WO2005053706A1 - 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
WO2005053706A1
WO2005053706A1 PCT/GB2004/005087 GB2004005087W WO2005053706A1 WO 2005053706 A1 WO2005053706 A1 WO 2005053706A1 GB 2004005087 W GB2004005087 W GB 2004005087W WO 2005053706 A1 WO2005053706 A1 WO 2005053706A1
Authority
WO
WIPO (PCT)
Prior art keywords
use according
prostaglandin
labour
female
medicament
Prior art date
Application number
PCT/GB2004/005087
Other languages
French (fr)
Inventor
Phillip Robert Bennett
Tamsin LINDSTRÖM
Original Assignee
Imperial College Innovations Limited
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 Limited filed Critical Imperial College Innovations Limited
Priority to EP04805915A priority Critical patent/EP1827451A1/en
Priority to US10/581,532 priority patent/US20070282004A1/en
Priority to JP2006542016A priority patent/JP2007513133A/en
Priority to CA002589908A priority patent/CA2589908A1/en
Priority to AU2004294800A priority patent/AU2004294800A1/en
Publication of WO2005053706A1 publication Critical patent/WO2005053706A1/en

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

Definitions

  • the present invention relates to agents for improving perinatal outcome in pre-term labour.
  • the present invention relates to the use of prostaglandins to prevent and/or reduce an inflammatory response in the reproductive system of a female, thereby delaying the onset of labour.
  • Pre-term labour defined as spontaneous labour occurring prior to 37 weeks of gestation (with 39 weeks being term) continues to be a major 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.
  • the uterus 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 needs to begin contracting.
  • PGs increase in maternal urine and blood and in fetal membranes in association with labour (Satoh et al, 1919; Skinner and Challis, 1985).
  • PGE 2 stimulates uterine contractions (Dyal and Crankshaw, 1985), indirectly increases fundamentally dominant myometrial contractility by upregulation of oxytocin receptors and synchronisation of contractions (Garfield et al, 1990), and acts in concert with LL-8 to remodel the cervix (reviewed in Kelly, 2002).
  • NFKB Nuclear Factor Kappa B
  • IL-6 interleukin-6
  • COX-1 and COX-2 COX-1 and COX-2
  • COX genes are also referred to as prostaglandin H synthase or PG synthase.
  • the resulting inflammatory infiltrate mediated by the cytokines
  • increase in prostaglandin synthesis leads to cervical ripening, fetal membrane rupture and myometrial contractions.
  • NF- ⁇ B/Rel Five members of the NF- ⁇ B/Rel family have been identified in mammals: NF- ⁇ Bl (p50 and its precursor pl05), NF- ⁇ B2 (p52 and its precursor plOO), p65 (RelA), c-rel, and Rel B. These proteins share a structurally conserved amino-terminal region termed the Rel homology domain (RHD).
  • the RHD is responsible for dimerisation, DNA binding, and interaction with the inhibitors of kappa B (Ii B) proteins. It also contains a nuclear localisation signal (NLS).
  • NF- ⁇ B In its active DNA-binding form NF- ⁇ B consists of heterogeneous dimers of various combinations of NF- ⁇ B subunits: each member of the NF-i B family, except for Rel B, can form homodimers, as well as heterodimers with one another.
  • the p65, c-rel and Rel B proteins contain a carboxy-te ⁇ ninal non-homologous transactivation domain, which activates transcription from i B sites in target genes; in contrast, p50 and p52 proteins lack a transactivation domain.
  • the various NF-icB dimers exhibit different binding affinities for specific KB sites (Kunsch et al, 1992, Phelps et al, 2000), and differentially stimulate transcription through distinct icB elements (Lin et al, 1995).
  • NF-icB dimers are normally sequestered in an inactive form in the cytoplasm by association with the inhibitory I B proteins, which include I ⁇ B ⁇ , Ii B ⁇ and I B ⁇ .
  • the Ii Bs are characterised by the presence of multiple ankyrin repeats which mediate binding to the RHD and mask the NLS of NF- B.
  • the major NF-icB signalling pathway which is activated by pro- inflammatory stimuli and LPS, targets IicB ⁇ - and IicB ⁇ -bound NF- ⁇ B (for review see Li and Verma 2002).
  • IKK I ⁇ B kinase
  • the LKK complex consists of several proteins, the main ones being IKK ⁇ (IKK1), LKK ⁇ (LKK2), and NF- B essential modulator (NEMO or IKK ⁇ ).
  • IKK1 IKK ⁇
  • LKK2 LKK ⁇
  • NEMO NF- B essential modulator
  • This modification targets IicB ⁇ for rapid degradation by the 26S proteasome.
  • the degradation of the IicB inhibitor exposes the NLS of NF-icB resulting in translocation of the p50/p65 dimer to the nucleus where it can bind to cB sites in the promoter of target genes and promote transcription.
  • NF-icB The critical inhibitory step in NF-icB inactivation involves binding of newly synthesised I ⁇ B ⁇ to NF-icB in the nucleus. IicB ⁇ is quickly res3mthesised following its degradation. The newly synthesised IicB ⁇ is localised in the nucleus and displaces NFicB from its DNA binding sites. IicB ⁇ contains leucine-rich nuclear export sequences (NES) (Johnson et al 1999), which then enable it to transport NF-icB back to the cytoplasm, thereby completing an autoregulatory post-induction repression.
  • NES leucine-rich nuclear export sequences
  • IicB ⁇ In many cells nearly half of the NF-icB is sequestered by the other major IKB isoform, IicB ⁇ (Whiteside et al, 1991). In contrast to IicB ⁇ , IicB ⁇ is not NF-icB inducible and does not exert a rapid post-induction repression of NF-icB activity. Rather, IicB ⁇ has been implicated in persistent NF-icB activation. Prolonged exposure to certain stimuli, such as LPS, leads to the long-term induction of NF-icB activity despite high levels of newly synthesised IicB ⁇ .
  • I ⁇ B ⁇ is un-phosphorylated and, in contrast to IicB ⁇ or the constitutively phosphorylated I ⁇ B ⁇ , can interact with NF-icB bound to target promoters without displacing it from the DNA (Suyang et al, 1996).
  • This interaction of un-phosphorylated I ⁇ B ⁇ with DNA-bound NF-icB is thought to protect NF-icB from nuclear export, and thus inhibition, by IicB ⁇ , and the outcome is a sustained NF-KB response.
  • PGs are a family of biologically active molecules having a diverse range of 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 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).
  • PGs bind to prostanoid receptors localised on the cell surface and act through second messenger systems (Narumiya, 1995).
  • PGD 2 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
  • PPARs Peroxisome proliferator-activated receptors
  • They exist in three distinct forms, PPAR- ⁇ , PPAR- ⁇ , and PPAR- ⁇ , which form heterodimers with the retinoic X receptor (RXR) and bind to PPAR response elements (PPREs) in the promoter of target genes to induce transcription.
  • PPAR- ⁇ can also repress gene transcription by negatively interfering with the NF-icB, AP-1, STAT and C/EBP pathways (Zhou et al, 1999; Subbaramaiah et al, 2001; Takata et al, 2002; Suzawa et al, 2003).
  • 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 with prematurity (Darnmann et al, 1999).
  • inflammatory responses caused by mechanical stretching of the uterus may contribute to the onset of labour.
  • Mechanical stretchmg of the uterus occurs to an extent as a normal part of pregnancy and may be responsible for some of the biochemical changes which occur near to term and which cause the normal onset of labour at term.
  • mechanical stretch may occur where the uterus is overdistended by multiple pregnancy or by excess amniotic fluid (clinically termed hydramnios or polyhydramnios).
  • hydramnios or polyhydramnios
  • 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 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.
  • ⁇ -sympathomimetics such as Ritodrine, Salbutamol and Terbutaline
  • ⁇ -sympathomimetics cause significant maternal cardiovascular, respiratory and metabolic side effects and may lead to pulmonary oedema, cardiac failure and maternal death.
  • pulmonary oedema cardiac failure and maternal death.
  • tachyphylaxis and become ineffective after 24 to 48 hours.
  • Meta-analysis of randomised controlled trials has shown that the value of ⁇ -sympathomimetics is only in the temporary delay of labour to allow in utero transfer or administration of steroid to improve fetal lung surfactant production.
  • corticosteroids 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 corticosteroids improves the outcome for the pre-term 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 h surfactant production and therefore prevents or reduces the severity of neonatal respiratory problems.
  • Such agents are l ⁇ iown to those skilled hi the art.
  • tocolytic drugs are ox ⁇ rtocin receptor antagonists, calcium channel blockers, sympathomimetics and nitric oxide donors.
  • a commonly used oxytocin receptor antagonist is Atosiban, that functions by blocking the oxytocin receptor, thereby preventing activation of the receptor by endogenous oxytocin that stimulates uterine contractions.
  • a commonly 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 commonly used 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 the activity of myosin light chain Idnase, an enzyme essential for contractions.
  • Indomethacin a cyclo-oxygenase inhibitor
  • Indomethacin is effective in preventing the contractions of pre-term labour. It is more effective in short term prolongation of pregnancy than the ⁇ -sympathomimetics and, unlike ⁇ -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 occurs only in a proportion, increasing with gestational age from 10% at 26 weeks to 50%> at 32 weeks.
  • indomethacin is limited in clinical practice to use ⁇ 32 weeks, and to short courses ( ⁇ 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). Because of these side effects some obstetricians now use Sulindac, which appears to be equally good as a tocolytic (Carlon et al 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 Jouppila 1995). However, Sulindac is far from an ideal choice of tocolytic agent.
  • new agents or regimens capable of reducing and/or preventing an inflammatory response in the reproductive system of a female are highly 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.
  • prostaglandins can be used to delay the onset and/or prevent the continuation of labour in a female.
  • the present invention provides the 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.
  • this is achieved by preventing and/or reducing an inflaimnatory response in the reproductive system of a female.
  • the invention stems from the unexpected finding that the cyclopentenone prostaglandins, such as 15-deoxy- ⁇ 12 ' 14 prostaglandin J 2 (15-dPGJ 2 ) and prostaglandin Aj (PGAi), inhibit and/or reduce NFicB activity within uterine cells of the female reproductive system.
  • cyclopentenone prostaglandins provide a means for the inliibition and/or reduction of NFicB activity in the reproductive system of a female.
  • Medicaments of the invention are believed to inhibit cytokine synthesis and inhibit the 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 ⁇ , ⁇ -unsaturated carbon3 and is formed by dehydration of the cyclopentane ring of a precursor prostaglandin.
  • 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 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 PGE 2 , PGD 2 and PGF 2 ⁇ synthase, respectively.
  • the cyclopentenone prostaglandins, prostaglandin A (PGA 2 ), prostaglandin Ai (PGA and prostaglandin J 2 (PGJ 2 ) are formed by dehydration of prostaglandin E 2 (PGE ), prostaglandin E x (PGE ⁇ and prostaglandin D 2 (PGD 2 ), respectively.
  • PGE prostaglandin E 2
  • PGE ⁇ prostaglandin E x
  • PGD 2 prostaglandin D 2
  • PGJ 2 is metabolised further to ⁇ 12 -prostaglanding J 2 ( ⁇ 12 -PGJ 2 ), and 15-deoxy- ⁇ 12 ' 14 prostaglandin J 2 (15-dPGJ 2 ).
  • cyclopentenone prostaglandins 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 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.
  • cyclopentenone prostaglandin we include any natural, unnatural or chemically-modified prostaglandin which has a cyclopentenone ring. Cyclopentenone prostaglandin is often abbreviated to "cyPG".
  • cyclopentenone prostaglandins include prostaglandin D 2 (PGD 2 ) and its metabolite 15-deoxy- ⁇ 12;1 prostaglandin (15-dPGJ 2 ). Also preferred is prostaglandin Ai (PGAi).
  • 15-dPGJ 2 may be obtained from Cayman Chemical, 1180 East Ellsworth Road, Aim Harbour, MI 48108 USA (catalogue number 18570); 9,10-di- hydro-15-deoxy- ⁇ 12:14 -Prostaglandin J 2 may be obtained from Alexis Biochemicals Ltd, PO Box 6757, Bingham, Nottingham, NG13 8LS, UK (catalogue number CAY-18590-M001).
