentrations (taken at amniocentesis) which suggest a high risk of inflammation within the uterus if abnormal. Methods for measuring such changes will be well known to those skilled in the art. 25 By "pregnant", we include the meaning that the female is carrying a fertilised egg in the uterus, or an embryo or neonate or fetus at any stage of gestational development. Preferably, the present invention provides a use wherein the female is 30 human and the duration of pregnancy is more than approximately 13 weeks 14 WO 2005/053706 PCT/GB2004/005087 of human pregnancy. More preferably, the duration of pregnancy is approximately between 20 and 32 weeks. Preferably, the medicament reduces and/or prevents an inflammatory 5 response in the reproductive system of a female associated with the onset or continuation of labour. The biochemical and physiological changes associated with the onset or continuation of labour have been mentioned above. 10 There are many situations where it is useful to substantially prevent or reduce at least one of the changes in the female reproductive system associated with the onset or continuation of labour. For example, it is well known that certain groups of pregnant females are at high risk of pre-tern labour. Females that have had one or more instances of pre-tenm labour 15 previously are at considerably higher risk of a further pre-tenn labour when pregnant. An increased risk of pre-term labour can also be determined by measuring oncofetal fibronectin levels and by cervical examination using methods well known in the art. 20 It is also useful to prevent or reduce at least one of the changes in the female reproductive system associated with the continuation of labour, particularly uterine contractions, temporarily in circumstances where this is desirable. For example, it may be desirable temporarily to inhibit uterine contractions during labour in order to clear the fetal lungs or in order to transfer the 25 female from one place to another. It is often desirable to transfer the female to a more suitable place where better care is available for her and the offspring. It is also useful to substantially prevent for a considerable duration pre-term 30 labour using the method of the invention. In particular, it is useful to inhibit 15 WO 2005/053706 PCT/GB2004/005087 pre-term uterine contractions from the time when they first occur (or soon thereafter) until the normal time of delivery. Preferably, the medicament reduces and/or prevents an inflammatory 5 response in the reproductive system of a female associated with infection by a pathogenic agent More preferably, the pathogenic agent is viral, bacterial or fungal. 10 Preferably, the medicament reduces and/or prevents an inflammatory response in the reproductive system of a female associated with stretch of the uterus. By "stretch of the uterus" we include mechanical stretching of the uterus 15 occurring where the uterus is overdistended by multiple pregnancy or by excess amniotic fluid (clinically tended hydramnios or polyhydramnios). There may also be more local stretch of the lower segment of the uterus, the cervix and overlying fetal membranes in cases where there is cervical weakness (clinically termed cervical incompetence). 20 Preferably, the medicament reduces and/or prevents one or more of the following conditions: pre-term labour; pathogenic infection; cervical ripening, uterine contractions. 25 By "pre-term labour", we include the meaning of spontaneous labour occurring before the usual calculated time for delivery. In humans, pre term labour is defined as spontaneous labour occurring before 37 weeks of gestation (with 39 weeks being term). The usual calculated time of delivery for females as defined by the invention will be well known in the arts of 30 human and veterinary medicine. 16 WO 2005/053706 PCT/GB2004/005087 Preferably, the medicament reduces and/or prevents fetal or neonatal damage. More preferably, the medicament reduces and/or prevents one or more of 5 the following conditions: astrogliosis; loss of myelin-producing oligodendrocytes; multifocal necroses resulting in cystic change (periventricular leucomalacia, PVL). By "astrogliosis" we include the meaning of hypertrophy (i.e. increasing 10 cell size) of the astroglia, that usually occurs in response to injury. Astroglia are the largest and most munerous neuroglial cells in the brain and spinal cord. Astrocytes (from "star" cells) are irregularly shaped with many long processes, including those with "end feet" which fon the glial (limiting) membrane and directly and indirectly contribute to the blood 15 brain barrier. They regulate the extracellular ionic and chemical environment, and "reactive astrocytes" (along with microglia) respond to injury. Astrocytes can release neuro-transmitters, but their role in signaling (as in many other functions) is not well understood. 20 By oligodendrocytess" we include the meaning of neuroglial cell of the central nervous system (CNS) in vertebrates whose function is to myelinate CNS axons. "Loss of myelin-producing oligodendrocytes" means that there a reduction in the number of these cells. 25 By "multifocal necroses" we include the meaning of death of tissue occurring at more than one site. By "cystic change" we include the meaning of the development of fluid filled spaces in the region where necrosis has taken place. By "periventricular leucomalacia" or "PVL" we include the meaning of damage to the periventrical cerebral white matter 30 which is seen following cytokine induced or hypoxia/ischeamia induced necroses and which can go on to become cystic change. 17 WO 2005/053706 PCT/GB2004/005087 A particularly preferred embodiment of the invention is the use of the cyclopentenone prostaglandin 15-deoxy-A' 12 4 -prostaglandin J 2 and/or prostaglandin A 1 . Alternatively, the cyclopentenone prostaglandin is provided in the form of a prodrug of 15-deoxy-A12' 14 -prostaglandin J 2 and/or prostaglandin A 1 . It will be appreciated by those skilled in the art that certain metabolic 10 precursors of cyclopentenone prostaglandins, may not possess phannacological activity as such, but may, in certain instances, be administered to a patient and thereafter metabolised in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". 15 All prodrugs of the cyclopentenone prostaglandins, particularly those of 15 deoxy-A 12 4 -prostaglandin J 2 and/or prostaglandin A 1 , are included within the scope of the invention. 20 Preferably, the prodrug is PGD 2 (the precursor of 15-dPGJ 2 ) or PGEi (the precursor of PGA 1 ). Preferably, the medicament further comprises a pharmaceutically acceptable excipient, diluent or carrier. 25 By "pharmaceutically acceptable" we mean that the carrier does not have a deleterious effect on the recipient. Typically, the carrier will be sterile and pyrogen free. 30 Preferably the medicament is in a fonn adapted for delivery by mouth, intravenous injection or intra-amniotic injection. 18 WO 2005/053706 PCT/GB2004/005087 Preferably, the medicament is in a form which is compatible with the amniotic fluid. More preferably, the medicament is in a form which has substantially the same pH and/or osmotic tension as amniotic fluid. 5 The anmiotic fluid has a distinct pH and a distinct osmotic tension. The anmiotic fluid pH and osmotic tension are well known to, or can be readily measured by, the person skilled in the art. 10 Preferably, the medicament further comprises an agent for treating a female who has or is at risk of one or more of the following conditions: pre-term labour; pathogenic infection; cervical ripening, uterine contractions. By an "agent for treating a female who has or is at risk of one or more of the 15 following conditions: pre-term labour; pathogenic infection; cervical ripening, uterine contractions" we include corticosteroids, tocolytic agents and anti-inflammatory prostaglandins. Preferably, the agent is a corticosteroid. 20 More preferably, the agent is capable of preventing and/or reducing respiratory distress syndrome. One function of corticosteroids is to mature the fetal lung, which leads to an 25 increase in surfactant production and therefore prevents or reduces the severity of neonatal respiratory problems More preferably, the agent is selected from dexamethasone or betamethasone. Such agents are known to those skilled in the art. 30 Administration of such agents may be two doses of 12mg intra-muscular (IM), 12 or 24 hours apart. 19 WO 2005/053706 PCT/GB2004/005087 Preferably, the agent is capable of delaying delivery. More preferably, the agent capable of delaying delivery is selected from: 5 oxytocin receptor antagonists; calcium channel blockers; sympathomimetics; nitric oxide donors Preferably, the agent is a tocolytic agent. 10 By "tocolytic" we include the meaning of a drug whose action is to stop uterine contractions. More preferably, the tocolytic agent is selected from: oxytocin receptor antagonists, calcium channel blockers, sympathomimetics, nitric oxide 15 donors. More preferably, the oxytocin receptor antagonist is Atosiban. More preferably, the calcium channel blocker is Nifedipine. More preferably, the sympathomimetic is Ritodrine. More preferably, the nitric oxide donor is 20 glyceryl trinitrate. Preferably, the inflanmatory response is mediated by NFfB in uterine cells. More preferably, the cyclopentenone prostaglandin is capable of inhibiting 25 and/or reducing NThB activity by preventing and/or reducing NFiB DNA binding in uterine cells. More preferably, the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFicB activity by preventing and/or reducing NFDB 30 mediated transcriptional regulation in uterine cells. 20 WO 2005/053706 PCT/GB2004/005087 More preferably, the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFicB activity by preventing and/or reducing NFiB production in uterine cells. 5 A further aspect of the invention is to provide a pharmaceutical composition comprising a cyclopentenone prostaglandin and a pharmaceutically acceptable carrier or exipient, the cyclopentenone prostaglandin being present in an amount effective to prevent and/or reduce an inflammatory response in the reproductive system of a female. 10 A further aspect of the invention is a method of treating inflammation within the reproductive system of a female, the method comprising administering an effective amount of a medicament of the invention. 15 A further aspect of the invention is to provide a method for identifying a cyclopentenone prostaglandin for delaying the onset and/or preventing the continuation of labour in a female comprising the step of testing the cyclopentenone prostaglandin to determine if it is capable of inhibiting and/or reducing NFxiB activity in uterine cells in a PPAR-y independent 20 manner. By "NFhcB activity" we include the DNA-binding activity of NFxB and/or NFiB-mediated transcriptional regulation. 25 Testing a cyclopentenone prostaglandin to determine if it is capable of inhibiting and/or reducing NFB activity in uterine cells in a PPAR-y independent manner can be perfonned by the methods described in Example 1, below. For example, whether a cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFicB activity in uterine cells in a 30 PPAR-y independent manner can be determined by using the PPAR-y inhibitor GW-9662, as shown in Figure 6, below. 21 WO 2005/053706 PCT/GB2004/005087 By "PPAR-y independent manner" we include the meaning that the activity of a cyclopentenone prostaglandin occurs without it binding to and/or activating the PPAR-y receptor. 5 It will be understood that a cyclopentenone prostaglandin to determine if it is capable of inhibiting and/or reducing NIFicB activity in uterine cells may be tested in vitro, in vivo or ex vivo. 10 A further aspect of the present invention is to provide a method for making a pharmaceutical composition for use in delaying the onset and/or preventing the continuation of labour in a female comprising providing a cyclopentenone prostaglandin identified by the method of the present invention and combining it with a pharmaceutically acceptable carrier. 15 Preferred, non-limiting examples which embody certain aspects of the invention will now be described, with reference to the following figures: Figure 1 : 15dPG. inhibition of NF-7cB DNA binding 20 Electro-mobility shift assay (EMSA) analysis of NF-kB DNA binding in nuclear protein extracts from (A) myometrial cells, (B) L+ amnion cells, and (C) L- amnion cells treated with 15dPGJ 2 or vehicle for 2 h +/- IL-lb stimulation (15 min). Consensus kB probe used to assess NF-kB DNA 25 binding, and consensus Oct-1 probe used as control. Figure 2: PPAR-yprotein expression Western immunoblots of (A) nuclear and cytosolic protein extracts from 30 myometrial and aniion cells with or without 15 min IL-1 I stimulation, and 22 WO 2005/053706 PCT/GB2004/005087 (B) nuclear extracts of myometrial cells treated with 15d-PGJ 2 +/- IL-lb. Probing with antibody to PPARy. Figure 3 : PPAR-a protein expression 5 Western innunoblots of nuclear and cytosolic protein extracts from inyometrial and amnion cells with or without 15 min IL-lb stimulation. Probing with antibody to PPARc. 10 Figure 4 : PPAR-y agonists do not inhibit NFkB DNA binding EMSA analysis of nuclear protein extracts from myometrial cells treated with (A) troglitazone, (B) GW1929 or vehicle for 2 h +/- IL-lb stimulation (15 min). Consensus icB probe used to assess NF-icB DNA binding, 15 consensus Oct-i probe used as control. For supershift analysis, extracts were preincubated with antibodies against p50 or p65. Figure 5 : Troglitazone and WY-14643 do not inhibit NFICB DNA binding 20 EMSA analysis of nuclear protein extracts from myometrial cells treated with (A) WY-14643 or vehicle, and (B) high doses of troglitazone, NWY 14643 or vehicle for 2 h followed by IL-lb stimulation (15 min). Consensus KB probe used to assess NFKB DNA binding, consensus Oct-1 and Sp-l probes used as controls. 25 Figure 6 PPAR-y antagonist GW9662 does not alleviate 15dPGJ? inhibition of NFKB DNA binding EMSA analysis of nuclear protein extracts from amnion cells treated with 30 l5dPGJ2 +/- GW9662 or vehicle for 2 h followed by IL-lp stimulation (15 23 WO 2005/053706 PCT/GB2004/005087 min). Consensus kB probe used to assess NFiB DNA binding, consensus Oct-I probe used as control. Figure 7 : PGAi inhibition of NFB DNA binding 5 EMSA analysis of nuclear protein extracts from (A) myometrial cells, and (B) anmion cells treated with PGA 1 or vehicle for 2 h followed by IL-lb stimulation (15 min). Consensus xB probe used to assess NFiB DNA binding, consensus Oct-i probe used as control. For supershift analysis, 10 extracts were preincubated with antibodies against p50 or p65. Figure 8 : Effect of cyPGs and PPAR agonists on NFicB transcriptional activity in annion 15 Amnion cells derived from L- or L+ placentas were transiently transfected with the NFKB-dependent reporter construct icB.BG.Luc, treated with 15dPGJ 2 , PGA 1 , troglitazone, WY-14643, or vehicle for 2 h, and then stimulated with IL-1 P (1 ng/ml) for 6 h. The mutated icBmut.Luc construct was used as a control to confim NFKB-mediated transactivation. Values are 20 nonnalised for b-gal reporter activity. Figure 9 : 15dPGJ 2 inhibition of NFiB transcriptional activity in nyonetriwn 25 Myometrial cells were transiently transfected with the NFicB-dependent reporter construct icB.BG.Luc, treated with 15dPGJ 2 or vehicle for 2 h, +/ IL-1 I3 (1 ng/ml) for 6 h. The mutated iBmut.Luc construct was used as a control to confirm NFxB-mediated transactivation. Values are norinalised for b-gal reporter activity. (NS = nonstimulated). 30 24 WO 2005/053706 PCT/GB2004/005087 Figure 10 : Effect of PGAi and PPAR agonists on NFKcB transcriptional activity in myometrium Myometrial cells were transiently transfected with the NFxB-dependent 5 reporter construct KB.BG.Luc, treated with troglitazone, WY-14643, PGA 1 or vehicle for 2 h, +/- IL-lb (1 ng/ml) for 6 h. The mutated xBmut.Luc construct was used as a control to confinn NFl'FB-mediated transactivation. Values are normalised for CMV-Renilla reporter activity. (NS = nonstimulated). 10 Figure 11 : PPAR-y agonist GW1929 does not inhibit NFiCB transcriptional activity Myometrial cells were transiently transfected with the NFicB-dependent 15 reporter construct iB.BG.Luc, treated with GW1929 or vehicle for 2 h, +/ IL-lb (1 ng/nl) for 6 h. Values are nornalised for CMV-Renilla reporter activity. (NS = nonstimulated). Figure 12 : Troglitazone and GW1929 potentiate PPAR-7 activation of a 20 PPRE reporter Myometrial cells were cotransfected with 0.4 mg of the PPAR-y-dependent reporter construct 3-PPRE-TK.pGL3 and 100 ng, 200 ng or 300 ng of a PPAR-y expression construct. Cells were treated with 10 mNI or 20 mM of 25 (A) troglitazone or (B) GW1929, or vehicle for 24 h. Values are normalised for CIV-renilla reporter activity. Similar results were obtained with transfection of amnion cells. Figure 13 : Troglitazone does not inhibit NF7B transcriptional activity in 30 PPAR--transfected cells 25 WO 2005/053706 PCT/GB2004/005087 Myometrial cells were transfected with 0.4 mg xB.BG.Luc reporter and 200 ng PPAR-y expression vector and treated with 10 mM troglitazone or vehicle for 7 h +/- IL-lb (1 ng/ml) for 17 h. Values are normalised for 5 CMV-renilla reporter activity. (NS= nonstimulated). Figure 14 PPAR-y overexpression does not potentiate 15d-PGJ? inhibition ofNFkB activity 10 Myometrial cells were transfected with 0.4 mg icB.BG.Luc reporter and 200 ng PPAR-y expression vector and treated with 15d-PGJ 2 for 2h followed by IL-l (1 ng/ml) for 6 h. Values are normalised for p galactosidase reporter activity. (NS= nonstimulated). 15 Figure 15 : 15dPGJ 2 inhibition of p65 nuclear localisation, p50 phosphorylation, and hcBa degradation Western irmnunoblots of nuclear or cytosolic protein extracts from (A) myometrial cells, (B) L- amnion cells, and (C) L+ aniion cells treated with 20 l5dPGJ 2 or vehicle for 2 h +/- IL-Is stimulation (15 min). Blots probed with antibodies against p65, p 5 0 or IicBa. Figure 16 : PGAi inhibition of p65 nuclear localisation and IcBa degradation 25 Western inmmunoblot of nuclear or cytosolic protein extracts from myornetrial cells treated with PGA 1 or vehicle for 2h followed by IL-1p stimulation (15 min). Blots probed with antibodies against p65or IicBa. 26 WO 2005/053706 PCT/GB2004/005087 Figure 17 : PGE 2 does not inhibit TNFa- and IL-1p-induced NFicB activation Analysis of nuclear protein extracts from myometrial cells treated with 5 PGE 2 or vehicle for 2 h +/- TNFa or IL-lb stimulation (15 min). (A) EMSA using consensus xB probe. (B) Western immunoblot probing for nuclear p 6 5 . Figure 18 : PGE 2 does not induce NFicB DNA binding 10 EMSA analysis of nuclear protein extracts from (A) L- amnion cells and (B) myometrial cells treated with vehicle, PGE 2 or IL- 15. Consensus icB probe used. 15 Figure 19 : J5dPGJ 2 inhibits IcBaphosphorylation Western immunoblots of cytosolic extracts from niyometrial cells (A) treated with 15dPGJ 2 for 2h +/- ILib for l5min; probed for IK, and (B) treated with 30mM 15dPGJ 2 , 40mM MG132 or vehicle for 2h, +/- IL-1P for 20 15min; probed for IcBa. Figure 20 : Effect of J5dPGJ and PPAR agonists on IL-1 P-induced COX-2 protein expression 25 Western irmnunoblot of cytosolic protein extracts from myometrial cells treated with 15dPGJ 2 , troglitazone, WY-14643 or vehicle for 2h, followed by IL-1 IP stimulation for 6h. Probed with antibodies to (A) COX-2, and (B) a smooth muscle actin. 27 WO 2005/053706 PCT/GB2004/005087 Figure 21 : Schematic of the structure of (A) prostaglandin Ai (PGAi) and (B) 15-deoxy -Ap' prostaglandin J 2 (15-dPGJ 2 ) Figure 22 : Effect of LPS and 15d-PGJ on inflammatory responses - IL- p 5 levels Concentrations of IL-1 I in placental homogenates collected from gestation day 16 mice 6 hours after intrauterine injection of 250 pg LPS + vehicle or 250ptg LPS + 4pg 15d-PGJ 2 . * denotes statistically significant difference (t 10 test (p<0.05)). Figure 23 : Effect of LPS and 15d-PGJ 2 on inflammatory responses phospho-p65 levels 15 Relative concentrations of phospho-p65 in placental homogenates collected from gestation day 16 mice 6 hours after intrauterine injection of 250ptg LPS + vehicle or 250ptg LPS + 4pLg 15d-PGJ 2 . * denotes statistically significant difference (t-test (p<0.05)). 20 Figure 24 :The cyclopentenone ring is essential for cyPG inhibition of NF KB. (A) NF-iB-DNA binding was measured by EMSA in nuclear protein extracts from nyometrial cells pre-treated with vehicle, 15d-PGJ 2 or PGA 1 25 for 2 h, followed by stimulation with IL-l (1 ng/ml) for 15 min. Antibodies against p50 and p65 were used for supershift analysis. (B) Myometrial cells were transiently transfected with a NF-i-iB-LUC reporter and a p-gal reporter plasmid, pre-treated with vehicle or PGA 1 for 2 h, and stimulated with IL- 1 P (1 ng/ml) for 6 h. Luciferase activity was normalized 30 for p-gal reporter readout. Values are presented as the mean +/- SEM obtained for each treatment done in triplicate. Western blot analysis of 28 WO 2005/053706 PCT/GB2004/005087 nuclear p65 and p50 expression in myometrial cells treated with (C) PGA 1 or (D) 15d-PGJ 2 for 2 h, followed by stimulation with IL-1p (1 ng/ml) for 15 min. (E) Western blot analysis of whole cell lysates from myometrial cells treated with 15d-PGJ2 or 9,10-dihydro-15d-PGJ2 for 2 h, followed by 5 stimulation with IL- 1p (1 ng/il) for 15 min. Membranes were probed with antibodies against p65 and Ser 536-phosphorylated p65. Similar results were obtained in amnion epithelial cells. EXAMPLE 1 - Experimental data 10 Methods Abbreviations EDTA Ethylenediaiminetetraacetic acid EGTA Ethyleneglycol bis-aminoethyltetra acetic acid DTT Dithiotreitol HEPES 4-(2 -hydroxyethyl)- 1 -piperazineethanesulfonic acid NP-40 Nonidet P-40 SDS-PAGE Sodium dodecyl sulphate - Polyacrylamide gel electrophoresis PVDF Polyvinylidene difluoride PBS-T Phosphate Buffered Saline plus Tween HRP Horseradish peroxidase PBS Phosphate Buffered Saline FCS Foetal Calf Serum DMEM Dulbecco's modified eagle's medium 15 Tissue biopsies and cell culture Local Ethics committee approval was obtained for the collection of these tissues and patients gave informed consent. 20 Hurnan myometrial cell culture Myometrial tissue was collected at term from the upper margin of uterine 29 WO 2005/053706 PCT/GB2004/005087 incision at the time of lower segment caesarean section either prior to the onset of labour (L-) or during fetal distress (L+). L+ samples were collected by Dr Mark Johnson and Dr S Soorrana at Chelsea & Westminster Hospital. Myometrial tissue was dissected, rinsed in PBS, and digested in 5 serum-free DMM containing 15mg/nl collagenase 1A (Sigma), 15mg/mil collagenase X, and 50mg/mil bovine serum albumin for 45min at 37'C. The cell suspension was filtered through a cell strainer, centrifuged at 400g for 5min, and the pellet re-suspended and plated out in DMEM, 10% FCS (Helena BioScience), 1% L-glutamine, 1% penicillin-streptomycin. Cells 10 were used between passage numbers 1-4. Human Annion Cell Culture Placentae were obtained from patients at term either at elective Caesarean 15 section prior to labour (L-) or following spontaneous labour onset and vaginal delivery (L+). Annion cells were prepared as described in Bennett et al., (1989). Briefly, the amnion was separated from the placenta, washed 3x in PBS, cut into strips, and incubated in 0.5mM EDTA in PBS for 15min. The strips were washed in PBS 2x and digested with 2.5mg/ml 20 dispase in serum-free DMEM for 35min at 37 0 C. The amnion was then shaken vigorously in DMEM, 10% FCS to dissociate the cells, the remaining strips discarded, and the cell suspension pelleted at 175g for 10nin and cultured in DMNEM, 10% FCS (Sigma), 1% L-glutamnine, 1% penicillin-streptomycin. 