  • PGAi may be obtained from Alexis Biochemicals Ltd (address as above; catalogue number 340-045- M005).
  • onset of labour and/or “continuation of labour” we mclude the biochemical and/or physiological changes associated with preparation of the tissues of the female reproductive system for delivery.
  • 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 indicates the degree of cervical ripening (described in Herman et al, 1993).
  • 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.
  • 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.
  • the present invention provides the 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.
  • Such medicaments are able to inhibit and/or reduce NFicB activity in uterine cells.
  • NFicB we include homo- and heterodimers of RelA (p65), RelB, NFicB 1 (p50), NF K B2 (p52) and cRel.
  • the RelA (p65), RelB, NFicB 1 (p50), NF ⁇ B2 (p52), and cRel genes and the sequence of the pofypeptide products are described in Li et al. (2002).
  • NFicB activity we include the activities of NFicB associated with the expression of genes controlled by any homo- or heterodimer of RelA (p65), RelB, NFicB 1 (p50), NF K B2 (p52) or cRel of the NFicB transcription factor family.
  • nuclear translocation of NFicB which can be measured, for example, by Western blottmg analysis of nuclear and cytosolic cellular fractions for the protein of interest (described in Sambrook et al., 1989; Lee et al, 2003); binding of NFicB 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 NFicB-mediated 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).
  • ESA Electro-Mobility Shift Assay
  • 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"
  • Cultures of amnion cells may be prepared from tissue by separating the entire amnion, except for the part overlying the placenta, from the chorion, followed by separating amnion epithelial cells from fibroblasts and maintaining the epithelial cells using mammalian cell culture techniques (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 maimnalian cell cultures will be well known to those skilled in the relevant art.
  • reproductive system of a female we mclude any cells and/or tissues and/or organs of a female directly or indirectly involved in the formation, nourishment, maintenance and development of a neonate, embryo or fetus at any gestational stage during pregnancy.
  • the medicament is for preventing and/or reducing an inflammatory response in the reproductive system of a female that is pregnant.
  • inflaimnatory response we include biochemical and physiological changes associated with inflammation mediated by cells of the host's immune system. Such changes are known in the arts of human and veterinary medicine, immunology, molecular biology and biological science.
  • 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.
  • the present invention provides a use wherein the female is human and the duration of pregnancy is more than approximately 13 weeks of human pregnancy. More preferably, the duration of pregnancy is approximately between 20 and 32 weeks.
  • the medicament reduces and/or prevents an inflammatory 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.
  • certain groups of pregnant females are at high risk of pre-term labour.
  • Females that have had one or more instances of pre-term labour previously are at considerably higher risk of a further pre-term 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 l ⁇ iown in the art.
  • pre-term labour it is also useful to substantially prevent for a considerable duration pre-term labour using the method of the invention.
  • the medicament reduces and/or prevents an inflammatory response in the reproductive system of a female associated with infection by a pathogenic agent
  • the pathogenic agent is viral, bacterial or fungal.
  • the medicament reduces and/or prevents an inflammatory response in the reproductive system of a female associated with stretch of the uterus.
  • stretch of the uterus we include mechanical stretching of the uterus occurring where the uterus is overdistended by multiple pregnancy or by excess amniotic fluid (clinically termed hydramnios or polyhydramnios).
  • cervical incompetence 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).
  • the medicament reduces and/or prevents one or more of the following conditions: pre-term labour; pathogenic infection; cervical ripening, uterine contractions.
  • preterm labour we include the meaning of spontaneous labour occurring before the usual calculated time for delivery.
  • preterm 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 l ⁇ iown in the arts of human and veterinary medicine.
  • the medicament reduces and/or prevents fetal or neonatal damage.
  • the medicament reduces and/or prevents one or more of the following conditions: astrogliosis; loss of myelin-producing oligodendrocytes; multifocal necroses resulting in cystic change (periventricular leucomalacia, PVL).
  • Astrogliosis we include the meaning of hypertrophy (i.e. increasing cell size) of the astroglia, that usually occurs in response to injury.
  • Astroglia are the largest and most numerous 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 form the glial (limiting) membrane and directly and indirectly contribute to the blood- brain barrier. They regulate the extracellular ionic and chemical environment, and "reactive astrocytes" (along with microglia) respond to injury. Astrocytes can release neuro-transmirters, but their role in signaling (as in many other functions) is not well understood.
  • oligodendrocytes 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.
  • multifocal necroses we include the meaning of death of tissue occurring at more than one site.
  • cystic change we include the meaning of the development of fluid filled spaces in the region where necrosis has taken place.
  • periventricular leucomalacia or “PVL” we include the meaning of damage to the periventrical cerebral white matter which is seen following cytokine induced or hypoxia/ischeamia mduced necroses and which can go on to become cystic change.
  • a particularly preferred embodiment of the invention is the use of the cyclopentenone prostaglandin 15-deoxy- ⁇ 12;14 -prostaglandin J 2 and/or prostaglandin Aj.
  • the cyclopentenone prostaglandin is provided in the form of a prodrug of 15-deoxy- ⁇ 12 ' 14 -prostaglandin J 2 and/or prostaglandin Ai.
  • prodrugs of the cyclopentenone prostaglandins particularly those of 15- deoxy- ⁇ 12 ' 1 -prostaglandin J 2 and/or prostaglandin A are included within the scope of the invention.
  • the prodrug is PGD 2 (the precursor of 15-dPG ) or PGEi (the precursor of PGAi).
  • the medicament further comprises a pharmaceutically acceptable excipient, diluent or carrier.
  • the carrier does not have a deleterious effect on the recipient.
  • the carrier will be sterile and pyrogen free.
  • the medicament is in a fo ⁇ n adapted for deliver ⁇ ' by mouth, intravenous injection or intra- amniotic injection.
  • the medicament is in a form which is compatible with the amniotic fluid. More preferably, the medicament is in a fo ⁇ n which has substantially the same pH and/or osmotic tension as amniotic fluid.
  • the amniotic fluid has a distinct pH and a distinct osmotic tension.
  • the amniotic fluid pH and osmotic tension are well l ⁇ iown to, or can be readily measured by, the person skilled in the art.
  • 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.
  • an “agent for treating a female who has or is at risk of one or more of the following conditions: pre-te ⁇ n labour; pathogenic infection; cervical ripening, uterine contractions” we include corticosteroids, tocolytic agents and anti-inflaimnatory prostaglandins.
  • the agent is a corticosteroid.
  • the agent is capable of preventing and/or reducing respiratory distress syndrome.
  • corticosteroids 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 neonatal respiratory problems
  • the agent is selected from dexamethasone or betamethasone.
  • dexamethasone or betamethasone Such agents are l ⁇ iown to those skilled in the art. Administration of such agents may be two doses of 12mg intra-muscular (IM), 12 or 24 hours apart. Preferably, the agent is capable of delaying delivery.
  • the agent capable of delaying delivery is selected from: oxytocin receptor antagonists; calcium channel blockers; sympathomimetics; nitric oxide donors
  • the agent is a tocolytic agent.
  • tocolytic we include the meaning of a drug whose action is to stop uterine contractions.
  • the tocolytic agent is selected from: oxytocin receptor antagonists, calcium channel blockers, sympathomimetics, nitric oxide donors.
  • 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 glyceryl trinitrate.
  • the inflammatory response is mediated by NFicB in uterine cells.
  • the cyclopentenone prostaglandin is capable of hibiting and/or reducing NFicB activity by preventing and/or reducing NFicB DNA- binding in uterine cells.
  • the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFicB activity by preventing and/or reducing NFicB - mediated transcriptional regulation in uterine cells. More preferably, the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFKB activity by preventing and/or reducing NFicB production in uterine cells.
  • a further aspect of the invention is to provide a pharmaceutical composition
  • 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.
  • 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.
  • 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 NFicB activity in uterine cells in a PPAR- ⁇ independent manner.
  • NFicB activity we include the DNA-binding activity of NFicB and/or NFicB-mediated transcriptional regulation.
  • Testing a cyclopentenone prostaglandin to determine if it is capable of mhibiting and/or reducing NFicB activity in uterine cells in a PPAR- ⁇ independent mamier can be perfo ⁇ ned by the methods described in Example 1, below.
  • whether a cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFKB activity hi uterine cells in a PPAR- ⁇ independent mamier can be determined by using the PPAR- ⁇ inhibitor GW-9662, as shown in Figure 6, below.
  • PPAR- ⁇ independent manner we include the meaning that the activity of a cyclopentenone prostaglandin occurs without it binding to and/or activating the PPAR- ⁇ receptor.
  • a further aspect of the present invention is to provide a method for making a pharmaceutical composition for use in deling 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 earner.
  • Electro-mobility shift assay analysis of NF-icB DNA binding h nuclear protein extracts from (A) 77?yometrial 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 bmding, and consensus Oct-1 probe used as control.
  • Amnion cells derived from L- or L+ placentas were transiently transfected with the NFKB -dependent reporter construct ⁇ cB.BG.Luc, treated with 15dPGJ 2 , PGAi, troglitazone, WY-14643, or vehicle for 2 h, and then stimulated with IL-l ⁇ (1 ng/ml) for 6 h.
  • the mutated icBmut.Luc construct was used as a control to confi ⁇ n NFicB-mediated transactivation. Values are normalised for b-gal reporter activity.
  • Myometrial cells were transiently transfected with the NFicB-dependent reporter construct icB.B G.Luc, treated with 15dPGJ 2 or vehicle for 2 h, +/- IL-l ⁇ (1 ng/ml) for 6 h.
  • Figure 10 Effect of PGA and PPAR agonists on NFKB transcriptional activity in myometrium
  • Myometrial cells were transiently transfected with the NFicB-dependent reporter construct icB.BG.Luc, treated with troglitazone, WY-14643, PGAi or vehicle for 2 h, +/- IL-lb (1 ng/ml) for 6 h.
  • Myometrial cells were cotransfected with 0.4 mg of the PPAR- ⁇ -dependent reporter construct 3-PPRE-TK.pGL3 and 100 ng, 200 ng or 300 ng of a PPAR- ⁇ expression construct.
  • Cells were treated with 10 mM or 20 mM of (A) troglitazone or (B) GW1929, or vehicle for 24 h. Values are normalised for CMV-renilla reporter activity. Similar results were obtained with transfection of amnion cells.
  • FIG. 13 Troglitazone does not inhibit NFicB transcriptional activity in PPAR- ⁇ -transfected cells
  • FIG 14 PPAR- ⁇ overexpression does not potentiate 15d-PGJ 2 inhibition of NFkB activity
  • Figure 15 15dPGJ 2 inhibition of p65 nuclear localisation, p50 phosphorylation, and IicB a degi-adation
  • Figure 16 PGA] inhibition of p65 nuclear localisation and IicBa degradation
  • Figure 20 Effect ofl5dPGJ 2 and PPAR agonists on IL-l ⁇ -induced COX-2 protein expression
  • FIG. 21 Schematic of the structure of (A) prostaglandin A j (PGA]) and (B) 15-deoxy- ⁇ " prostaglandin J 2 (15-dPGJ 2 )
  • Figure 24 The cyclopentenone ring is essential for cyPG inhibition of NF ⁇
  • NF- ⁇ cB-DNA binding was measured by EMSA in nuclear protem extracts from myometrial cells pre-treated with vehicle, 15d-PGJ 2 or PGAi 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.
  • Myometrial cells were transiently transfected with a NF-icB-LUC reporter and a ⁇ -gal reporter plasmid, pre-treated with vehicle or PGAi for 2 h, and stimulated with IL-l ⁇ (1 ng/ml) for 6 h. Luciferase activity was normalized for ⁇ -gal reporter readout.