25 Protein Extracts from cultured cells Nuclear and cytosolic protein extracts were obtained from cultured ainion cells as described by Schreiber et al (1989). For nuclear/cytosolic 30 -fractionation, confluent cell monolayers were scraped and lysed using a buffer containing 10mM HEPES, 10mM KCl, 0.1mM EDTA, 0.1mM 30 WO 2005/053706 PCT/GB2004/005087 EGTA, 2mM DTT, 1% (v/v) NP-40 and complete protease inhibitor tablets (CPIs, Roche), diluted to manufacturer's instructions. Cell lysates were incubated on ice for 1 0min and NP-40 added to a final concentration of 1% (v/v). Lysates were vortexed for 10secs and centrifuged for 30sees at 4'C, 5 12000g. The supernatants were retained as the cytosolic protein extracts. The pellets were resuspended in buffer containing 1OmM HEPES, 10mM KCl, 0.1mM EDTA, 0.1mM EGTA, 2mM DTT, 400mM NaCl, 1% NP-40 (v/v) and CPIs. Samples were shaken vigorously for 15min in an ice bath. The nuclear protein extracts were obtained in the supernatant following a 10 5min centrifugation at 4'C, 12000g. For whole cell lysates, confluent cell monolayers were scraped and lysed in a hiah-salt extraction buffer containing 0.4M KCl, 20mM IIEPES, 20% (v/v) glycerol, 1mM DTT, and CPIs. 15 Protein Extracts fiom fresh tissue biopsies Tissue samples were rinsed in ice-cold PBS, dissected, flattened between aluminium foil, flash-frozen in liquid nitrogen, and stored at -80'C. 20 Samples were reduced to powder in liquid nitrogen using a pestle and mortar. Powdered tissue was homogenized in a Dounce homogeniser on ice in a buffer containing 0.6% (v/v) NP-40, 150mM HEPES, 1mM EDTA, 0.5mM PMSF and any unbroken tissue was removed by centrifugation for 30sec at 2000rpm at 0 0 C. The supernatant was incubated on ice for 5min, 25 centrifuged for 10min at 4000rpm at 0 0 C, and the nuclear pellets resuspended in 25% (v/v) glycerol 20niM HEPES, 0.42M NaCl, 1.2 mM MgCl,, 0.2mM EDTA, 0.5mM DTT, and CPIs. All extracts were aliquoted, frozen on dry ice and stored at -80'C. The 30 extracts were processed for protein quantitation by the Lowry method using 31 WO 2005/053706 PCT/GB2004/005087 Bio-Rad protein assay reagents (Bio-Rad Laboratories) according to manufacturer's instructions. Electro-mrobilit; shift assay (EMSA) Oliaonucleotide labelling Sense and antisense strands (175mnole/ml each) were incubated in annealing buffer (10mM Tris-HCl pH7.5, 100mM NaCl, 1mM EDTA) for 10 10min at 65'C, and allowed to cool at room temperature for 2h. 3.5 pmole double-stranded oligonucleotides were end-labelled with 0.37MBq 3 2 P(yATP) by incubating for 30min at 37'C with T4 polynucleotide kinase. Labelled oligonucleotides were recovered by centrifugation at 3000rpm for 2min through MicroSpin G-25 or G-50 sephadex columns (Amersham 15 Biosciences). EMSA 3-5 tg protein extracts were incubated on ice for lh with non-radiolabelled 20 non-specific oligonucleotide (poly(dI-dC) or Oct-1) in, a binding buffer (20% (v/V) glycerol, 5mM MgCl 2 , 2mM EDTA, 50mM Tris-HCl pH7.5, 250mM NaCl, 2mnM DTT), followed by a 45min incubation with 0.035pmole 3 2 P(IyATP)-end labelled oligonucleotide probes: 25 consensus NF-icB: 5'-AGT TGA GGG GAC TTT CCC AGG C-3' consensus Oct-1: 5'-TGT CGA ATG CAA ATC ACT AGA A-3' consensus SP-1: 5'-ATT CGA TCG GGG CGG GGC GAG upstream COX-2 KB: 5'-CGG GAG AGG GGA TTC CCT GCG C-3' downstream COX-2 kB: 5'-AGA GTG GGG ACT ACC CCC TCT-3' 30 32 WO 2005/053706 PCT/GB2004/005087 Oct-I or SP-1 consensus sequences were used as a controls for a NF-IrB specific effect. The resulting protein/DNA complexes were separated in a 4% acrylamide gel, the gel dried under vacuum at 80 0 C and exposed to X ray film. For supershift analysis, samples were incubated with 2pg 5 antibodies for 30min on ice prior to incubation with oligonucleotides. Non radio-labelled oligonucleotides were used at 100-fold molar excess for specific and non-specific competition for DNA binding. Reagents for EMSA were obtained from Promega Life Sciences, Delta House, Chilworth Research Centre, Southampton S016 7NS, United Kingdom. 10 SDS-PAGE and Western blotting analysis Protein samples (20-70pig) were mixed with Laemmli sample buffer (1:1) containing p-mercaptoethanol (5%), and boiled for 5min. Proteins were 15 then separated by SDS-PAGE (12-14% gels) and transferred onto PVDF membrane (Amersham Phannacia Biotech). The membranes were blocked overnight in 5% non-fat milk prepared in PBS-T buffer, at 4'C. The blots were incubated with the primary antibody in 1% non-fat milk in PBS-T buffer for lh, and washed three times (10min each) in PBS-T with vigorous 20 shaking. The blots were then incubated with HRP-conjugated secondary antibody (diluted 1:2000 in 1% non-fat milk in PBS-T buffer) for lh and washed three times (10min each) in PBS-T. Signal detection was achieved using enhanced chemi-luminescence (ECL plus system, Amersham Pharmacia Biotech) according to manufacturer's instructions. 25 To re-probe a membrane, blots were incubated for 3 0min in 50'C stripping buffer (2% SDS, 62.5mM Tris-HCI pH6.7, 100mM 2-mercaptoethanol), washed 2x in PBS-T, placed in blotto overnight, and then probed with a new antibody as above. 30 33 WO 2005/053706 PCT/GB2004/005087 3 0
-
5 0pg protein extracts were subjected to SDS-PAGE and Western immuno-blotting. Secondary antibodies were IgG-ERP and ECL Plus detection kit (Amershain Phannacia Biotech. Amershain Place, Little Chalfont, Bucks, HP7 9NA) was used for visualisation. Transfections and luciferase assay Cells at 70-80% confluence in 24-well plates were transfected using the liposome Transfast (Pronega). 0.5 tg per well of luciferase reporter 10 construct was transfected using a 1:1 ratio of transfection (i.e., 3pl Transfast per 1pg DNA) in serum-free DMEM for lh. DN\EM, 10% FCS was then added and the cells were incubated at 37'C for 24h. The medium was replaced with DMEM, 2% FCS for a further 24h, and the cells treated with various agonists/inhibitors or vehicle for 6-8h. Transfections were analysed 15 in a dual firefly/renilla (Packard BioSciences/Calbiochem) luciferase assay or firefly/p-galactosidase (Pronega/Galacton) assay using a luminometer. pGL3.6kB.IBG.luc was the reporter construct used to assess NF-KB mediated transcription, while the mutant pGL3.6xBmut.luc and empty 20 pGL3.BG.luc were used as controls (Schwarzer et al., 1998). pGL3.6icB.BG.luc: a NF<-iB-dependent reporter construct with 6 copies of the NF-icB binding site. It contains two tandem repeats of the sequence 5' GGG GAC TTT C CC TGG GGA CTT TCC CTG GGG ACT TTC CC-3, 25 which contains three copies of the decameric NF-icB binding site (underlined) upstream of a minimal p-globin promoter driving a luciferase gene. 34 WO 2005/053706 PCT/GB2004/005087 pGL3.6rcBmut.luc: this reporter construct is as above except that the core NF-icB binding site is mutated to 5'-GCC ACT TTC C-3' (mutated bases underlined). 5 pGL3 .BG.luc: this reporter construct contains only the minimal p-globin promoter. Cells were co-transfected with the renilla vector pRL-CMV or a p galactosidase vector pCH110 as internal controls for transfection 10 efficiencies. In vitro translation and plasnid preps For recombinant production of p6 5 , a pSG5/p65 expression construct was 15 transcribed and translated using a TNT Coupled Reticulocyte Lysate System (Promega), according to manufacturer's instructions. QIAGEN Maxi Prep kits were used for plasmid isolation from transformed JM109 E. coli cells, and all constructs were subsequently precipitated with polyethylene glycol. 20 Reagents/Antibodies Recombinant cytosine IL-l@ and TNFa from R&D Systems; 15d-PGJ 2 , PGAi, troglitazone, GW-9662, and 16,16-dimethyl-PGE 2 from Cayman 25 Chemical; WY-14643, MG132 proteasome inhibitor, and PG490 (triptolide) from Calbiochem; HRP-conjugated secondary antibodies and antibodies to p50, p65, c-rel, Rel B, COX-2, fictc, IxdIBp, and PPARy from Santa Cruz; antibodies to p52, Bcl-3 and smooth muscle actin from Upstate Biotechnologies. Antibody to PPAR-y from Affinity BioReagents, to 30 phospho-p65 from Cell Signaling, to COX-1 from Alexis Biochemicals, and to lamin B from Oncogene Research Products. 35 WO 2005/053706 PCT/GB2004/005087 Mouse model of'pIeterm labour Surgery was performed on timed pregnant NF1 mice at day 16 of gestation. 5 After deep maternal anaesthesia was attained, a minilaparotomy was performed in the lower abdomen. The uterine horns were exposed through the incision and preterm labour was induced by the intrauterine injection of 250pig lipopolysaccharide (LPS, Sigma) into the gravid hom. This was immediately followed by injection of 4pg 5d-PGJ 2 (Cayman), or an equal 10 volume of vehicle (methyl acetate), at the same site. The uterus was then returned to the abdomen and the fascia and skin were closed with continuous vicryl sutures. Effect of LPS and 15d-PGJ 2 on inflannatory responses 15 Mice were sacrificed 6 hours after injection of LPS ± 15d-PGJ2. Placentae were washed in phosphate buffered saline (PBS), flash frozen in liquid nitrogen and stored at -80'C until further processing. Fetuses were washed in PBS, then immediately fixed in 4% parafonnaldehyde for 24h and then 20 stored in 70% ethanol until further processing. Placentae were homogenized for 1 minute in the presence of lysis buffer comprising 400mM KCl, 20mM HEPES pH7.4. 1mM dithiothreitol, 20% glycerol and 5% (v/v) protease inhibitor cocktail. 25 Homogenate levels of Interlekin-lf (IL-1p) and tumour necrosis factor a (TNFa) were determined in placental lysates by ELISA (R and D systems) according to manufacturers instructions. Total protein concentrations were determined for each homogenate and IL-1j and TNFa levels were expressed as pg/mg total protein. Homogenates were also subjected to 30 polyacrylamide gel electrophoresis. Loading volumes were adjusted according to the protein content of each homogenate such that a constant 36 WO 2005/053706 PCT/GB2004/005087 amount of protein was run in each lane. Phosphorylated p65 (phospho-p65) was detected by western inrnunoblotting using a specific antibody (Santa Cruz) and quantified by densitometric analysis. Results CyPGs, but not PPAR agonists, inhibit NF-icB DNA binding in ainion and 10 nyometrial cells. 15d-PGJ 2 inhibited ILI-p-induced NF-1CB DNA binding in a dose dependent manner in myometrial cells, as well as in L- and L+ amnion cells (Fig. 1). Protein binding to a consensus Oct-I or Sp-1 probe was unaffected 15 by either IL-i [P or 15d-PGJ 2 treatment, confirming that the effects observed are NF-icB-specific. Since PPAR-y is the putative endogenous receptor for 15d-PGJ2, and PPAR expression may be affected by cytokines (Tontonoz et al., 1998, Tanaka et 20 al., 1999), PPAR-y protein expression was examined in myonetrial and amnion cells. PPAR-y was shown to be expressed predominantly in the nucleus of both cell types, and its expression was not affected by IL-i [P or 15d-PGJ 2 treatment (Fig. 2). 15d-PGJ 2 can also transactivate PPAR-a, though more wealdy than PPAR-y (Forman et al., 1995). PPAR-a 25 expression in myometrial and anion cells was found to be predominantly cytoplasmic (Fig. 3). The ability of synthetic PPAR agonists to mimic the inhibitory effects of 15d-PGJ 2 was examined. The PPAR-y agonist troglitazone had no effect on 30 NF-xB DNA binding at 10-50jM doses, although it did cause a slight reduction at 100 LM (Fig. 4, 5). Troglitazone can transactivate PPAR-y at 37 WO 2005/053706 PCT/GB2004/005087 1p M and induces weak interactions between PPAR-y and the co-activators p300 and steroid receptor co-activator (SRC-1) at 1OpM doses; adipogenesis is positively regulated by PPAR-y, and troglitazone can induce expression of adipogenic markers at 5-10ptM doses (Prusty et al., 2002). 5 Thus, at 100gM concentrations, it is unlikely that troglitazone is exerting a specific effect through PPAR-7. Since structurally distinct PPAR ligands may differentially affect coactivator/corepressor recruitment, a new potent PPAR-y agonist, which lacks the TZD moiety, was also used. This GW1929 ligand failed to inhibit NF-1cB DNA binding (Fig. 6). The 10 synthetic PPAR-a agonist WY-14643 can transactivate PPARa at 5-25KM doses in a GAL4 chimera transfection system (Kehrer et al., 2001), but WY-14643 had no effect on NF-1<B DNA binding, at 10-100pM concentrations. To further investigate a potential role for PPAR-y in mediating the inhibitory effects of 15d-PGJ 2 , NF-icB DNA binding was 15 assessed in cells treated with 15d-PGJ 2 in the presence of the selective PPAR-y inhibitor GW-9662. GW9662 binds irreversibly to PPAR-y through covalent modification of Cys 2 11 in the ligand-binding domain (Leesnitzer et al., 2002). GW-9662 failed to alleviate 15d-PGJ2 inhibition of NF-icB (Fig. 6). 20 In contrast, PGA 1 , which does not act as a PPAR ligand but does contain a cyclopentenone ring, was able to inhibit NF-icB DNA binding in amonion and myometrial cells, albeit at much higher doses than 15d-PGJ2 (Fig. 7). 25 CyPGs, but not PPAR agonists, inhibit NF-icB transcriptional activity. To determine whether the cyPG effects on NF-1cB DNA binding extend to inhibition of NF-icB transactivation potential, amnion cells were transfected with the NF-icB-dependent reporter icB.BG.Luc and treated with 15d-PGJ, 30 PGA 1 , troglitazone, WY-14643 or vehicle, followed by IL-1p stimulation 38 WO 2005/053706 PCT/GB2004/005087 (Fig. 8). Constitutive reporter activity was seen in both L- and L+ anion cells, although the levels were lower and showed a greater increase with IL 13. in L- cells, in agreement with previous studies by Allport et al (2001). Both 15d-PGJ 2 and PGA 1 inhibited IL-1p-induced NF-icB transcriptional 5 activity, whereas troglitazone and WY-14643 did not. In myometrial cells, 15d-PGJ2 inhibited IL-lp-induced NF-kB transcriptional activity in a dose-dependent manner, reducing reporter activity to basal levels (Fig. 9). IL-1p-induced NF-kB transcriptional 10 activity was also reduced to basal levels by PGA 1 , but not troglitazone, GW19 2 9 or WY-14643 (Fig. 10, 11). GW1929 and troglitazone were shown to be functional as PPAR-y ligands, potentiating PPAR-y-mediated transcription of a PPRE-dependent reporter 15 in both cell types. Endogenous PPAR-y levels were not sufficient to drive the PPRE reporter in the transfection system used, with transcription requiring overexpression of the receptor. Troglitazone was also unable to inhibit a NF-icB-dependent reporter in PPARy-transfected cells, and PPARy overexpression did not promote 15d-PGJ 2 inhibition of 20 NF-icB transcriptional activity (Fig. 12, 13, 14). CyPGs, but not PGE, inhibit NF-0B activation and IicB degradation. 15d-PGJ 2 inhibited IL-1p-induced p65 nuclear translocation and p50 25 phosphorylation in myometrial cells and in L-, L+ amnion cells in a dose dependent manner (Fig. 15). This was paralleled by inhibition of IL-lp induced IKBa and IxB3 degradation. Similarly, PGA 1 inhibited p65 nuclear translocation and IiclBdegradation in myometrial cells (Fig. 16). 39 WO 2005/053706 PCT/GB2004/005087 16,16-Dimethyl-PGE 2 , a PGE2 analogue with increased half-life, did not inhibit NF-xB DNA binding (controlled for with Oct-i binding) or IL-1p induced p65 nuclear translocation in myometrial and amnion cells (Fig. 17). This is not unexpected, since, in contrast to the cyPGs, PGE 2 is known to be 5 pro-inflammatory, does not contain a cyclopentenone ring, and does not activate PPAR-y (Forman et al., 1995). 16,16-dimethyl-P GE 2 did not inhibit NF-xfB DNA binding or p6 5 nuclear translocation in myometrial cells (Fig. 18). However, neither did it stimulate NF-icB activity as reported in T cells (Dumais et al., 1998), nor did it synergise with IL-1BDor TNFa. 10 Effect of 15-dPGJ 2 on NF-rB upstream activators and downstream targets. In contrast to the proteasome inhibitor MG132, which prevented IL-1 induced licBa degradation and resulted in the accumulation of undegraded, 15 phosphorylated IrBa, accumulation of phosphorylated I-Ba was not detected following 15-dPGJ, treatment, suggesting that 15-dPGJ2 may be affecting IKKs or other upstream kinases (Fig. 19). Both IL-l P and 15 dPGJ 2 treatment had no effect on IKKa or IKKP protein expression, although it is more likely that 15d-PGJ 2 would inhibit the kinase activity of 20 the Ius. Since COX-2 is an important target gene for NF-xB in labour, the effect of 15-dPGJ 2 and PPAR agonists on COX-2 expression was assessed. IL-1p induced COX-2, expression was inhibited by 15-dPGJ 2 , but not by 25 troglitazone or WY-14643 (Fig. 20). Similar results were obtained in L and L+ amnion cells. Effect of LPS on pre-term delivery. 40 WO 2005/053706 PCT/GB2004/005087 Pre-tern delivery of pups occurred by 16 hours after injection of LPS using the mouse model of preterm labour, as set out in the methods above. Effect of LPS and ]5d-PGJ, on inflamInatory responses In all mice, levels of TNFa and IL-[p were significantly higher in the placentae proximal to the injection site compared to those in the opposite horn. Levels of IL- 1P were approximately 40% lower in proximal placentae injected with LPS + 15d-PGJ 2 compared to those given LPS + vehicle (Fig. 10 22). This difference was statistically significant (p<0.05). In contrast, TNFa levels were not significantly altered according to drug treatment. Significantly, placental levels of IL-I were not altered according the proximity of the placenta to the site of injection, indicating that 15 inflammatory response can be distributed throughout the uterus, irrespective of the site of infection. However, phospho-p65 levels were approximately 35% lower in proximal placentae injected with LPS + 15d-PGJ 2 compared to those given LPS + vehicle (Fig. 23) and this difference was statistically significant (p<0.05). 20 The cyclopentenone ring is essentialfor cyPG inhibition ofNF-icB Several studies have demonstrated that 15d-PGJ 2 is a PPAR agonist, whilst other prostaglandins, such as PGAI, are not (Forman et al., 1995; Kliewer et 25 al., 1995; Ferry et al., 2001). We have shown that PGA 1 shares the effect of 15d-PGJ 2 on NF-icB., but that 9,10-dihydro-15d-PGJ 2 (an analogue of 15d
PGJ
2 which retains PPARy agonist activity but in which the cyclopentenone ring has been disrupted) could not reproduce the effects of 15d-PGJ 2 (Fig. 24). Taken together, these findings indicate that the inhibitory effects of 30 15d-PGJ 2 on NF- iciB in aimion epithelial and myometrial cells can be 41 WO 2005/053706 PCT/GB2004/005087 attributed to its electrophilic ring, that similar effects would be expected with other cyclopentenone prostaglandins but not with other, non cyclopentenone PPAR agonists. 5 Conclusions NF-icB inhibition by cyPGs 15d-PGJ 2 inhibited IL1-$-induced NF-,-B DNA binding and NF-icB 10 mediated transactivation in myometrial cells, as well as in L- and L+ anrion cells. 15d-PGJ2 inhibited the nuclear translocation and activation of NF-iB, at least in part, by preventing the degradation of IKBa by IL-1p. In myometrial and amnion cells, which expressed both PPAR-a and PPAR 15 y receptors, neither PPAR-y nor PPAR-a agonists were able to inhibit IL 1 P-induced NF-KB DNA binding or NF-kB transcriptional activity at doses shown to inhibit NF-irB in other cell types (Chinetti et al., 1998; Gupta et al., 2001), or even at higher concentrations. In a study investigating the potential functional interactions between PPAR-y and NF-icB in adipocytes, 20 PPAR-y agonists did not impair TNFc-induced NF-KB activation, nuclear translocation, or DNA binding activity; rather, they antagonised the transcriptional regulatory activity of NF-icB, and PPAR-y overexpression was required to demonstrate such inhibition (Ruan et al., 2003). In the present study, while PPAR-y overexpression potentiated transactivation of a 25 PPRE, it did not enable the PPAR-y agonists to inhibit NF-kB transcription. In addition, 15d-PGJ 2 was able to inhibit NF-xB transcription in the absence of exogenous PPAR-y and overexpression of this receptor did not promote inhibition. 42 WO 2005/053706 PCT/GB2004/005087 IL-1p-induced COX-2 expression was inhibited by 15d-PGJ-, but not by PPAR agonists. While PPAR agonists are known to be anti-inflanmatory and can inhibit COX-2 expression (Staels et al., 1998; Subbaramaiah et al., 2001), they have also been reported to enhance COX-2 expression in certain 5 cell types (Meade et al., 1999; Ikawa et al., 2001; Pang et al., 2003). CyPGs such as 15d-PGJ are characterised by the presence of a cyclopentenone ring system containing an electrophilic carbon. This ring can react covalently with nucleophiles such as the free sulfhydryls of 10 glutathione and cysteine residues in cellular proteins. Receptor-independent actions of 15d-PGJ2 have been attributed to its cyclopentenone ring. NF-icB proteins contain a conserved cysteine residue in their DNA-binding doinain (DBD) and alkylation of this cysteine impairs DNA binding (Toledano et al., 1993). In the present study, PGA 1 , a cyPG that does not act as a PPAR 15 y ligand, was able to inhibit NF-icB DNA binding and trans activation, albeit at higher concentrations than 15d-PGJ2. This ability of PGA 1 , but not PGE 2 or PPAR agonists, to mimic the effects of 15d-PGJ2 suggests that these cyPGs may inhibit NF-7cB in amnion and myometrial cells by virtue of their cyclopentenone ring. Indeed, our results indicate that the inhibitory effects 20 of 15d-PGJ 2 on NFicB in amnion epithelial and myometrial cells can be attributed to its electrophilic ring and that similar effects would be expected with other cyclopentenone prostaglandins but not with other, non cyclopentenone PPAR agonists. 25 While direct modification of NF-icB cysteines has not been addressed in this study, both 15d-PGT 2 - and PGA 1 -mediated inhibition of NF-cB was shown to involve the inhtibition of IiB a degradation, suggesting that events further upstream in the NF-KB cascade are being targeted. 43 WO 2005/053706 PCT/GB2004/005087 Thus, while PPAR activation may not be effectively anti-inflammatory in amnion and myometrium, the use of cyPGs should prove useful in repressing NF-icB, and therefore an array of pro-inflarmnatory and labour associated genes, in these tissues. CyPG administration offers an attractive 5 alternative approach to anti-inflammatory treatment since a potential specificity of cyPGs for IK p/hicBc would spare other potentially beneficial pathways of NF-icB activation (e.g., the processing of p105 and formation of p50 homodimers), which might be disrupted by more broad spectrum NF-B inhibitors. The use of the cyPGs, able to simultaneously 10 trigger the inhibition of the pro-inflammatory NF-icB and harness the anti inflammatory activities of endogenous cytoprotective molecules represents a novel therapeutic approach in the treatment of pretenn labour and neurodevelopmental disorders of the neonate. 15 This study provides evidence that the mouse model used is an effective model for the study of pretern delivery and agents that may delay the onset of pretend delivery. The finding of lower levels of IL- 1p and phospho-p65 in mice treated with the cyclopentenone prostaglandin 15d-PGJ2 suggests that this compound is effective at blocking the inflammatory pathway 20 induced by LPS treatment in vivo. 44 WO 2005/053706 PCT/GB2004/005087 EXAMPLE 2 - Preferred pharmaceutical formulations and modes and doses of administration. The compounds of the present invention may be delivered using an 5 injectable sustained-release drug delivery system. These are designed specifically to reduce the frequency of injections. An example of such a system is Nutropin Depot which encapsulates recombinant human growth hormone (rhGH) in biodegradable microspheres that, once injected, release rhGH slowly over a sustained period. 10 The compounds of the present invention can be administered by a surgically implanted device that releases the drug directly to the required site. For example, Vitrasert releases ganciclovir directly into the eye to treat CMV retinitis. The direct application of this toxic agent to the site of disease 15 achieves effective therapy without the drug's significant systemic side effects. Electroporation therapy (EPT) systems can also be employed for administration. A device which delivers a pulsed electric field to cells 20 increases the permeability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drug delivery. Compounds can also be delivered by electroincorporation (EI). EI occurs when small particles of up to 30 microns in diameter on the surface of the 25 skin experience electrical pulses identical or similar to those used in electroporation. In EI, these particles are driven through the stratum corneum and into deeper layers of the skin. The particles can be loaded or coated with drugs or genes or can simply act as "bullets" that generate pores in the skin through which the drugs can enter. 30 45 WO 2005/053706 PCT/GB2004/005087 An alternative method of administration is the ReGel injectable system that is thermosensitive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into known, safe, biodegradable polymers. The active 5 drug is delivered over time as the biopolymers dissolve. The compounds of the invention can also be delivered orally. The process employs a natural process for oral uptake of vitamin B 12 in the body to co deliver proteins and peptides. By riding the vitamin B 12 uptake system, the 10 protein or peptide can move through the intestinal wall. Complexes are synthesised between vitamin B 1 2 analogues and the drug that retain both significant affinity for intrinsic factor (IF) in the vitamin B 12 portion of the complex and significant bioactivity of the drug portion of the complex. 15 Compounds can be introduced to cells by "Trojan peptides". These are a class of polypeptides called penetratins which have translocating properties and are capable of carrying hydrophilic compounds across the plasma membrane. This system allows direct targeting of oligopeptides to the cytoplasm and nucleus, and may be non-cell type specific and highly 20 efficient (Derossi et al., 1998). Preferably, the pharmaceutical formulation of the present invention is a unit dosage containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of the active ingredient. 25 The compounds of the invention can be administered orally or by any parenteral route, in the form of a pharmaceutical formulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable 30 dosage fonn. Depending upon the disorder and patient to be treated, as well 46 WO 2005/053706 PCT/GB2004/005087 as the route of administration, the compositions may be administered at varying doses. Formulations in accordance with the present invention suitable for oral 5 administration may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in oil liquid emulsion. The active ingredient may also be presented as a bolus, 10 electuary or paste. A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing 15 form such as a powder or granules, optionally mixed with a binder (e.g. povidone, gelatin, hydroxypropyhnethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g. sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Moulded tablets may be made by moulding in a suitable 20 machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethylcellulose in varying proportions to provide desired release profile. 