  • EDTA Ethylenediaminetetraacetic acid
  • EGTA Ethyleneglycol bis-aminoethyltetra acetic acid
  • NP-40 Nonidet P-40 SDS-PAGE Sodium dodec3'l sulphate - Polyacrylamide gel electrophoresis PVDF Pobyvhtylidene 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
  • M3'ometrial tissue was collected at tenn from the upper margin of uterine 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 & Riverside Hospital.
  • Myometrial tissue was dissected, rinsed in PBS, and digested in serum-free DMEM containing 15mg/ml collagenase 1A (Sigma), 15mg/ml collagenase X, and 50mg/ml 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 were used between passage numbers 1-4.
  • Placentae were obtained from patients at term either at elective Caesarean section prior to labour (L-) or following spontaneous labour onset and vaginal delivery (L+).
  • Amnion 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 dispase in serum-free DMEM for 35min at 37°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 lOmin and cultured in DMEM, 10% FCS (Sigma), 1% L-glutamine, 1% penicillin-streptomycin.
  • Nuclear and cytosolic protein extracts were obtained from cultured amnion cells as described by Schreiber et al (1989). For nuclear/cytosolic fractionation, confluent cell monolayers were scraped and lysed using a buffer containing lOmM HEPES, lOmM KC1, O.lmM EDTA, O. lmM 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 lOmin and NP-40 added to a final concentration of 1% (v/v).
  • Lysates were vortexed for lOsecs and centrifuged for 30secs at 4°C, 12000g.
  • the supematants were retained as the cytosolic protein extracts.
  • the pellets were resuspended in buffer containing lOmM HEPES, lOmM KC1, O.lmM EDTA, O.lmM 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 5min centrifugation at 4°C, 12000g.
  • Tissue samples were rinsed in ice-cold PBS, dissected, flattened between aluminium foil, flash-frozen in liquid nitrogen, and stored at -80°C. 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, ImM EDTA, 0.5mM PMSF and any unbroken tissue was removed by centrifugation for 30sec at 2000rpm at 0°C.
  • the supernatant was incubated on ice for 5min, centrifuged for lOmin at 4000rpm at 0°C, and the nuclear pellets resuspended in 25% (v/v) glycerol 20mM HEPES, 0.42M NaCl, 1.2 mM MgCl 2 , 0.2mM EDTA, 0.5mM DTT, and CPIs.
  • Sense and antisense strands (175nmole/ml each) were incubated in annealing buffer (lOmM Tris-HCl pH7.5, lOOmM NaCl, ImM EDTA) for lOmin at 65°C, and allowed to cool at room temperature for 2h.
  • annealing buffer lOmM Tris-HCl pH7.5, lOOmM NaCl, ImM EDTA
  • 3-5 ⁇ g protein extracts were incubated on ice for lh with non-radiolabelled non-specific oligonucleotide (poly(dl-dC) or Oct-1) in a binding buffer (20% (v/V) glycerol, 5mM MgCl 2 , 2mM EDTA, 50mM Tris-HCl pH7.5, 250mM NaCl, 2mM DTT), followed by a 45min incubation with 0.035pmole 2 P( ⁇ ATP)-end labelled oligonucleotide probes:
  • RNA/DNA complexes were separated in a 4% acrylamide gel, the gel dried under vacuum at 80°C and exposed to X- ray film.
  • samples were incubated with 2 ⁇ g 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 SOI 6 7NS, United Kingdom.
  • Protein samples (20-70 ⁇ g) were mixed with Laemmli sample buffer (l .T) containing ⁇ -mercaptoethanol (5%), and boiled for 5min. Proteins were then separated by SDS-PAGE (12-14% gels) and transfe ⁇ ed onto PVDF membrane (Amersham Pharmacia 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 (lOmin each) in PBS-T with vigorous 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 (lOmin each) in PBS-T.
  • HRP-conjugated secondary antibody diluted 1:2000 in 1% non-fat milk in PBS-T buffer
  • washed three times (lOmin each) in PBS-T washed three times (lOmin each) in PBS-T.
  • Signal detection was achieved using enhanced chemi-luminescence (ECL plus system, Amersham Pharmacia Biotech) according to manufacturer's instructions.
  • blots were incubated for 30min in 50°C stripping buffer (2% SDS, 62.5mM Tris-HCl pH6.7, lOOrnM 2-mercaptoethanol), washed 2x in PBS-T, placed in blotto overnight, and then probed with a new antibody as above.
  • 50°C stripping buffer 2% SDS, 62.5mM Tris-HCl pH6.7, lOOrnM 2-mercaptoethanol
  • luciferase reporter construct was transfected using a 1 :1 ratio of transfection (i.e., 3 ⁇ l Transfast per I ⁇ g DNA) in serum-free DMEM for lh.
  • DMEM, 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 ceUs treated with various agonists/inhibitors or vehicle for 6-8h.
  • Transfections were analysed in a dual firefly/renilla (Packard BioSciences/Calbiochem) luciferase assay or f ⁇ refiy/ ⁇ -galactosidase (Promega/Galacton) assay using a luminometer.
  • a dual firefly/renilla Packard BioSciences/Calbiochem
  • f ⁇ refiy/ ⁇ -galactosidase Promega/Galacton
  • pGL3.6 ⁇ B.BG.luc was the reporter construct used to assess NF- ⁇ B- mediated transcription, while the mutant pGL3.6 ⁇ Bmut.luc and empty ⁇ GL3.BG.luc were used as controls (Schwarzer et al, 1998).
  • pGL3.6 ⁇ B.BG.luc a NF-icB -dependent reporter construct with 6 copies of the F-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'. which contains three copies of the decameric NF- ⁇ B binding site (underlined) upstream of a minimal ⁇ -globin promoter driving a luciferase gene.
  • pGL3.6 ⁇ Bmut.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).
  • pGL3.BG.luc this reporter construct contains only the minimal ⁇ -globin promoter.
  • Cells were co-transfected with the renilla vector pRL-CMV or a ⁇ - galactosidase vector pCHHO as internal controls for transfection efficiencies.
  • pSG5/p65 expression construct was 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 transfo ⁇ ned JM109 E. coli cells, and all constructs were subsequently precipitated with polyethylene glycol.
  • Recombinant cytoldne IL-l ⁇ and TNF ⁇ from R&D Systems 15d-PGJ 2 , PGAi, troglitazone, GW-9662, and 16,16-dimethyl-PGE 2 from Cayman Chemical; WY-14643, MG132 proteasome inliibitor, and PG490 (triptolide) from Calbiochem; HRP-conjugated secondary antibodies and antibodies to p50, p65, c-rel, Rel B, COX-2, IicB ⁇ , IicB ⁇ , and PPAR ⁇ from Santa Cruz; antibodies to p52, Bcl-3 and smooth muscle actin from Upstate Biotechnologies. Antibody to PPAR- ⁇ from Affinity BioReagents, to phospho-p65 from Cell Signalmg, to COX-1 from Alexis Biochemicals, and to lamin B from Oncogene Research Products. Mouse model of preterm labour
  • 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%o paraformaldehyde for 24h and then stored in 70% ethanol until further processing. Placentae were homogenized for 1 minute in the presence of lysis buffer comprising 400mM KC1, 20mM HEPES pH7.4, ImM dithiothreitol, 20% glycerol and 5% (v/v) protease inhibitor cocktail.
  • lysis buffer comprising 400mM KC1, 20mM HEPES pH7.4, ImM dithiothreitol, 20% glycerol and 5% (v/v) protease inhibitor cocktail.
  • IL-l ⁇ Interlekin-l ⁇
  • TNF ⁇ tumour necrosis factor ⁇
  • CyPGs but not PPAR agonists, inhibit NF- ⁇ B DNA binding in amnion and myometi'ial cells.
  • 15d-PGJ 2 inhibited ILl- ⁇ -induced NF- ⁇ B 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-1 or Sp-1 probe was unaffected by either IL-l ⁇ or 15d-PGJ 2 treatment, conf ⁇ ning that the effects observed are NF-KB -specific.
  • PPAR- ⁇ is the putative endogenous receptor for 15d-PGJ 2 , and PPAR expression may be affected by cytokines (Tontonoz et al, 1998, Tanaka et al, 1999), PPAR- ⁇ protein expression was examined in myometrial and amnion cells. PPAR- ⁇ was shown to be expressed predominantly in the nucleus of both cell types, and its expression was not affected by IL-l ⁇ or 15d-PGJ 2 treatment (Fig. 2). 15d-PGJ 2 can also transactivate PPAR- ⁇ , though more weakly than PPAR- ⁇ (Forman et al, 1995). PPAR- ⁇ expression in myometrial and amnion cells was found to be predominantly cytoplasmic (Fig. 3).
  • troglitazone had no effect on NF-icB DNA binding at 10-50 ⁇ M doses, although it did cause a slight reduction at lOO ⁇ M (Fig. 4, 5).
  • Troglitazone can transactivate PPAR- ⁇ at I ⁇ M and induces weak interactions between PPAR- ⁇ and the co-activators p300 and steroid receptor co-activator (SRC-1) at lO ⁇ M doses; adipogenesis is positively regulated by PPAR- ⁇ , and troglitazone can induce expression of adipogenic markers at 5-lO ⁇ M doses (Prusty et al., 2002).
  • NF- ⁇ B DNA binding was assessed in cells treated with 15d-PGJ 2 in the presence of the selective PPAR- ⁇ inhibitor GW-9662.
  • GW9662 binds irreversibly to PPAR- ⁇ through covalent modification of Cys " in the ligand-bmd ng domain (Leesnitzer et al., 2002).
  • GW-9662 failed to alleviate 15d-PGJ 2 inliibition of NF-icB (Fig. 6).
  • PGA b which does not act as a PPAR ligand but does contain a cyclopentenone ring, was able to inhibit NF-icB DNA binding in amnion and myometrial cells, albeit at much higher doses than 15d-PGJ 2 (Fig. 7).
  • CyPGs but riot PPAR agonists, inhibit NF- ⁇ B transcriptional activity.
  • amnion cells were transfected with the NF-icB-dependent reporter icB.BG.Luc and treated with 15d-PGJ 2 , PGAi, troglitazone, WY-14643 or vehicle, followed by IL-1 ⁇ stimulation (Fig. 8).
  • Constitutive reporter activity was seen in both L- and L+ amnion cells, although the levels were lower and showed a greater increase with IL- I ⁇ . in L- cells, in agreement with previous studies by Allport et al (2001). Both 15d-PGJ 2 and PGA ⁇ inhibited IL-l ⁇ -induced NF-icB transcriptional activity, whereas troglitazone and WY-14643 did not.
  • IL-l ⁇ -induced NF-icB transcriptional activity in myometrial cells, 15d-PGJ 2 ihibited IL-l ⁇ -induced NF-icB transcriptional activity in a dose-dependent manner, reducing reporter activity to basal levels (Fig. 9). IL-l ⁇ -induced NF-icB transcriptional activity was also reduced to basal levels by PGA], but not troglitazone, GW1929 or WY-14643 (Fig. 10, 11).
  • GW1929 and troglitazone were shown to be functional as PPAR- ⁇ ligands, potentiating PPAR- ⁇ -mediated transcription of a PPRE-dependent reporter in both cell types. Endogenous PPAR- ⁇ 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 PPAR ⁇ -transfected cells, and PPAR ⁇ overexpression did not promote 15d-PGJ 2 inhibition of NF-icB transcriptional activity (Fig. 12, 13, 14).
  • CyPGs, but notPGE 2 inhibit NF-icB activation and IicB degradation.
  • 16,16-Dimeth ⁇ -PGE2 a PGE 2 analogue with increased half-life, did not inhibit NF- ⁇ B DNA binding (controlled for with Oct-1 binding) or IL-l ⁇ - induced p65 nuclear translocation in myometrial and amnion cells (Fig. 17).
  • PGE 2 is l ⁇ iown to be pro-inflammatory, does not contain a cyclopentenone ring, and does not activate PPAR- ⁇ (Forman et al, 1995).
  • 16,16-dimeth3 -PGE 2 did not inhibit NF-icB DNA binding or p65 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-l ⁇ or TNF ⁇ .