25 Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and 30 mouth-washes comprising the active ingredient in a suitable liquid carrier. 47 WO 2005/053706 PCT/GB2004/005087 In human therapy, the compounds of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice. 5 For example, the compounds of the invention can be administered orally, buccally or sublingually in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for innnediate-, delayed- or controlled-release applications. The 10 compounds of the invention may also be administered via intracavernosal injection. Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and 15 glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as 20 magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, 25 cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the compounds of the invention may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof 30 48 WO 2005/053706 PCT/GB2004/005087 The compounds of the invention can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They 5 are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by 10 standard pharmaceutical techniques well-known to those skilled in the art. Fonnulations suitable for parenteral administration include aqueous and non aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood 15 of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The fonnulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried lyophilisedd) condition requiring only the addition of the sterile liquid carrier, 20 for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described. Generally, in humans, oral or parenteral administration of the compounds of 25 the invention is the preferred route, being the most convenient. For veterinary use, the compounds of the invention are administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will detennine the dosing regimen and 30 route of administration which will be most appropriate for a particular animal. 49 WO 2005/053706 PCT/GB2004/005087 The formulations of the pharmaceutical compositions of the invention may conveniently be presented in unit dosage fonn and may be prepared by any of the methods well known in the art of pharmacy. Such methods include 5 the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. 10 Preferred unit dosage formulations are those containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of an active ingredient. A preferred delivery system of the invention may comprise a hydrogel 15 impregnated with a compound of the invention, which is preferably carried on a tampon which can be inserted into the cervix and withdrawn once an appropriate cervical ripening or other desirable affect on the female reproductive system has been produced. 20 It should be understood that in addition to the ingredients particularly mentioned above the fonulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents. 25 50 WO 2005/053706 PCT/GB2004/005087 EXAMPLE 3 - Exemplafy pharmaceuticalformulations Whilst it is possible for a compound of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one 5 or more acceptable carriers. The carrier(s) must be "acceptable" in the sense of being compatible with the compound of the invention and not deleterious to the recipients thereof. Typically, the carriers will be water or saline which will be sterile and pyrogen-free. 10 The following examples illustrate pharmaceutical formulations according to the invention in which the active ingredient is a compound of the invention. Example 3A: Tablet 15 Active ingredient 100 mg Lactose 200 mg Starch 50 mg Polyvinylpyrrolidone 5 mg Magnesium stearate 4 mg 20 359 mg Tablets are prepared from the foregoing ingredients by wet granulation followed by compression. 25 51 WO 2005/053706 PCT/GB2004/005087 Example 3B: Ophthahnic Solution Active ingredient 0.5 g Sodium chloride, analytical grade 0.9 g 5 Thiomersal 0.001 g Purified water to 100 ml pH adjusted to 7.5 Example 3C: Tablet Formulations 10 The following formulations A and B are prepared by wet granulation of the ingredients with a solution of povidone, followed by addition of magnesium stearate and compression. 15 Formulation A ma/tablet m/tablet (a) Active ingredient 250 250 (b) Lactose B.P. 210 26 (c) Povidone B.P. 15 9 20 (d) Sodium Starch Glycolate 20 12 (e) Magnesium Stearate 5 3 500 300 25 Formulation B m2/tablet ma/tablet (a) Active ingredient 250 250 (b) Lactose 150 (c) Avicel PH 101* 60 26 30 (d) Povidone B.P. 15 9 (e) Sodium Starch Glycolate 20 12 52 WO 2005/053706 PCT/GB2004/005087 (f) Magnesium Stearate 5 3 500 300 5 Formndation C ma/tablet Active ingredient 100 Lactose 200 Starch 50 10 Povidone 5 Magnesium stearate 4 359 15 The following fonnulations, D and E, are prepared by direct compression of the admixed ingredients. The lactose used in formulation E is of the direction compression type. Forniulation D 20 ma/capsule Active Ingredient 250 Pregelatinised Starch NF15 150 400 25 Formulation E mg/capsule Active Ingredient 250 Lactose 150 Avicel * 100 30 500 53 WO 2005/053706 PCT/GB2004/005087 Formulation F (Controlled Release Formulation) The fonmulation is prepared by wet granulation of the ingredients (below) 5 with a solution of povidone followed by the addition of magnesium stearate and compression. m/tablet Active Ingredient 500 Hydroxypropylmethylcellulose 112 10 (Methocel K4M Premiurm)* Lactose B.P. 53 Povidone B.P.C. 28 Magnesium Stearate 7 15 700 Drug release takes place over a period of about 6-8 hours and was complete after 12 hours. Example 3D: Capsule Formulations 20 Formulation A A capsule formulation is prepared by admixing the ingredients of Formulation D in Example C above and filling into a two-part hard gelatin 25 capsule. Formulation B (infi-a) is prepared in a similar manner. Formulation B ma/capsule Active ingredient 250 30 Lactose B.P. 143 Sodium Starch Glycolate 25 54 WO 2005/053706 PCT/GB2004/005087 Magnesium Stearate 2 420 5 Formzulation C ma/capsule Active ingredient 250 Macrogol 4000 BP 350 10 600 Capsules are prepared by melting the Macrogol 4000 BP, dispersing the active ingredient in the melt and filling the melt into a two-part hard gelatin capsule. 15 Fornulation D mg/capsule Active ingredient 250 Lecithin 100 Arachis Oil 100 20 450 Capsules are prepared by dispersing the active ingredient in the lecithin and arachis oil and filling the dispersion into soft, elastic gelatin capsules. 25 Formulation E (Controlled Release Capsule) The following controlled release capsule formulation is prepared by extruding ingredients a, b, and c using an extruder, followed by spheronisation of the 30 extrudate and drying. The dried pellets are then coated with release controlling membrane (d) and filled into a two-piece, hard gelatin capsule. 55 WO 2005/053706 PCT/GB2004/005087 mg/capsule Active ingredient 250 Microcrystalline Cellulose 125 Lactose BP 125 5 Ethyl Cellulose 13 513 Example 3E: Injectable Formulation 10 Active ingredient 0.200 g Sterile, pyrogen free phosphate buffer (pH7.0) to 10 nl The active ingredient is dissolved in most of the phosphate buffer (35-40'C), 15 then made up to volume and filtered through a sterile imicropore filter into a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals. Example 3F: Intramuscular injection 20 Active ingredient 0.20 g Benzyl Alcohol 0.10 g Glucofurol 75* 1.45 g Water for Injection q.s. to 3,00 ml 25 The active ingredient is dissolved in the glycofurol. The benzyl alcohol is then added and dissolved, and water added to 3 ml. The mixture is then filtered through a sterile micropore filter and sealed in sterile 3 ml glass vials (type 1). 30 56 WO 2005/053706 PCT/GB2004/005087 Example 3G: Syrup Suspension Active ingredient 0.2500 g Sorbitol Solution 1.5000 g 5 Glycerol 2.0000 g Dispersible Cellulose 0.0750 g Sodium Benzoate 0.0050 g Flavour, Peach 17.42.3169 0.0125 ml Purified Water q.s. to 5.0000 ml 10 The sodium benzoate is dissolved in a portion of the purified water and the sorbitol solution added. The active ingredient is added and dispersed. In the glycerol is dispersed the thickener (dispersible cellulose). The two dispersions are mixed and made up to the required volume with the purified water. 15 Further thickening is achieved as required by extra shearing of the suspension. Example 3H: Suppository mg/suppository Active ingredient (63 im)* 250 20 Hard Fat, BP (Witepsol H15 - Dynamit Nobel) 1770 2020 *The active ingredient is used as a powder wherein at least 90% of the 25 particles are of 63 .m diameter or less. One fifth of the Witepsol H15 is melted in a steam-jacketed pan at 45"C maximum. The active ingredient is sifted through a 200 pLm sieve and added to the molten base with mixing, using a silverson fitted with a cutting head, 30 until a smooth dispersion is achieved. Maintaining the mixture at 45"C, the remaining Witepsol H15 is added to the suspension and stirred to ensure a 57 WO 2005/053706 PCT/GB2004/005087 homogenous mix. The entire suspension is passed through a 250 sim stainless steel screen and, with continuous stirring, is allowed to cool to 40'C. At a temperature of 38"C to 40'C 2.02g of the mixture is filled into suitable plastic moulds. The suppositories are allowed to cool to room temperature. 5 Example 31: Pessaries mg/pessary Active in-redient 250 Anhydrate Dextrose 380 10 Potato Starch 363 Maonesium Stearate 7 1000 15 The above ingredients are mixed directly and pessaries prepared by direct compression of the resulting mixture. Example 3J: Creams and ointments 20 Described in Remington. Example 3K: Microsphere fornulations The compounds of the invention may also be delivered using microsphere 25 fornulations, such as those described in Cleland (1997; 200 1). Example 3L: Dry Powder Inhalation The compounds of the invention may be delivered by inhalation, with the 30 aid of a dry powder inhaler delivering micronised particles in metered quantities as described in Ansel (1999). 58 WO 2005/053706 PCT/GB2004/005087 Example 3M: Aerosol Inhalation The compounds of the invention may be delivered by inhalation, with the 5 aid of a suitable inhaler delivering micronised particles in metered quantities employing a non CFC propellant as described in Ansel (1999). 59 WO 2005/053706 PCT/GB2004/005087 References Allport VC, Pieber D, Slater DM, Newton R, White JO, Bennett PR. Human labour is associated with nuclear factor-xB activity which mediates 5 cyclo-oxygenase-2 expression and is involved with the functional progesterone withdrawal, Mol Hun Reprod 2001; 7: 581-5 86. Ansel. Pharmaceutical Dosage Forms and Drug Delivery Systems, 1999, Lippincott Williams and Wilkins. 10 Bendixen AC, Shevde NK, Dienger KM, Willson TM, Funk CD, Pike 1W. IL-4 inhibits osteoclast formation through a direct action on osteoclast precursors via peroxisome proliferator-activated receptor 41. Proc Nat! AcadSci USA 2001; 98: 2443-2448. 15 Bennett PR, Rose MP, Myatt L, Elder MG. Preterm labor: stimulation of arachidonic acid metabolism in human amnion cells by bacterial products. Am JObstet Gynecol. 1987, 156:649-5. 20 Bennett, P. and Slater, D. The role of cyclo-oxygenases in the onset of labour., in improved non-steroid anti-inflammatory drugs: COX-2 enzyme inhibitors. J. Vane, R. Botting, and G. Botting, Editors. 1996, Kluwer Academic: London. P. 112-118. 25 Brown, N.L., Alvi, S.A., Elder, M.G., Bennett, P.R. and Sullivan, M.H. A spontaneous induction of fetal nembrane prostaglandin production precedes clinical labour. JEndocrinol 1998; 157: R1-R6. Carlon et al., Obstet Gynecol, 1992. 85(5), 769-774. 30 60 WO 2005/053706 PCT/GB2004/005087 Chinetti G, Griglio S, Antonucci M, Torra IP, Delerive P, Majd Z, Fruchart J-C, Chapman J, Najib J, Staels B. Activation of proliferator-activated receptors a and y induces apopotosis of human monocyte-derived macrophages. JBiol Chem 1998; 273: 25573-25580. 5 Cleland, J.L. (1997) Pharm. Biotechnol. 10:1-43. Cleland et al. (2001) J Control. Release 72:13-24. 10 Corey, E. J. The logic of chemical synthesis-mulitstep synthesis of complex carbogenic molecules. Angew Chem Int Ed Engl, 1991, 30:455-465. Crankshaw, D.J. and Dyal, R. Effects of some naturally occurring prostanoids and some cyclo-oxygenase inhibitors on the contractility of the 15 human lower uterine segment in vitro. Can J Physiol Pharmacol, 1994. 