  • TNF ⁇ and IL-l ⁇ were significantly higher in the placentae proximal to the injection site compared to those in the opposite horn.
  • Levels of IL-l ⁇ were approximately 40% lower in proximal placentae injected with LPS + 15d-PGJ 2 compared to those given LPS + vehicle (Fig. 22). This difference was statistically significant (p ⁇ 0.05).
  • TNF ⁇ levels were not significantly altered according to drug treatment.
  • the cyclopentenone ring is essential for cyPG inhibition ofNF-icB
  • 15d-PG is a PPAR agonist
  • other prostaglandins such as PGAi
  • PGAi 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 PPAR ⁇ agonist activity but in which the cyclopentenone ring has been disrupted) could not reproduce the effects of 15d-PGJ 2 (Fig. 24).
  • 15d-PGJ 2 inhibited ILl- ⁇ -induced NF-icB DNA binding and NF-icB- mediated transactivation in myometrial cells, as well as in L- and L+ amnion cells. 15d-PGJ 2 inhibited the nuclear translocation and activation of NF-icB, at least in part, by preventing the degradation of IicB ⁇ by IL-l ⁇ .
  • PPAR- ⁇ agonists did not impair TNF ⁇ -mduced NF-icB activation, nuclear translocation, or DNA binding activity; rather, they antagonised the transcriptional regulatory activity of NF-icB, and PPAR- ⁇ overexpression was required to demonstrate such inhibition (Ruan et al., 2003).
  • PPAR- ⁇ overexpression potentiated transactivation of a PPRE, it did not enable the PPAR- ⁇ agonists to inhibit NF-icB transcription.
  • 15d-PGJ 2 was able to inhibit NF- ⁇ B transcription in the absence of exogenous PPAR- ⁇ and overexpression of this receptor did not promote inhibition.
  • IL-l ⁇ -induced COX-2 expression was inhibited by 15d-PGJ 2 but not by PPAR agonists. While PPAR agonists are l ⁇ iown to be anti-inflammatory 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 cell types (Meade et al, 1999; Ikawa et al, 2001; Pang et al, 2003).
  • CyPGs such as 15d-PGJ 2 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 glutathione and cysteine residues in cellular proteins. Receptor-independent actions of 15d-PGJ 2 have been attributed to its cyclopentenone ring.
  • NF-icB proteins contain a conserved cysteine residue in their DNA-binding domain (DBD) and alkylation of this cysteine impairs DNA binding (Toledano et al, 1993).
  • PGAi a cyPG that does not act as a PPAR- ⁇ ligand, was able to inhibit NF-icB DNA binding and transactivation, albeit at higher concentrations than 15d-PGJ 2 .
  • This ability of PGAi, but not PGE 2 or PPAR agonists, to mimic the effects of 15d-PGJ 2 suggests that these cyPGs may inhibit NF- ⁇ B in amnion and myometrial cells by virtue of their cyclopentenone ring.
  • CyPG administration offers an attractive alternative approach to anti-inflammatory treatment since a potential specificity of cyPGs for IKK ⁇ /IicB ⁇ would spare other potentially beneficial pathways of NF-icB activation (e.g., the processing of pi 05 and fonnation of p50 homodimers), which might be disrupted by more broad- spectrum NF-icB inhibitors.
  • the use of the cyPGs, able to simultaneously 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 preterm labour and neurodevelopmental disorders of the neonate.
  • mice model used is an effective model for the study of prete ⁇ n delivery and agents that may delay the onset of pretenn delivery.
  • the finding of lower levels of IL-1 ⁇ and phospho-p65 in mice treated with the cyclopentenone prostaglandin 15d-PGJ 2 suggests that this compound is effective at blocking the inflammatory pathway induced by LPS treatment in vivo.
  • EXAMPLE 2 Preferred pharmaceutical formulations and modes and doses of administration.
  • the compounds of the present invention may be delivered using an 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.
  • the compounds of the present invention can be administered by a surgically implanted device that releases the drug directfy to the required site.
  • a surgically implanted device that releases the drug directfy to the required site.
  • Vitrasert releases ganciclovir directly into the eye to treat CMV retinitis.
  • the direct application of this toxic agent to the site of disease achieves effective therapy without the drug's significant systemic side- effects.
  • Electroporation therapy (EPT) systems can also be employed for administration.
  • EPT Electroporation therapy
  • a device which delivers a pulsed electric field to cells increases the penneability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drug delivery.
  • Compounds can also be delivered by electroincorporation (El).
  • El occurs when small particles of up to 30 microns in diameter on the surface of the skin experience electrical pulses identical or similar to those used in electroporation. In El, 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.
  • An alternative method of administration is the ReGel injectable system that is thermos ens itive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into l ⁇ iown, safe, biodegradable polymers. The active 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 ⁇ 2 in the body to co- deliver proteins and peptides.
  • the protein or peptide can move through the intestinal wall.
  • Complexes are synthesised between vitamin B ⁇ 2 analogues and the drug that retain both significant affinity for intrinsic factor (IF) in the vitamin 12 portion of the complex and significant bioactivity of the drug portion of the complex.
  • 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 efficient (Derossi et al, 1998).
  • the pharmaceutical fo ⁇ nulation 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 mgredient.
  • the compounds of the invention can be administered orally or by any parenteral route, in the fo ⁇ n of a pharmaceutical fo ⁇ nulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage fo ⁇ n.
  • a pharmaceutical fo ⁇ nulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage fo ⁇ n.
  • the compositions may be administered at varying doses.
  • Formulations in accordance with the present invention suitable for oral administration ma3' 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, 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 fo ⁇ n such as a powder or granules, optional ⁇ mixed with a binder (e.g. povidone, gelatin, h3 ⁇ droxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g. sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • a binder e.g. povidone, gelatin, h3 ⁇ droxypropylmethyl cellulose
  • lubricant e.g. sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose
  • preservative e.g. sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose
  • disintegrant
  • Moulded tablets may be made by moulding in a suitable machme a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be fo ⁇ nulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydrox3'piOpylmethylcellulose h varying proportions to provide desired release profile.
  • 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 mouth-washes comprising the active ingredient in a suitable liquid carrier.
  • the compounds of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient diluent or can ⁇ er selected with regard to the intended route of administration and standard pharmaceutical practice.
  • 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 immediate-, delayed- or controlled-release applications.
  • the compounds of the invention may also be administered via intracavemosal injection.
  • Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glyc ne, disintegrants such as starch (preferably com, 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 magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glyc ne
  • disintegrants such as starch (preferably com, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and
  • Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • Preferred excipients in this regard include lactose, starch, cellulose, milk sugar or high molecular weight polyethylene glycols.
  • 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.
  • the compounds of the invention can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventriculai y, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be admhiistered by infusion techniques.
  • parenterally for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventriculai y, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be admhiistered by infusion techniques.
  • 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 standard
  • Fo ⁇ nulations 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 of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requ ⁇ ing only the addition of the sterile liquid carrier, 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 land previously described.
  • oral or parenteral administration of the compounds of the invention is the prefe ⁇ ed route, being the most convenient.
  • 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 route of administration which will be most appropriate for a particular animal.
  • the fo ⁇ nulations of the pharmaceutical compositions of the invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include 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 mgredient with liquid carriers or finely divided solid earners or both, and then, if necessary, shaping the product.
  • Prefe ⁇ ed unit dosage fo ⁇ nulations are those containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of an active mgredient.
  • a preferred delivery system of the invention may comprise a hydrogel hnpregnated with a compound of the invention, which is preferably earned 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.
  • a compound of the invention Whilst it is possible for a compound of the invention to be administered alone, it is preferable to present it as a pharmaceutical fo ⁇ nulation, together with one 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.
  • the carriers will be water or saline which will be sterile and pyrogen-free.
  • Example 3B Ophthalmic Solution
  • Active ingredient 0.5 g Sodium chloride, analytical grade 0.9 g Thiomersal 0.001
  • Purified water 100 ml pH adjusted to 7.5
  • fo ⁇ nulations A and B are prepared by wet granulation of the ingredients with a solution of povidone, followed by addition of magnesium stearate and compression.
  • Formulation C mg/tablet Active ingredient 100 Lactose 200 Starch 50
  • the following fo ⁇ nulations, D and E are prepared by direct compression of the admixed ingredients.
  • the lactose used in formulation E is of the direction compression type.
  • the fo ⁇ nulation is prepared by wet granulation of the ingredients (below) with a solution of povidone followed by the addition of magnesium stearate and compression.
  • Active Ingredient 500 Hydroxypropylmethylcellulose 112 (Methocel K4M Premium) ® Lactose B.P. 53 Povidone B.P.C. 28 Magnesium Stearate 7 700
  • Drug release takes place over a period of about 6-8 hours and was complete after 12 hours.
  • a capsule formulation is prepared by admixing the ingredients of Fo ⁇ nulation D in Example C above and filling into a two-part hard gelatin capsule.
  • Formulation B (infra) is prepared in a similar manner.
  • Formulation C mg/capsule Active ingredient 250 Macrogol 4000 BP 350 600
  • 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.
  • Capsules are prepared by dispersing the active ingredient in the lecithm and arachis oil and filling the dispersion into soft, elastic gelatin capsules.
  • the following controlled release capsule fo ⁇ nulation is prepared by extruding ingredients a, b, and c using an extruder, followed by spheronisation of the extradate and drymg. The dried pellets are then coated with release- controlling membrane (d) and filled into a two-piece, hard gelatin capsule.
  • mg/capsule Active ingredient 250 Microci stalline Cellulose 125 Lactose BP 125 Ethyl Cellulose 13
  • Active ingredient 0.200 g Sterile, pyrogen free phosphate buffer (pH7.0) to 10 ml
  • the active ingredient is dissolved in most of the phosphate buffer (35-40°C), then made up to volume and filtered through a sterile micropore filter mto a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals.
  • the sodium benzoate is dissolved in a portion of the purified water and the sorbitol solution added.
  • the active ingredient is added and dispersed.
  • the glycerol is dispersed the thickener (dispersible cellulose). The two dispersions are mixed and made up to the required volume with the purified water. Further thickening is achieved as required by extra shearing of the suspension.
  • Example 3H Suppository mg/suppository Active ingredient (63 ⁇ m)* 250 Hard Fat, BP (Witepsol HI 5 - Dynamit Nobel) 1770
  • the active ingredient is used as a powder wherein at least 90% of the particles are of 63 ⁇ m diameter or less.
  • Witepsol HI 5 is melted hi a steam-jacketed pan at 45 °C maximum.
  • the active mgredient is sifted through a 200 ⁇ m sieve and added to the molten base with mixing, usmg a silverson fitted with a cutting head, until a smooth dispersion is achieved. Maintaining the mixture at 45 °C, the remaining Witepsol HI 5 is added to the suspension and stirred to ensure a homogenous mix.
  • the entire suspension is passed through a 250 ⁇ m stainless steel screen and, with continuous stirring, is allowed to cool to 40 °C. At a temperature of 38°C to 40 ° C 2.02 g of the mixture is filled into suitable plastic moulds. The suppositories are allowed to cool to room temperature.
  • Example 31 Pessaries mg/pessary Active ingredient 250 Anhydrate Dextrose 380 Potato Starch 30J Magnesium Stearate 7
  • Example 3J Creams and ointments
  • the compounds of the invention may also be delivered using microsphere fo ⁇ nulations, such as those described in Cleland (1997; 2001).
  • the compounds of the invention may be delivered by inhalation, with the aid of a dry powder inhaler delivering micronised particles in metered quantities as described in Ansel (1999).
  • Example 3M Aerosol Inhalation
  • the compounds of the invention may be delivered by inhalation, with the aid of a suitable inhaler delivering micronised paiticles in metered quantities employing a non CFC propellant as described in Ansel (1999).
  • 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.
  • Skinner KA and Challis JR Changes in the synthesis and metabolism of prostaglandins by human fetal membranes and decidua at labour.
  • 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.
  • Staels B Koenig W, Habib A, Merval R, Lebret M, To ⁇ a IP, Delerive P, Fadel A, Chinetti G, Fmchart JC, Najib J, Maclouf J, Tedgui A.