72(8): p. 870-4. Dammann 0 and Leviton A. Brain damage in preterm newborns: Might enhancement of developmentally regulated endogenous protection open a 20 door for prevention? Pediatrics 1999; 104: 541-550. Derossi et al. (1998), Trends Cell Biol 8, 84-87. Dignam et al., Accurate transcription initiation by RNA polymerase II in a 25 soluble extract from isolated mammalian nuclei. Nucleic Acids Res., 11:1475-1489. Dyal R and Cranskshaw DJ. The effects of some synthetic prostanoids on the contractility of the human lower uterine segment in vitro. Am J Obstet 30 Gynecol 1988; 158: 281-285. 61 WO 2005/053706 PCT/GB2004/005087 Elliot CL, Loudon JA, Brown N, Slater DM, Bennett PR, Sullivan MH. IL lbeta and IL-8 in human fetal membranes: changes with gestational age, labor, and culture conditions. Am J Reprod Immunol 2001; 46: 260-267. 5 Ferry G, Bruneau V, Beauverger P, et al. 2001 Binding of prostaglandins to human PPARgamma: tool assessment and new natural ligands. Eur J Pharnacol., 417:77-89. Forman BM, Tontonoz P, Chen J, Brun RP, Spiegehnan BM, Evans RM. 10 15-deoxy-A -prostaglandin J 2 is a ligand for the adipocyte determination factor PPARy. Cell 1995; 83: 803-812. Ganstrom et al., Acta Obstet Gynecol Scand, 1987. 66:429-431. 15 Garfield RE and Hertzberg EL. Cell-to-cell coupling in the myometrium: Emil Bozier's prediction. Prog Clin Biol Res 1990; 327: 673-681. Gibson CS, MacLennen AH, Goldwater PN, Dekker GA. Antenatal causes of cerebral palsy: associations between inherited thrombophilias, viral and 20 bacterial infection, and inherited susceptibility to infection. Obstet Gynecol Survey 2003; 58: 209-220. Gupta RA, Polk DB, Krishna U, Israel DA, Yan F, DuBois RN, Peek RM Jr. Activation of peroxisome proliferator-activated receptor y suppresses 25 nuclear factor xB-mediated apoptosis induced by Helicobacter Pylori I gastric epithelial cells. JBiol Chem 2001; 276: 31059-31066. Herman A, Groutzd A, Bukovsky I, Arieli S, Sherman D, Caspi E. A simplified pre-induction scoring method for the prediction of successful 30 vaginal delivery based on multivariate analysis of pelvic and other obstetrical factors. JPerinat Med. 1993, 21:117-24. 62 WO 2005/053706 PCT/GB2004/005087 Huang JT, Welch JS, icote M, Binder CJ, Willson TM, Kelly C, Witztum JL, Funk CD, Conrad D, Glass CK. Interleukin-4-dependent production of PPAR-y ligands in macrophages by 12/15-lipoxygenase. Nature 1999; 400: 5 378-382. Ikawa H, Kameda H, Kamitani H, Back SJ, Nixon JB, His LC, Eling TE. Effect of PPAR activators on cytokine-stimulated cyclooxygenase-2 expression in human colorectal carcinoma cells. Exp Cell Res 2001; 267: 10 73-80. Johnson C, Van Antwerp D, Hope TJ. An N-terminal nuclear export signal is required for the nucleocytoplasmic shuttling of IxBca. EMBO J 1999; 23: 6682-6693. 15 Keelan JA, Marvin KW, Sato TA, Coleman M, McCowan LM, Mitchell MD. Cytokine abundance in placental tissues: evidence of inflammatory activation in gestational membranes with term and preterm parturition. Am JObstet Gynecol 1999; 181: 1530-1536. 20 Keirse (1995) "Indomethacin tocolysis in pre-term labour" in Pregnancy and Childbirth Module (Eds. Enkin, M.W., Keirse, MJ.N.C., Renfrew, M.J., Neilson, J P.) Cochrane Database of Systematic Reviews, No 04383, Oxford). 25 Kelly RW. Inflammatory mediators and cervical ripening. J Reprod Immnunol 2002; 57: 217-224. Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, Lebmann JM 30 1995 A prostaglandin J2 metabolite binds peroxisone proliferator-activated receptor gamma and promotes adipocyte differentiation. Cell 83:813-9. 63 WO 2005/053706 PCT/GB2004/005087 Kunsch C, Ruben SM, Rosen CA. Selection of optimal kappa B/Rel DNA binding motifs: interaction of both subunits of NF-lkappaB with DNA is required for transcriptional activation. Mol Cell Biol 1992; 12: 4412-4421. Lee, Curr. Opin. Biotechnol., 2001, 11:81-84. Lee Y, Allport V, Sykes A, Lindstrom T, Slater D. Bennett P. The effects of labour and of interleukin 1 beta upon the expression of nuclear factor 10 kappa B related proteins in human amnion. Mol Hum Reprod 2003; 9: 213 8. Leesnitzer LM, Parks DJ, Bledsoe RK, Cobb JE, Collins JL, Consler TG, Davis RG, Hull-Ryde EA, Lenhard JM, Patel L, Plunket KD, Shenk JL, 15 Stirnmel JB, Therapontos C, Willson TM, Blanchard SG. Functional consequences of cysteine modification in the ligand binding sites of peroxisome proliferator activated receptors by GW9662. Biochem 2002; 41: 6640-50. 20 Li Q and Verma IM. NF-KB regulation in the immune system. Nature Reviews, 2002. 2:725-735. Lin R, Gewert D, Hiscott J. Differential transcriptional activation in vitro by NF-kB/Rel proteins. JBiol Chem 1995; 270: 3123-3131. 25 Maul H, Nagel S, Welsch G, Schafer A, Winkler M, Rath W. Messenger ribonucleic acid levels of interleukin- 1 beta, interleukin-6 and interleukin-8 in the lower uterine segment increased significantly at final cervical dilatation during term parturition, while those of tumor necrosis factor alpha 30 remained unchanged. Eu- J Obstet Gynecol Reprod Biol 2002; 102:143-7. 64 WO 2005/053706 PCT/GB2004/005087 Meade EA, McIntyre TM, Zinnernan GA, Prescott SM. Peroxisome proliferators enhance cyclooxygenase-2 expression in epithelial cells. J Biol Chem 1999; 274: 8328-8334. 5 Mitchell MD, Edwin SS, Lundin-Schiller S, Silver RM, Smotkin D, Trautman MS. Mechanism of interleukin-1 beta stimulation of human amnion prostaglandin biosynthesis: mediation via a novel inducible cyclooxygenase. Placenta 1993; 14: 615-625. 10 Miyahara T, Schrum L, Rippe R, Xiong S, Yee HF Jr, Motomura K, Anania FA, Willson TM, Tsukamoto H. Peroxisome proliferator-activated receptors and hepatic stellate cell activation. J Biol Chem 2000; 275: 35715-35722. 15 Moise et al. Effect of advancing gestational age on the frequency of fetal ductal constriction in association with maternal indomethacin use" Am. J Obster. Gynecol., 1995, 170(45), 1204-5. Narumiya S. Structures, properties and distributions of prostanoid 20 receptors. Adv Prost Thromb Leuk Res 1995; 23: 17-22. Narumiya S, Ohno K, Fukushima M, Fujiwara M. Site and mechanism of growth inhibition by prostaglandins. III. Distribution and binding of prostaglandin A2 and delta 12-prostaglandin J2 in nuclei. JPharmacol Exp 25 Ther 1987; 242: 306-11. Nasuhura et al., JBC, 1999. 274:19965. Pang L, Nie M, Corbett L, Knox AJ. Cyclooxygenase-2 expression by 30 nonsteroidal anti-inflanunatory drugs in human airway smooth muscle cells: 65 WO 2005/053706 PCT/GB2004/005087 Role of peroxisome proliferator-activated receptors. J Jmmunol 2003: 170: 1043-1051. Phelps CB, Sengchanthalangsy LL, Malek S, Ghosh G. Mechanism of 0B 5 DNA binding by Rel/NF-kB dimers. JBiol Chem 2000; 275: 24392-24399. Pieber D, Allport VC, Hills F, Johnson M, Bennett PR. Interactions between progesterone receptor isoforms in myometrial cells in human labour. Mol Hum Reprod. 2001, 7:875-9. 10 Prusty D, Park B-H, Davis KE, Farmer SR. Activation of MEK/ERK signaling promotes adipogenesis by enhancing peroxisome proliferators activated receptor y (PPARy) and C/EBPt gene expression during the differentiation of 3T3-L1 preadipocytes. JBiol Chem 2002; 277: 46226 15 46232. Rasanen and Jouppila. Fetal cardiac function and ductus arteriosus during indomethacin and sulindac therapy for threatened pre-term labour; A randomised study. Am J Obstet Gynecol 1995. 173(1), 20-25. 20 Remington: The Science and Practise of Pharmacy, 19 th ed., The Philadelphia College of Pharmacy and Science, ISBN 0-912734-04-3. Responded et al., Fetal echocardiography during indomethacin treatment. 25 Ultrasound Obstet Gynecol, 1995. 5, 86-89. Romero R, Parvizi ST, Oyarzun E, Mazor \4, Wu YK, Avila C. Athanassiadis AP, Mitchell MID. Amniotic fluid interleukin-1 in spontaneous labour at tenn. JReprod Med 1990; 35: 235-238. 30 66 WO 2005/053706 PCT/GB2004/005087 Romero R, Espinoza J, Chaiworapongsa T, Kalache K. Infection and prematurity and the role of preventive strategies. Senin Neonatol. 2002; 7:259-74. 5 Ruan H, Pownall HJ, Lodish HF. Troglitazone antagonizes TNF-c-induced reprogramnming of adipocyte gene expression by inhibiting the transcriptional regulatory functions of NF-i<B. J Biol Chem 2003; Manuscript M303 141200. 10 Rush RW, Keirse MJNC, Howat P, Baum JD, Anderson AB, Turnbull AC. Contribution of preterm delivery to perinatal mortality. Br Med J 1976; 2: 965. Satoh K, Yasumizu T, Fukuoka H, Kinoshita K, Kaneko Y, Tsuchiya M, 15 S akamoto S. Prostaglandin F2 alpha metabolite levels in plasma, amniotic fluid, and urine during pregnancy and labor. Am J Obstet Gynecol 1979; 133: 886-890. Sambrook et al., Molecular Cloning. A laboratory manual. 1989. Cold 20 Spring Harbour pub. Schreiber et al., Rapid detection of octomer binding proteins with mini extracts prepared form a small number of cells. Nucl. Acids Res, 1989. 17:6419. 25 Skinner KA and Challis JR. Changes in the synthesis and metabolism of prostaglandins by human fetal membranes and decidua at labour. Am J Obstet Gynecol 1985; 151: 519-523. 67 WO 2005/053706 PCT/GB2004/005087 Slater DM, Berger L, Newton R, Moore GE, Bennett PR. Changes in the expression of types 1 and 2 cyclo-oxygenase in human fetal membranes at term. Am JObstet Gynecol 1995; 172: 77-82. 5 Staels B, Koenig W, Habib A, Merval R, Lebret M, Torra IP, Delerive P, Fadel A, Chinetti G, Fruchart JC, Najib f, Maclouf J, Tedgui A. Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARganma activators. Nature 1998; 393: 790-793. 10 Straus D. S. and Glass C. K. Cyclopentenone prostaglandins: new insights on biological activities and cellular targets. Med Res Rev, 2001. 21:185 210. Subbaramaiah K, Lin DT, Hart JC, Dannenberg AJ. Peroxisome 15 proliferator-activated receptor y ligands suppress the transcriptional activation of cyclooxygenase-2. JBiol Chem 2001; 276: 12440-12448. Suyang H, Phillips R, Douglas I, Ghosh S. Role of unphosphorylated, newly synthesized IxB f in persistent activation of NF-B. Mol Cell Biol 20 1996; 16: 5444-5449. Suzawa M, Takada I, Yanagisawa J, Ohtake F, Ogawa S, Yanauchi T, Kadowaki T, Takeuchi Y, Shibuya H, Gotoh Y, Matsumoto K, Kato S. Cytokines suppress adipogenesis and PPAR-y function through the 25 TAKl/TABl/NIK cascade. Nature Cell Biol2003; 5: 224-230. Suzuki et al., Three component coupling synthesis of prostaglandins. A simplified, general procedure. Tetrahedron, 1990, 46:4809-4822. 30 Takata Y, Kitami Y, Yang Z-H, Nakamura M, Okura T, Hiwada K. Vascular inflammation is negatively autoregulated by interaction between 68 WO 2005/053706 PCT/GB2004/005087 CCAAT/enhancer-binding protein-6 and peroxisome proliferator-activated receptor-y. Circ Res 2002; 91: 427-433. Takeuchi et al. Adv. Drug. Delic. Rev., 2001, 47:39-54. 5 Tanaka T, Itoh H, Doi K, Fukunaga Y, Hosoda K, Shintani M, Yamashita J, Chun TH, Inoue M, Masatsugu K, Sawada N, Saito T, Inoue G, Nishimura H, Yoshimasa Y, Nakao K. Down regulation of peroxisome proliferator activated receptor g expression by inflannatory cytokines and its reversal 10 by thiazolidinediones. Diabetologia 1999; 42: 702-710. Toledano M, Ghosh D, Trinh F, Leonard WJ. N-terminal DNA-binding domains contribute to differential DNA-binding specificities of NF-kappa B p50 and p65. Mol Cell Biol 1993; 13: 852-860. 15 Tontonoz P, Nagy L, Alvarez J, Thomazy V, Evans R. PPARy promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell 1998; 93: 241-252. 20 Tulzer et al., Doppler- -echocardiography of fetal ductus arteriosus constriction versus increased right ventricular output. JA CC, 1991. 18(2), 532-36. Turnbull, A. The fetus and birth, in Elsevier, London. 1977. 25 Urakubo A, Jarskog LF, Lieberman JA, Gilmore JH. Prenatal exposure to maternal infection alters cytokine expression in the placenta, amnuiotic fluid, and fetal brain. Schizophrenia research 2001; 47: 27-36. 69 WO 2005/053706 PCT/GB2004/005087 Van Meir, C.A., er al., Chorionic prostaglandin catabolism is decreased in the lower uterine segment with term labour. Placenta, 1997. 18(2-3): p.109 14. 5 Van Meir, C.A., et al., Immunoreactive 15-hydroxyprostaglandin dehydrogenase (PGDH) is reduced in fetal membranes from patients at pre term delivery in the presence of infection. Placenta, 1996. 17(5-6): p.291-7. Volpe IJ. Neurobiology of periventricular leukomalacia in the premature 10 infant. Pediatr Res 2001;50:553-62. Ward C, Dransfield I, Murray J, Farrow SN, Haslett C, Rossi AG. Prostaglandin D 2 and its metabolites induce caspase-dependent granulocyte apoptosis that is mediated via inhibition of IicBa degradation using a 15 peroxisome proliferator-activated receptor-y-independent mechanism. J 1mmunol2002; 168: 6232-6243. Whiteside ST, Epinat J-C, Rice NR, Israel A. I kappa B epsilon, a novel member of the IicB family, controls RelA and cRel NF-icB activity. EMBO 20 J1997(b); 16: 1413-1426. Zhou YC and Waxman DJ. Cross-talk between Janus-Kinase-signal transducer activator of transcription (JAK-STAT) and peroxisome proliferator-activated a (PPARa.) signaling pathways. J Biol Chem 1999; 25 274: 2672-2681. 30 70