Abstract

The present invention provides the 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. Preferably the cyclopentenone prostaglandin prevents and/or reduces an inflammatory response in the reproductive system of a female. Preferably, the cyclopentenone prostaglandin is 15-deoxy-Δ12,14-prostaglandin J2 or prostaglandin A1, or a precursor thereof. The invention further provides a pharmaceutical composition comprising cyclopentenone prostaglandin and methods of use thereof.

Description

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 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 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.
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 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 labour process. Interleukin- lβ (L -lβ) 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 and blood and in fetal membranes in association with labour (Satoh et al, 1919; Skinner and Challis, 1985). PGE2 stimulates uterine contractions (Dyal and Crankshaw, 1985), indirectly increases fundamentally dominant myometrial contractility by upregulation of oxytocin receptors and synchronisation of contractions (Garfield et al, 1990), and acts in concert with LL-8 to remodel the cervix (reviewed in Kelly, 2002).
The onset of labour is associated with activation of the Nuclear Factor Kappa B (NFKB) transcription factor system in the amnion which plays a role in the expression of pro-inflammatory genes such as interleukin- 8 (LL- 8), interleukin-6 (IL-6) and cyclo-oxygenases 1 and 2 (COX-1 and COX-2). 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.
Five members of the NF-κB/Rel family have been identified in mammals: NF-κBl (p50 and its precursor pl05), NF-κB2 (p52 and its precursor plOO), p65 (RelA), c-rel, and Rel B. These proteins share a structurally conserved amino-terminal region termed the Rel homology domain (RHD). The RHD is responsible for dimerisation, DNA binding, and interaction with the inhibitors of kappa B (Ii B) proteins. It also contains a nuclear localisation signal (NLS). In its active DNA-binding form NF-κB consists of heterogeneous dimers of various combinations of NF-κB subunits: each member of the NF-i B family, except for Rel B, can form homodimers, as well as heterodimers with one another. The p65, c-rel and Rel B proteins contain a carboxy-teπninal non-homologous transactivation domain, which activates transcription from i B sites in target genes; in contrast, p50 and p52 proteins lack a transactivation domain. The various NF-icB dimers exhibit different binding affinities for specific KB sites (Kunsch et al, 1992, Phelps et al, 2000), and differentially stimulate transcription through distinct icB elements (Lin et al, 1995).
In resting cells, NF-icB dimers are normally sequestered in an inactive form in the cytoplasm by association with the inhibitory I B proteins, which include IκBα, Ii Bβ and I Bε. The Ii Bs are characterised by the presence of multiple ankyrin repeats which mediate binding to the RHD and mask the NLS of NF- B. The major NF-icB signalling pathway, which is activated by pro- inflammatory stimuli and LPS, targets IicBα- and IicBβ-bound NF-κB (for review see Li and Verma 2002). p50/p65 dimers are the most abundant form of NF-icB in most cell types, and activation of IicBα-bound p50/p65 dimers is the best characterised pathway. In this 'classical' pathway, diverse stimuli trigger signal transduction cascades that ultimately converge on the activation of a specific IκB kinase (IKK). The LKK complex consists of several proteins, the main ones being IKKα (IKK1), LKKβ (LKK2), and NF- B essential modulator (NEMO or IKKγ). The activated IKK complex phosphorylates IicBα at serines 32 and 36, which results in the poly- ubiquitination of IκBα at lysines 21 and 22. This modification targets IicBα for rapid degradation by the 26S proteasome. The degradation of the IicB inhibitor exposes the NLS of NF-icB resulting in translocation of the p50/p65 dimer to the nucleus where it can bind to cB sites in the promoter of target genes and promote transcription.
Most stimuli cause only the transient activation of NF-icB. The critical inhibitory step in NF-icB inactivation involves binding of newly synthesised IκBα to NF-icB in the nucleus. IicBα is quickly res3mthesised following its degradation. The newly synthesised IicBα is localised in the nucleus and displaces NFicB from its DNA binding sites. IicBα contains leucine-rich nuclear export sequences (NES) (Johnson et al 1999), which then enable it to transport NF-icB back to the cytoplasm, thereby completing an autoregulatory post-induction repression. In many cells nearly half of the NF-icB is sequestered by the other major IKB isoform, IicBβ (Whiteside et al, 1991). In contrast to IicBα, IicBβ is not NF-icB inducible and does not exert a rapid post-induction repression of NF-icB activity. Rather, IicBβ has been implicated in persistent NF-icB activation. Prolonged exposure to certain stimuli, such as LPS, leads to the long-term induction of NF-icB activity despite high levels of newly synthesised IicBα. Following stimulus-induced degradation, the newly synthesised IκBβ is un-phosphorylated and, in contrast to IicBα or the constitutively phosphorylated IκBβ, can interact with NF-icB bound to target promoters without displacing it from the DNA (Suyang et al, 1996). This interaction of un-phosphorylated IκBβ with DNA-bound NF-icB is thought to protect NF-icB from nuclear export, and thus inhibition, by IicBα, and the outcome is a sustained NF-KB response.
PGs are a family of biologically active molecules having a diverse range of 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 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 prostaglandin dehydrogenases are thought to protect the uterus from basal 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). 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). 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- activated transcription factors belonging to the nuclear receptor superfamily. They exist in three distinct forms, PPAR-α, PPAR-δ, and PPAR-γ, which form heterodimers with the retinoic X receptor (RXR) and bind to PPAR response elements (PPREs) in the promoter of target genes to induce transcription. PPAR-γ can also repress gene transcription by negatively interfering with the NF-icB, AP-1, STAT and C/EBP pathways (Zhou et al, 1999; Subbaramaiah et al, 2001; Takata et al, 2002; Suzawa et al, 2003).
The aetiology of pre-teπn labour is multi-factorial but bacterial infection is 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 bom before 32 weeks of gestation. During ascending intrauterine infection, micro-organisms can stimulate the production of pro- inflammatory cytokines, such as tumour necrosis factor α (TNFα) and IL- lβ, as well as PGs and other inflaimnatoiy mediators, resulting in the 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 with prematurity (Darnmann et al, 1999).
In addition, inflammatory responses caused by mechanical stretching of the uterus may contribute to the onset of labour. Mechanical stretchmg of the uterus occurs to an extent as a normal part of pregnancy and may be responsible for some of the biochemical changes which occur near to term and which cause the normal 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 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). 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 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 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-term birth also cause fetal cerebral damage. Characteristically, damage is localised to the white matter, involving both a diffuse astrogliosis with subsequent loss of myelin-producing oligodendrocytes, as well as 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). There are currently no drugs available which will safely and effectively inhibit pre-term contractions. The most commonly used agents, β-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. 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 β-sympathomimetics is only in the temporary delay of labour to allow in utero transfer or administration of steroid to improve fetal lung surfactant production.
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 corticosteroids improves the outcome for the pre-term 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 h surfactant production and therefore prevents or reduces the severity of neonatal respiratory problems. Such agents are lαiown to those skilled hi the art.
Current obstetric management of pre-term labour (or threatened pre-term labour or pre-term premature rupture of membranes) is to attempt to delay delivery using 'tocolytic' drugs to allow time for steroid administration. Typically, effective tocolytic drugs are ox}rtocin receptor antagonists, calcium channel blockers, sympathomimetics and nitric oxide donors.
A commonly used oxytocin receptor antagonist is Atosiban, that functions by blocking the oxytocin receptor, thereby preventing activation of the receptor by endogenous oxytocin that stimulates uterine contractions. A commonly 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 commonly used 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 the activity of myosin light chain Idnase, 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 prolongation of pregnancy than the β-sympathomimetics and, unlike β-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 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 (< 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). Because of these side effects some obstetricians now use Sulindac, which appears to be equally good as a tocolytic (Carlon et al 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 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 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 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 delaying the onset and/or preventing the continuation of labour in a female.
Preferably, this is achieved by preventing and/or reducing an inflaimnatory response in the reproductive system of a female.
The invention stems from the unexpected finding that the cyclopentenone prostaglandins, such as 15-deoxy-Δ12'14prostaglandin J2 (15-dPGJ2) and prostaglandin Aj (PGAi), inhibit and/or reduce NFicB activity within uterine cells of the female reproductive system. Thus, cyclopentenone prostaglandins provide a means for the inliibition and/or reduction of NFicB activity in the reproductive system of a female. Medicaments of the invention are believed to inhibit cytokine synthesis and inhibit the 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 α,β -unsaturated carbon3 and is formed by dehydration of the cyclopentane ring of a 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 A2. Arachidonic acid is then converted sequentially to PGG2 and PGH2 by the cyclo-oxygenase and peroxidase activities of the PGH synthases, PGH 1 and 2. The prostaglandins PGE2, PGD2 and PGF are subsequently synthesised from PGH2 via the action of the PGE2, PGD2 and PGF synthase, respectively. The cyclopentenone prostaglandins, prostaglandin A (PGA2), prostaglandin Ai (PGA and prostaglandin J2 (PGJ2) are formed by dehydration of prostaglandin E2 (PGE ), prostaglandin Ex (PGE^ and prostaglandin D2 (PGD2), respectively. PGJ2 is metabolised further to Δ12-prostaglanding J212-PGJ2), and 15-deoxy-Δ12'14prostaglandin J2 (15-dPGJ2).
Other umiatural 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 which are added to the desired chemical head-group. This S3mthesis is versatile and allows the synthesis of a variety of natural and umiatural 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 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. 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 D2 (PGD2) and its metabolite 15-deoxy-Δ12;1 prostaglandin (15-dPGJ2). Also preferred is prostaglandin Ai (PGAi).
15-dPGJ2 may be obtained from Cayman Chemical, 1180 East Ellsworth Road, Aim Harbour, MI 48108 USA (catalogue number 18570); 9,10-di- hydro-15-deoxy-Δ12:14-Prostaglandin J2 may be obtained from Alexis Biochemicals Ltd, PO Box 6757, Bingham, Nottingham, NG13 8LS, UK (catalogue number CAY-18590-M001). PGAi may be obtained from Alexis Biochemicals Ltd (address as above; catalogue number 340-045- M005).
By "onset of labour" and/or "continuation of labour" we mclude the biochemical and/or physiological changes associated with preparation of the 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 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. 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.
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 inflammatory response in the reproductive system of a female. Such medicaments are able to inhibit and/or reduce NFicB activity in uterine cells.
By "NFicB" we include homo- and heterodimers of RelA (p65), RelB, NFicB 1 (p50), NFKB2 (p52) and cRel. The RelA (p65), RelB, NFicB 1 (p50), NFκB2 (p52), and cRel genes and the sequence of the pofypeptide products are described in Li et al. (2002).
By "NFicB activity" we include the activities of NFicB associated with the expression of genes controlled by any homo- or heterodimer of RelA (p65), RelB, NFicB 1 (p50), NFKB2 (p52) or cRel of the NFicB transcription factor family. In particular, we include: nuclear translocation of NFicB which can be measured, for example, by Western blottmg analysis of nuclear and cytosolic cellular fractions for the protein of interest (described in Sambrook et al., 1989; Lee et al, 2003); binding of NFicB 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 NFicB-mediated 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 NFicB are well known by those skilled in the art of biochemistry and molecular biology. 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"
Cultures of amnion cells may be prepared from tissue by separating the entire amnion, except for the part overlying the placenta, from the chorion, followed by separating amnion epithelial cells from fibroblasts and maintaining the epithelial cells using mammalian cell culture techniques (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 maimnalian cell cultures will be well known to those skilled in the relevant art.
By "reproductive system of a female", we mclude any cells and/or tissues and/or organs of a female directly or indirectly involved in the formation, nourishment, maintenance and development of a neonate, embryo or fetus at any gestational stage during pregnancy. In particular we include the cells and/or tissues of the uterus, placenta, amnion, chorion, decidua, cervix and vagina.
Preferably, the medicament is for preventing and/or reducing an inflammatory response in the reproductive system of a female that is pregnant.
By "inflaimnatory response" we include biochemical and physiological changes associated with inflammation mediated by cells of the host's 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, 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 mflammation within the uterus. Other clinical measures of inflammation within the uterus are maternal temperature, white blood cell 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 lαiown to those skilled in the art.
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 human and the duration of pregnancy is more than approximately 13 weeks 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 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. 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-term labour. Females that have had one or more instances of pre-term labour previously are at considerably higher risk of a further pre-term 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 lαiown in the art.
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 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 labour using the method of the invention. In particular, it is useful to inhibit 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 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. 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 occurring where the uterus is overdistended by multiple pregnancy or by excess amniotic fluid (clinically termed 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).
Preferably, the medicament reduces and/or prevents one or more of the following conditions: pre-term labour; pathogenic infection; cervical ripening, uterine contractions.
By "pre-term labour", we include the meaning of spontaneous labour occurring before the usual calculated time for delivery. In humans, preterm 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 lαiown in the arts of human and veterinary medicine. Preferably, the medicament reduces and/or prevents fetal or neonatal damage.
More preferably, the medicament reduces and/or prevents one or more of 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 cell size) of the astroglia, that usually occurs in response to injury. Astroglia are the largest and most numerous 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 form the glial (limiting) membrane and directly and indirectly contribute to the blood- brain barrier. They regulate the extracellular ionic and chemical environment, and "reactive astrocytes" (along with microglia) respond to injury. Astrocytes can release neuro-transmirters, but their role in signaling (as in many other functions) is not well understood.
By "oligodendrocytes" 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.
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 which is seen following cytokine induced or hypoxia/ischeamia mduced necroses and which can go on to become cystic change. A particularly preferred embodiment of the invention is the use of the cyclopentenone prostaglandin 15-deoxy-Δ12;14-prostaglandin J2 and/or prostaglandin Aj.
Alternatively, the cyclopentenone prostaglandin is provided in the form of a prodrug of 15-deoxy-Δ12'14-prostaglandin J2 and/or prostaglandin Ai.
It will be appreciated by those skilled in the art that certain metabolic precursors of cyclopentenone prostaglandins, may not possess pharmacological 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".
All prodrugs of the cyclopentenone prostaglandins, particularly those of 15- deoxy-Δ12'1 -prostaglandin J2 and/or prostaglandin A are included within the scope of the invention.
Preferably, the prodrug is PGD2 (the precursor of 15-dPG ) or PGEi (the precursor of PGAi).
Preferably, the medicament further comprises a pharmaceutically acceptable excipient, diluent or carrier.
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.
Preferably the medicament is in a foπn adapted for deliver}' by mouth, intravenous injection or intra- amniotic injection. Preferably, the medicament is in a form which is compatible with the amniotic fluid. More preferably, the medicament is in a foπn which has substantially the same pH and/or osmotic tension as amniotic fluid.
The amniotic fluid has a distinct pH and a distinct osmotic tension. The amniotic fluid pH and osmotic tension are well lαiown to, or can be readily measured by, the person skilled in the art. 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 following conditions: pre-teπn labour; pathogenic infection; cervical ripening, uterine contractions" we include corticosteroids, tocolytic agents and anti-inflaimnatory prostaglandins.
Preferably, the agent is a corticosteroid.
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 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 lαiown to those skilled in the art. Administration of such agents may be two doses of 12mg intra-muscular (IM), 12 or 24 hours apart. Preferably, the agent is capable of delaying delivery.
More preferably, the agent capable of delaying delivery is selected from: oxytocin receptor antagonists; calcium channel blockers; sympathomimetics; nitric oxide donors
Preferably, the agent is a tocolytic agent. 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 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 glyceryl trinitrate.
Preferably, the inflammatory response is mediated by NFicB in uterine cells.
More preferably, the cyclopentenone prostaglandin is capable of hibiting and/or reducing NFicB activity by preventing and/or reducing NFicB DNA- binding in uterine cells.
More preferably, the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFicB activity by preventing and/or reducing NFicB - mediated transcriptional regulation in uterine cells. More preferably, the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFKB activity by preventing and/or reducing NFicB production in uterine cells.
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.
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. 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 NFicB activity in uterine cells in a PPAR-γ independent manner.
By "NFicB activity" we include the DNA-binding activity of NFicB and/or NFicB-mediated transcriptional regulation.
Testing a cyclopentenone prostaglandin to determine if it is capable of mhibiting and/or reducing NFicB activity in uterine cells in a PPAR-γ independent mamier can be perfoπned by the methods described in Example 1, below. For example, whether a cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFKB activity hi uterine cells in a PPAR-γ independent mamier can be determined by using the PPAR-γ inhibitor GW-9662, as shown in Figure 6, below. By "PPAR-γ independent manner" we include the meaning that the activity of a cyclopentenone prostaglandin occurs without it binding to and/or activating the PPAR-γ receptor.
It will be understood that a cyclopentenone prostaglandin to determine if it is capable of inhibiting and/or reducing NFicB activity in uterine cells may be tested in vitro, in vivo or ex vivo. A further aspect of the present invention is to provide a method for making a pharmaceutical composition for use in deling 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 earner.
Prefeπed, non-limiting examples which embody certain aspects of the invention will now be described, with reference to the following figures:
Figure 1 : 15dPGJ2 inhibition ofNF-κB DNA bindint σ
Electro-mobility shift assay (EMSA) analysis of NF-icB DNA binding h nuclear protein extracts from (A) 77?yometrial cells, (B) L+ amnion cells, and (C) L- amnion cells treated with 15dPGJ2 or vehicle for 2 h +/- IL-lb stimulation (15 min). Consensus kB probe used to assess NF-kB DNA bmding, and consensus Oct-1 probe used as control.
Figure 2 : PPAR-γ protein expression
Western immunoblots of (A) nuclear and cytosolic protein extracts from myometrial and amnion cells with or without 15 min IL-lβ stimulation, and (B) nuclear extracts of myometrial cells treated with 15d-PGJ2 +/- IL-lb. Probing with antibody to PPARγ.
Figure 3 : PPAR-aprotein expression
Western immunoblots of nuclear and cytosolic protein extracts from myometrial and amnion cells with or without 15 min IL-lb stimulation. Probing with antibody to PPARα. Figure 4 : PPAR-γ 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, consensus Oct-1 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 NFKB DNA binding
EMSA analysis of nuclear protein extracts from myometrial cells treated with (A) WY-14643 or vehicle, and (B) high doses of troglitazone, WY- 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-1 probes used as controls.
Figure 6 : PPAR-γ antagonist GW9662 does not alleviate 15dPGJ2 inhibition ofNF B DNA binding
EMSA analysis of nuclear protem extracts from amnion cells treated with 15dPGJ2 +/- GW9662 or vehicle for 2 h followed by IL- 1 β stimulation ( 15 min). Consensus kB probe used to assess NFKB DNA binding, consensus Oct-1 probe used as control.
Figure 7 : PGA] inhibition of NFKB DNA binding
EMSA analysis of nuclear protein extracts from (A) myometrial cells, and (B) amnion cells treated with PGAi or vehicle for 2 h followed by IL-lb stimulation (15 min). Consensus icB probe used to assess NFKB DNA binding, consensus Oct-1 probe used as control. For supershift analysis, extracts were preincubated with antibodies against p50 or p65.
Figure 8 : Effect of cyPGs and PPAR agonists on NFicB transcriptional activity in amnion
Amnion cells derived from L- or L+ placentas were transiently transfected with the NFKB -dependent reporter construct ιcB.BG.Luc, treated with 15dPGJ2, PGAi, troglitazone, WY-14643, or vehicle for 2 h, and then stimulated with IL-lβ (1 ng/ml) for 6 h. The mutated icBmut.Luc construct was used as a control to confiπn NFicB-mediated transactivation. Values are normalised for b-gal reporter activity.
Figure 9 : 15dPGJ2 inhibition of NFicB transcriptional activity in myometrium
Myometrial cells were transiently transfected with the NFicB-dependent reporter construct icB.B G.Luc, treated with 15dPGJ2 or vehicle for 2 h, +/- IL-l β (1 ng/ml) for 6 h. The mutated icBmut.Luc construct was used as a control to confirm NFicB -mediated transactivation. Values are nonnalised for b-gal reporter activity. (NS = nonstimulated). Figure 10 : Effect of PGA and PPAR agonists on NFKB transcriptional activity in myometrium
Myometrial cells were transiently transfected with the NFicB-dependent reporter construct icB.BG.Luc, treated with troglitazone, WY-14643, PGAi or vehicle for 2 h, +/- IL-lb (1 ng/ml) for 6 h. The mutated icBmut.Luc construct was used as a control to confmn NFicB-mediated transactivation. Values are normalised for CMV-Renilla reporter activity. (NS = nonstimulated).
Figure 11 : PPAR-γ agonist GW1929 does not inhibit NFicB transcriptional activity
Myometrial cells were transiently transfected with the ΝFicB-dependent reporter construct icB.BG.Luc, treated with GW1929 or vehicle for 2 h, +/- IL-lb (1 ng/ml) for 6 h. Values are nonnalised for CMV-Renilla reporter activity. (NS = nonstimulated).
Figure 12 : Troglitazone and GW 1929 potentiate PPAR-γ activation of a PPRE reporter
Myometrial cells were cotransfected with 0.4 mg of the PPAR-γ-dependent reporter construct 3-PPRE-TK.pGL3 and 100 ng, 200 ng or 300 ng of a PPAR-γ expression construct. Cells were treated with 10 mM or 20 mM of (A) troglitazone or (B) GW1929, or vehicle for 24 h. Values are normalised for CMV-renilla reporter activity. Similar results were obtained with transfection of amnion cells.
Figure 13 : Troglitazone does not inhibit NFicB transcriptional activity in PPAR-γ-transfected cells Myometrial cells were transfected with 0.4 mg icB.BG.Luc reporter and 200 ng PPAR-γ 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 CMV-renilla reporter activity. (NS = nonstimulated).
Figure 14 : PPAR-γ overexpression does not potentiate 15d-PGJ2 inhibition of NFkB activity Myometrial cells were transfected with 0.4 mg icB.BG.Luc reporter and 200 ng PPAR-γ expression vector and treated with 15d-PGJ2 for 2h followed by IL-l β (1 ng/ml) for 6 h. Values are noπnalised for β- galactosidase reporter activity. (NS = nonstimulated).
Figure 15 : 15dPGJ2 inhibition of p65 nuclear localisation, p50 phosphorylation, and IicB a degi-adation
Western immunoblots of nuclear or cytosolic protein extracts from (A) myometrial cells, (B) L- amnion cells, and (C) L+ amnion cells treated with 15dPGJ2 or vehicle for 2 h +/- IL-lβ stimulation (15 min). Blots probed with antibodies agamst p65, p50 or IicBα.
Figure 16 : PGA] inhibition of p65 nuclear localisation and IicBa degradation
Western immunoblot of nuclear or c3r osolic protein extracts from myometrial cells treated with PGAi or vehicle for 2h followed by IL-lβ stimulation (15 min). Blots probed with antibodies aga st p65or IicB a. Figure 17 : PGE2 does not inhibit TNFa- and IL-lβ-induced NFKB activation
Analysis of nuclear protein extracts from myometrial cells treated with PGE2 or vehicle for 2 h +/- TNFα or IL-lb stimulation (15 min). A) EMSA using consensus icB probe. (B) Western immunoblot probing for nuclear p65.
Figure 18 : PGE2 does not induce NFicB DNA binding
EMSA analysis of nuclear protein extracts from (A) L- amnion cells and (B) myometrial cells treated with vehicle, PGE2 or IL-lβ. Consensus icB probe used. Figure 19 : 15dPGJ inhibits IKB a phosphorylation
Western immunoblots of cytosolic extracts from myometrial cells (A) treated with 15dPGJ2 for 2h +/- TLlb for 15min; probed for IKK, and (B) treated with 30mM 15dPGJ2, 40mM MG132 or vehicle for 2h, +/- IL-lβ for 15min; probed for IicBα.