Claims (39)

1. Use of a cyclopentenone prostaglandin in the manufacture of a medicament for delaying the onset and/or preventing the continuation of labour in a female.
2. Use -of a cyclopentenone prostaglandin in the manufacture of a medicament for preventing and/or reducing an inflammatory response in the reproductive system of a female.
3. A use according to Claim 2 wherein the female is pregnant.
4. A use according to Claim 1 or 3 wherein the female is human and the duration of pregnancy is more than approximately 13 weeks.
5. A use according to Claim 4 wherein the duration of pregnancy is approximately between 20 and 32 weeks.
6. A use according to any preceding claim wherein the medicament reduces and/or prevents an inflammatory response in the reproductive system of a female associated with the onset or continuation of labour.
7. A use according to any preceding claim wherein the medicament reduces and/or prevents an inflammatory response in the reproductive system of a female associated with infection by a pathogenic agent.
8. A use according to Claim 7 wherein the pathogenic agent is viral, bacterial or fungal. 71 WO 2005/053706 PCT/GB2004/005087
9. A use according to Claim 6 wherein the inflanunatory response is activated by stretch of the uterus.
10. A use according to any preceding claim wherein the medicament reduces and/or prevents one or more of the following conditions: pre term labour; pathogenic infection; cervical ripening, uterine contractions.
11. A use according to any preceding claim wherein the medicament reduces and/or prevents fetal or neonatal damage.
12. A use according to Claim 11 wherein the fetal or neonatal damage is brain damage.
13. A use according to Claim 12 wherein the fetal or neonatal damage is one or more of the following conditions: astrogliosis; loss of myelin producing oligodendrocytes; multifocal necroses resulting in cystic change (periventricular leucomalacia, PVL).
14. A use according to any preceding claim wherein the cyclopentenone prostaglandin is 15-deoxy-A' 2 '4 -prostaglandin J 2 and/or prostaglandin A 1 and/or is a prodrug of 15-deoxy-A 12 " 4 prostaglandin J 2 and/or prostaglandin A 1 .
15. A use according to Claim 14 wherein the prodrug is PGD 2 or PGE 1 .
16. A use according to any preceding claim wherein the medicament further comprises a pharmaceutically acceptable excipient, diluent or carrier. 72 WO 2005/053706 PCT/GB2004/005087
17. A use according to any preceding claim wherein the medicament is in a fonn adapted for delivery by mouth.
18. A use according to any preceding claim wherein the medicament is in a form adapted for delivery by intravenous injection.
19. A use according to any preceding claim wherein the medicament is in a form adapted for delivery by intra-amniotic injection.
20. A use according to any preceding claim wherein the medicament is in a form which is compatible with the amniotic fluid.
21. A use according to any preceding claim wherein the medicament further comprises an agent for treating a female who has or is at risk of one or more of the following conditions: pre-term labour; pathogenic infection; cervical ripening, uterine contractions.
22. A use according to Claim 21 wherein the agent is a corticosteroid.
23. A use according to Claim 21 or 22 wherein the agent is capable of preventing and/or reducing respiratory distress syndrome in the neonate.
24. A use according to Claim 23 wherein the agent is selected from dexamethasone or betamethasone.
25. A use according to Claim 21 wherein the condition is preterm labour and the agent is capable of delaying delivery.
26. A use according to Claim 21 wherein the condition is uterine contractions and the agent is a tocolytic agent. 73 WO 2005/053706 PCT/GB2004/005087
27. A use according to Claim 26 wherein the tocolytic agent is selected from oxytocin receptor antagonists, calcium channel blockers, sympathomimetics, nitric oxide donors.
28. A use according to Claim 27 wherein the oxytocin receptor antagonist is Atosiban.
29. A use according to Claim 27 wherein the calcium channel blocker is Nifedipine.
30. A use according to Claim 27 wherein the syrnpathomimetic is Ritodrine.
31. A use according to Claim 27 wherein the nitric oxide donor is glyceryl trinitrate.
32. A use according to any preceding claim wherein the inflammatory response is mediated by NFicB in uterine cells.
33. A use according to Claim 32 wherein the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFicB activity by preventing and/or reducing NFicB DNA-binding in uterine cells.
34. A use according to Claim 33 wherein the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFicB activity by preventing and/or reducing NFicB-mediated transcriptional regulation in uterine cells. 74 WO 2005/053706 PCT/GB2004/005087
35. A use according to Claim 34 wherein the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFirB activity by preventing and/or reducing NFKB production in uterine cells.
36. A pharmaceutical composition comprising a cyclopentenone prostaglandin and a pharmaceutically acceptable carrier or exipient, the cyclopentenone prostaglandin being present in an amount effective to prevent and/or reduce an inflammatory response in the reproductive System of a female.
37. A method of treating inflammation within the reproductive system of a female, the method comprising administering an effective amount of a medicament as defined in any one of the preceding claims to a subject in need thereof.
38. A method for identifying a cyclopentenone prostaglandin for delaying the onset and/or preventing the continuation of labour in a female comprising the step of testing the cyclopentenone prostaglandin to determine if it is capable of inhibiting and/or reducing NFicB activity in uterine cells in a PPAR-y independent manner.
39. A method for making a pharmaceutical composition for use in delaying the onset and/or preventing the continuation of labour in a female comprising providing a cyclopentenone prostaglandin identified by the method of Claim 38 and combining it with a pharmaceutically acceptable carrier. 75
AU2004294800A 2003-12-02 2004-12-02 Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour Abandoned AU2004294800A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB0327975.9A GB0327975D0 (en) 2003-12-02 2003-12-02 Methods of treatment
GB0327975.9 2003-12-02
PCT/GB2004/001380 WO2005053705A1 (en) 2003-12-02 2004-03-29 Use of a cyclopentenone prostaglandin for delaying the onset and/or preventing the continuation of labour
AUPCT/GB2004/001380 2004-03-29
PCT/GB2004/005087 WO2005053706A1 (en) 2003-12-02 2004-12-02 Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour

Publications (1)

Publication Number Publication Date
AU2004294800A1 true AU2004294800A1 (en) 2005-06-16

Family

ID=34655231

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2004294800A Abandoned AU2004294800A1 (en) 2003-12-02 2004-12-02 Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour

Country Status (5)

Country Link
EP (1) EP1827451A1 (en)
JP (1) JP2007513133A (en)
AU (1) AU2004294800A1 (en)
CA (1) CA2589908A1 (en)
WO (1) WO2005053706A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015008166A2 (en) * 2013-07-12 2015-01-22 Casey Patrick J Method for the harvesting, processing, and storage of proteins from the mammalian feto-placental unit and use of such proteins in compositions and medical treatment

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9418483D0 (en) * 1994-09-14 1994-11-02 British Tech Group Compound for use in pre-term labour
WO1999018942A1 (en) * 1997-10-10 1999-04-22 Imperial College Innovations Ltd. Use of csaidtm compounds for the management of uterine contractions

Also Published As

Publication number Publication date
WO2005053706A1 (en) 2005-06-16
EP1827451A1 (en) 2007-09-05
JP2007513133A (en) 2007-05-24
CA2589908A1 (en) 2005-06-16

Similar Documents

Publication Publication Date Title
Olson The role of prostaglandins in the initiation of parturition
Sugimoto et al. Distribution and function of prostanoid receptors: studies from knockout mice
Hertelendy et al. Regulation of myometrial smooth muscle functions
Yu et al. Differential impact of prostaglandin H synthase 1 knockdown on platelets and parturition
Reinl et al. Na+-leak channel, non-selective (NALCN) regulates myometrial excitability and facilitates successful parturition
Wahid et al. Toll-like receptor-4 antagonist (+)-naltrexone protects against carbamyl-platelet activating factor (cPAF)-induced preterm labor in mice
Nicol et al. Becoming female: ovarian differentiation from an evolutionary perspective
US20070282004A1 (en) Use of a Cyclopentenone Prostaglandin for Delaying for the Onset and/or Preventing the Continuation of Labour
Russell et al. Ovulation: the coordination of intrafollicular networks to ensure oocyte release
AU2004294800A1 (en) Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour
Li et al. The role of endogenous Smad7 in regulating macrophage phenotype following myocardial infarction
Mitchell et al. Prostaglandin endoperoxide H synthase inhibitors and other tocolytics in preterm labour
US9161925B2 (en) Therapeutic uses of beta-3 adrenergic receptor agonist derivatives in particular to modulate apoptosis
Sunita et al. Histone deacetylase: A potential therapeutic target for ovarian dysfunction
JP4663968B2 (en) EG-VEGF / prokineticin 2-receptor antagonist
US20030220266A1 (en) Method of treating a disease
Zhao et al. Na+-Leak Channel, Non-Selective (NALCN) Regulates Myometrial Excitability and Facilitates Successful Parturition.
US11464831B2 (en) Compositions and methods using IL-8 for improving health of mammals
Pearson Adiponectin in equine reproduction
Habelrih et al. Preventing Preterm Birth: Exploring Innovative Solutions
Liao et al. Uterine receptor activation in response to uterotonic agents in late-pregnancy rat myometrium following pretreatment with oxytocin: an in vitro study
Reinl et al. Please let us know how this document benefits you.
Kohan-ghadr Morphological and functional characterization of placenta during gestation in bovine clones derived by somatic nuclear transfer
Lee et al. Progesterone-regulated genes in the endometrium
Simmons Insulin-Like Growth Factor Binding Proteins-1 and-3, and Hydroxysteroid (11-Beta) Dehydrogenase One: Potential Roles in Ruminant Conceptus Development and Endometrial Function

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application