Figure 20 : Effect ofl5dPGJ2 and PPAR agonists on IL-lβ-induced COX-2 protein expression
Western immunoblot of cytosolic protein extracts from myometrial cells treated with 15dPGJ2, troglitazone, WY-14643 or vehicle for 2h, followed by IL-lβ stimulation for 6h. Probed with antibodies to (A) COX-2, and (B) a smooth muscle actin. Figure 21 : Schematic of the structure of (A) prostaglandin A j (PGA]) and (B) 15-deoxy-Δ " prostaglandin J2 (15-dPGJ2)
Figure 22 : Effect of LPS and 15d-PGJ2 on inflammatory responses - IL-lβ levels
Concentrations of IL-1 β in placental homogenates collected from gestation day 16 mice 6 hours after intrauterine injection of 250μg LPS + vehicle or 250μg LPS + 4μg 15d-PGJ2. * denotes statistically significant difference (t- test (p<0.05)).
Figure 23 : Effect of LPS and 15d-PGJ2 on inflammatory responses — phospho-p65 levels
Relative concentrations of phospho-p65 in placental homogenates collected from gestation day 16 mice 6 hours after intrauterine injection of 250μg LPS + vehicle or 250μg LPS + 4μg 15d-PGJ2. * denotes statistically significant difference (t-test (p<0.05)).
Figure 24 : The cyclopentenone ring is essential for cyPG inhibition of NF¬
(A) NF-ιcB-DNA binding was measured by EMSA in nuclear protem extracts from myometrial cells pre-treated with vehicle, 15d-PGJ2 or PGAi for 2 h, followed by stimulation with IL-lβ (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-icB-LUC reporter and a β-gal reporter plasmid, pre-treated with vehicle or PGAi for 2 h, and stimulated with IL-lβ (1 ng/ml) for 6 h. Luciferase activity was normalized for β-gal reporter readout. Values are presented as the mean +/- SEM obtained for each treatment done in triplicate. Western blot analysis of nuclear p65 and p50 expression in myometrial cells treated with (C) PGAi or (D) 15d-PGJ2 for 2 h, followed by stimulation with IL-lβ (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 stimulation with IL-lβ (1 ng/ml) for 15 min. Membranes were probed with antibodies against p65 and Ser 536-phosphor} ated p65. Similar results were obtained in amnion epithelial cells.
EXAMPLE 1 - Experimental data
Methods
Abbreviations
EDTA Ethylenediaminetetraacetic acid EGTA Ethyleneglycol bis-aminoethyltetra acetic acid DTT Dithiotreitol HEPES 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid NP-40 Nonidet P-40 SDS-PAGE Sodium dodec3'l sulphate - Polyacrylamide gel electrophoresis PVDF Pobyvhtylidene 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
Tissue biopsies and cell culture
Local Ethics committee approval was obtained for the collection of these tissues and patients gave infoπned consent.
Human myometrial cell culture
M3'ometrial tissue was collected at tenn from the upper margin of uterine 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 serum-free DMEM containing 15mg/ml collagenase 1A (Sigma), 15mg/ml collagenase X, and 50mg/ml 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 were used between passage numbers 1-4.
Human Amnion Cell Culture
Placentae were obtained from patients at term either at elective Caesarean section prior to labour (L-) or following spontaneous labour onset and vaginal delivery (L+). Amnion 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 dispase in serum-free DMEM for 35min at 37°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 lOmin and cultured in DMEM, 10% FCS (Sigma), 1% L-glutamine, 1% penicillin-streptomycin.
Protein Extracts from cultured cells
Nuclear and cytosolic protein extracts were obtained from cultured amnion cells as described by Schreiber et al (1989). For nuclear/cytosolic fractionation, confluent cell monolayers were scraped and lysed using a buffer containing lOmM HEPES, lOmM KC1, O.lmM EDTA, O. lmM 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 lOmin and NP-40 added to a final concentration of 1% (v/v). Lysates were vortexed for lOsecs and centrifuged for 30secs at 4°C, 12000g. The supematants were retained as the cytosolic protein extracts. The pellets were resuspended in buffer containing lOmM HEPES, lOmM KC1, O.lmM EDTA, O.lmM 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 5min centrifugation at 4°C, 12000g.
For whole cell lysates, confluent cell monolayers were scraped and lysed in a high-salt extraction buffer containing 0.4M KC1, 20mM HEPES, 20% (v/v) glycerol, ImM DTT, and CPIs.
Protem Extracts fiΛomfi"esh 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. 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, ImM EDTA, 0.5mM PMSF and any unbroken tissue was removed by centrifugation for 30sec at 2000rpm at 0°C. The supernatant was incubated on ice for 5min, centrifuged for lOmin at 4000rpm at 0°C, and the nuclear pellets resuspended in 25% (v/v) glycerol 20mM HEPES, 0.42M NaCl, 1.2 mM MgCl2, 0.2mM EDTA, 0.5mM DTT, and CPIs.
All extracts were aliquoted, frozen on dry ice and stored at -80°C. The extracts were processed for protein quantitation by the Lowry method usmg Bio-Rad protein assay reagents (Bio-Rad Laboratories) according to manufacturer's instructions.
Electro-mobility shift assay (EMSA)
Oligonucleotide labelling
Sense and antisense strands (175nmole/ml each) were incubated in annealing buffer (lOmM Tris-HCl pH7.5, lOOmM NaCl, ImM EDTA) for lOmin at 65°C, and allowed to cool at room temperature for 2h. 3.5 pmole double-stranded oligonucleotides were end-labelled with 0.37MBq o2P(γATP) by incubating for 30min at 37°C with T4 polynucleotide kinase. Labelled oligonucleotides were recovered by centrifugation at SOOOrpm for 2min through MicroSpin G-25 or G-50 sephadex columns (Amersham Biosciences).
EMSA
3-5μg protein extracts were incubated on ice for lh with non-radiolabelled non-specific oligonucleotide (poly(dl-dC) or Oct-1) in a binding buffer (20% (v/V) glycerol, 5mM MgCl2, 2mM EDTA, 50mM Tris-HCl pH7.5, 250mM NaCl, 2mM DTT), followed by a 45min incubation with 0.035pmole 2P(γATP)-end labelled oligonucleotide probes:
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 icB : 5 '-CGG GAG AGG GGA TTC CCT GCG C-3 ' downstream COX-2 KB : 5 ' -AGA GTG GGG ACT ACC CCC TCT-3 ' Oct-1 or SP-1 consensus sequences were used as a controls for a NF-icB- specific effect. The resulting protein/DNA complexes were separated in a 4% acrylamide gel, the gel dried under vacuum at 80°C and exposed to X- ray film. For supershift analysis, samples were incubated with 2μg 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 SOI 6 7NS, United Kingdom.
SDS-PAGE and Western blotting analysis
Protein samples (20-70μg) were mixed with Laemmli sample buffer (l .T) containing β-mercaptoethanol (5%), and boiled for 5min. Proteins were then separated by SDS-PAGE (12-14% gels) and transfeπed onto PVDF membrane (Amersham Pharmacia 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 (lOmin each) in PBS-T with vigorous 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 (lOmin each) in PBS-T. Signal detection was achieved using enhanced chemi-luminescence (ECL plus system, Amersham Pharmacia Biotech) according to manufacturer's instructions.
To re-probe a membrane, blots were incubated for 30min in 50°C stripping buffer (2% SDS, 62.5mM Tris-HCl pH6.7, lOOrnM 2-mercaptoethanol), washed 2x in PBS-T, placed in blotto overnight, and then probed with a new antibody as above. 30-50μg protein extracts were subjected to SDS-PAGE and Western immuno-blotting. Secondary antibodies were IgG-HRP and ECL Plus detection kit (Amersham Pharmacia Biotech, Amersham 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 (Promega). 0.5 μg per well of luciferase reporter construct was transfected using a 1 :1 ratio of transfection (i.e., 3μl Transfast per Iμg DNA) in serum-free DMEM for lh. DMEM, 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 ceUs treated with various agonists/inhibitors or vehicle for 6-8h. Transfections were analysed in a dual firefly/renilla (Packard BioSciences/Calbiochem) luciferase assay or fϊrefiy/β-galactosidase (Promega/Galacton) assay using a luminometer.
pGL3.6κB.BG.luc was the reporter construct used to assess NF-κB- mediated transcription, while the mutant pGL3.6κBmut.luc and empty ρGL3.BG.luc were used as controls (Schwarzer et al, 1998).
pGL3.6κB.BG.luc: a NF-icB -dependent reporter construct with 6 copies of the F-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'. which contains three copies of the decameric NF-κB binding site (underlined) upstream of a minimal β-globin promoter driving a luciferase gene. pGL3.6κBmut.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). pGL3.BG.luc: this reporter construct contains only the minimal β-globin promoter.
Cells were co-transfected with the renilla vector pRL-CMV or a β- galactosidase vector pCHHO as internal controls for transfection efficiencies.
In vitro translation and plasmid preps
For recombinant production of p65, a pSG5/p65 expression construct was 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 transfoπned JM109 E. coli cells, and all constructs were subsequently precipitated with polyethylene glycol.
Reagents/Antibodies
Recombinant cytoldne IL-lβ and TNFα from R&D Systems; 15d-PGJ2, PGAi, troglitazone, GW-9662, and 16,16-dimethyl-PGE2 from Cayman Chemical; WY-14643, MG132 proteasome inliibitor, and PG490 (triptolide) from Calbiochem; HRP-conjugated secondary antibodies and antibodies to p50, p65, c-rel, Rel B, COX-2, IicBα, IicBβ, and PPARγ from Santa Cruz; antibodies to p52, Bcl-3 and smooth muscle actin from Upstate Biotechnologies. Antibody to PPAR-γ from Affinity BioReagents, to phospho-p65 from Cell Signalmg, to COX-1 from Alexis Biochemicals, and to lamin B from Oncogene Research Products. Mouse model of preterm labour
Surgery was performed on timed pregnant MF1 mice at day 16 of gestation. After deep maternal anaesthesia was attained, a minilaparotomy was perfoπned in the lower abdomen. The uterine horns were exposed through the incision and preterm labour was induced by the intrauterine injection of 250μg lipopolysaccharide (LPS, Sigma) into the gravid horn. This was immediately followed by injection of 4μg 5d-PGJ2 (Cayman), or an equal 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 on inflammatory responses
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%o paraformaldehyde for 24h and then stored in 70% ethanol until further processing. Placentae were homogenized for 1 minute in the presence of lysis buffer comprising 400mM KC1, 20mM HEPES pH7.4, ImM dithiothreitol, 20% glycerol and 5% (v/v) protease inhibitor cocktail.
Homogenate levels of Interlekin-l β (IL-lβ) and tumour necrosis factor α (TNFα) 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-l β and TNFα levels were expressed as pg/mg total protein. Homogenates were also subjected to polyacrylamide gel electrophoresis. Loading volumes were adjusted according to the protein content of each homogenate such that a constant amount of protein was run in each lane. Phosphorylated p65 (phospho-p65) was detected by western hnmunoblotting using a specific antibod3 (Santa Cruz) and quantified by densitometric analysis.
Results
CyPGs, but not PPAR agonists, inhibit NF-κB DNA binding in amnion and myometi'ial cells.
15d-PGJ2 inhibited ILl-β-induced NF-κB 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-1 or Sp-1 probe was unaffected by either IL-lβ or 15d-PGJ2 treatment, confϊπning that the effects observed are NF-KB -specific.
Since PPAR-γ is the putative endogenous receptor for 15d-PGJ2, and PPAR expression may be affected by cytokines (Tontonoz et al, 1998, Tanaka et al, 1999), PPAR-γ protein expression was examined in myometrial and amnion cells. PPAR-γ was shown to be expressed predominantly in the nucleus of both cell types, and its expression was not affected by IL-l β or 15d-PGJ2 treatment (Fig. 2). 15d-PGJ2 can also transactivate PPAR-α, though more weakly than PPAR-γ (Forman et al, 1995). PPAR-α expression in myometrial and amnion cells was found to be predominantly cytoplasmic (Fig. 3).
The ability of synthetic PPAR agonists to mimic the inhibitory effects of
15d-PGJ2 was examined. The PPAR-γ agonist troglitazone had no effect on NF-icB DNA binding at 10-50μM doses, although it did cause a slight reduction at lOOμM (Fig. 4, 5). Troglitazone can transactivate PPAR-γ at IμM and induces weak interactions between PPAR-γ and the co-activators p300 and steroid receptor co-activator (SRC-1) at lOμM doses; adipogenesis is positively regulated by PPAR-γ, and troglitazone can induce expression of adipogenic markers at 5-lOμM doses (Prusty et al., 2002). Thus, at lOOμM concentrations, it is unlilcely that troglitazone is exerting a specific effect through PPAR-γ. Since structurally distinct PPAR ligands may differentially affect coactivator/corepressor recruitment, a new potent PPAR-γ agonist, which lacks the TZD moiety, was also used. This GW1929 ligand failed to inhibit NF-icB DNA binding (Fig. 6). The synthetic PPAR-α agonist WY-14643 can transactivate PPARα at 5-25μM doses in a GAL4 chimera transfection system (Kehrer et al, 2001), but WY-14643 had no - effect on NF-icB DNA binding, at 10-100μM concentrations. To further investigate a potential role for PPAR-γ in mediating the inhibitory effects of 15d-PGJ2, NF-κB DNA binding was assessed in cells treated with 15d-PGJ2 in the presence of the selective PPAR-γ inhibitor GW-9662. GW9662 binds irreversibly to PPAR-γ through covalent modification of Cys" in the ligand-bmd ng domain (Leesnitzer et al., 2002). GW-9662 failed to alleviate 15d-PGJ2 inliibition of NF-icB (Fig. 6).
In contrast, PGAb which does not act as a PPAR ligand but does contain a cyclopentenone ring, was able to inhibit NF-icB DNA binding in amnion and myometrial cells, albeit at much higher doses than 15d-PGJ2 (Fig. 7).
CyPGs, but riot PPAR agonists, inhibit NF-κB transcriptional activity.
To determine whether the cyPG effects on NF-icB 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-PGJ2, PGAi, troglitazone, WY-14643 or vehicle, followed by IL-1 β stimulation (Fig. 8). Constitutive reporter activity was seen in both L- and L+ amnion cells, although the levels were lower and showed a greater increase with IL- Iβ. in L- cells, in agreement with previous studies by Allport et al (2001). Both 15d-PGJ2 and PGAΪ inhibited IL-l β-induced NF-icB transcriptional activity, whereas troglitazone and WY-14643 did not.
In myometrial cells, 15d-PGJ2 ihibited IL-lβ-induced NF-icB transcriptional activity in a dose-dependent manner, reducing reporter activity to basal levels (Fig. 9). IL-l β-induced NF-icB transcriptional activity was also reduced to basal levels by PGA], but not troglitazone, GW1929 or WY-14643 (Fig. 10, 11).
GW1929 and troglitazone were shown to be functional as PPAR-γ ligands, potentiating PPAR-γ-mediated transcription of a PPRE-dependent reporter in both cell types. Endogenous PPAR-γ 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 PPARγ-transfected cells, and PPARγ overexpression did not promote 15d-PGJ2 inhibition of NF-icB transcriptional activity (Fig. 12, 13, 14).
CyPGs, but notPGE2, inhibit NF-icB activation and IicB degradation.
15d-PGJ2 inhibited IL-lβ-induced p65 nuclear translocation and p50 phosphorylation in myometrial cells and in L-, L+ amnion cells in a dose- dependent manner (Fig. 15). This was paralleled by inhibition of IL-lβ- induced IicBα and IicBβ degradation. Similarly. PGAi inhibited p65 nuclear translocation and IκB (^degradation in myometrial cells (Fig. 16). 16,16-Dimeth} -PGE2, a PGE2 analogue with increased half-life, did not inhibit NF-κB DNA binding (controlled for with Oct-1 binding) or IL-lβ- induced p65 nuclear translocation in myometrial and amnion cells (Fig. 17). This is not unexpected, since, in contrast to the cyPGs, PGE2 is lαiown to be pro-inflammatory, does not contain a cyclopentenone ring, and does not activate PPAR-γ (Forman et al, 1995). 16,16-dimeth3 -PGE2 did not inhibit NF-icB DNA binding or p65 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-lβor TNFα.
Effect of 15-dPGJ? on NF- B upsti-eam activators and downsti- earn targets.
In contrast to the proteasome inhibitor MG132, which prevented IL-l β- induced IicBα degradation and resulted in the accumulation of undegraded, phosphorylated IicBα, accumulation of pb.0sph0r3d.ated IicBα was not detected following 15-dPGJ2 treatment, suggesting that 15-dPGJ2 may be affecting IKKs or other upstream kinases (Fig. 19). Both IL-l β and 15- dPGJ2 treatment had no effect on IKKα or IKKβ protein expression, although it is more likely that 15d-PGJ2 would inhibit the kinase activity of the IKKs.
Since COX-2 is an important target gene for NF-icB in labour, the effect of 15-dPGJ2 and PPAR agonists on COX-2 expression was assessed. IL-lβ- induced COX-2, expression was inhibited by 15-dPG , but not by troglitazone or WY-14643 (Fig. 20). Similar results were obtained in L- and L+ amnion cells.
Effect of LPS on pre-term delivery. Pre-term 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 15d-PGJ2 on inflammatory responses
In all mice, levels of TNFα and IL-l β were significantly higher in the placentae proximal to the injection site compared to those in the opposite horn. Levels of IL-l β were approximately 40% lower in proximal placentae injected with LPS + 15d-PGJ2 compared to those given LPS + vehicle (Fig. 22). This difference was statistically significant (p<0.05). In contrast, TNFα levels were not significantly altered according to drug treatment.
Significantly, placental levels of IL-lβ were not altered according the proximity of the placenta to the site of injection, indicating that 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-PGJ2 compared to those given LPS + vehicle (Fig. 23) and this difference was statistically significant (p<0.05).
The cyclopentenone ring is essential for cyPG inhibition ofNF-icB
Several studies have demonstrated that 15d-PG is a PPAR agonist, whilst other prostaglandins, such as PGAi, are not (Foπnan et al, 1995; Kliewer et al, 1995; Ferry et al, 2001). We have shown that PGAi shares the effect of 15d-PGJ2 on NF-icB, but that 9,10-dihydro-15d-PGJ2 (an analogue of 15d- PGJ2 which retains PPARγ agonist activity but in which the cyclopentenone ring has been disrupted) could not reproduce the effects of 15d-PGJ2 (Fig. 24). Taken together, these findings indicate that the inhibitory effects of 15d-PGJ2 on NF-icB in amnion epithelial and myometrial cells can be attributed to its electrophilic ring, that similar effects would be expected with other cyclopentenone prostaglandins but not with other, non- cyclop entenone PPAR agonists. Conclusions
NF-icB inhibition by cyPGs
15d-PGJ2 inhibited ILl-β-induced NF-icB DNA binding and NF-icB- mediated transactivation in myometrial cells, as well as in L- and L+ amnion cells. 15d-PGJ2 inhibited the nuclear translocation and activation of NF-icB, at least in part, by preventing the degradation of IicBα by IL-lβ.
In myometrial and amnion cells, which expressed both PPAR-α and PPAR- γ receptors, neither PPAR-γ nor PPAR-α agonists were able to inhibit IL- 1 β-induced NF-κB DNA binding or NF-icB transcriptional activity at doses shown to inhibit NF-κB 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-γ and NF-κB in adipocytes, PPAR-γ agonists did not impair TNFα-mduced NF-icB activation, nuclear translocation, or DNA binding activity; rather, they antagonised the transcriptional regulatory activity of NF-icB, and PPAR-γ overexpression was required to demonstrate such inhibition (Ruan et al., 2003). In the present study, while PPAR-γ overexpression potentiated transactivation of a PPRE, it did not enable the PPAR-γ agonists to inhibit NF-icB transcription. In addition, 15d-PGJ2 was able to inhibit NF-κB transcription in the absence of exogenous PPAR-γ and overexpression of this receptor did not promote inhibition. IL-lβ-induced COX-2 expression was inhibited by 15d-PGJ2 but not by PPAR agonists. While PPAR agonists are lαiown to be anti-inflammatory 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 cell types (Meade et al, 1999; Ikawa et al, 2001; Pang et al, 2003).
CyPGs such as 15d-PGJ2 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 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 domain (DBD) and alkylation of this cysteine impairs DNA binding (Toledano et al, 1993). In the present study, PGAi, a cyPG that does not act as a PPAR- γ ligand, was able to inhibit NF-icB DNA binding and transactivation, albeit at higher concentrations than 15d-PGJ2. This ability of PGAi, but not PGE2 or PPAR agonists, to mimic the effects of 15d-PGJ2 suggests that these cyPGs may inhibit NF-κB in amnion and myometrial cells by virtue of their cyclopentenone ring. Indeed, our results indicate that the inhibitory effects of 15d-PGJ2 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- c3'clopentenone PPAR agonists.
While direct modification of NF-icB cysteines has not been addressed in this study, both 15d-PGJ2- and PGApinediated inliibition of NF-icB was shown to involve the inliibition of IicBα degradation, suggesting that events further upstream in the NF-κB cascade are being targeted. 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 airay of pro-inflammatory and labour- associated genes, in these tissues. CyPG administration offers an attractive alternative approach to anti-inflammatory treatment since a potential specificity of cyPGs for IKKβ/IicBα would spare other potentially beneficial pathways of NF-icB activation (e.g., the processing of pi 05 and fonnation of p50 homodimers), which might be disrupted by more broad- spectrum NF-icB inhibitors. The use of the cyPGs, able to simultaneously 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 preterm labour and neurodevelopmental disorders of the neonate.
This study provides evidence that the mouse model used is an effective model for the study of preteπn delivery and agents that may delay the onset of pretenn delivery. The finding of lower levels of IL-1 β and phospho-p65 in mice treated with the cyclopentenone prostaglandin 15d-PGJ2 suggests that this compound is effective at blocking the inflammatory pathway induced by LPS treatment in vivo.
EXAMPLE 2 - Preferred pharmaceutical formulations and modes and doses of administration.
The compounds of the present invention may be delivered using an 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.
The compounds of the present invention can be administered by a surgically implanted device that releases the drug directfy 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 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 increases the penneability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drug delivery.
Compounds can also be delivered by electroincorporation (El). El occurs when small particles of up to 30 microns in diameter on the surface of the skin experience electrical pulses identical or similar to those used in electroporation. In El, 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. An alternative method of administration is the ReGel injectable system that is thermos ens itive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into lαiown, safe, biodegradable polymers. The active 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 Bι2 in the body to co- deliver proteins and peptides. By riding the vitamin Bι2 uptake system, the protein or peptide can move through the intestinal wall. Complexes are synthesised between vitamin Bι2 analogues and the drug that retain both significant affinity for intrinsic factor (IF) in the vitamin 12 portion of the complex and significant bioactivity of the drug portion of the complex. 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 efficient (Derossi et al, 1998).
Preferably, the pharmaceutical foπnulation 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 mgredient.
The compounds of the invention can be administered orally or by any parenteral route, in the foπn of a pharmaceutical foπnulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage foπn. Depending upon the disorder and patient to be treated, as well as the route of administration, the compositions may be administered at varying doses.
Formulations in accordance with the present invention suitable for oral administration ma3' 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, 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 foπn such as a powder or granules, optional^ mixed with a binder (e.g. povidone, gelatin, h3^droxypropylmethyl 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 machme a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be foπnulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydrox3'piOpylmethylcellulose h varying proportions to provide desired release profile.
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 mouth-washes comprising the active ingredient in a suitable liquid carrier. 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 canϊer selected with regard to the intended route of administration and standard pharmaceutical practice.
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 immediate-, delayed- or controlled-release applications. The compounds of the invention may also be administered via intracavemosal injection.
Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glyc ne, disintegrants such as starch (preferably com, 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 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, 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. The compounds of the invention can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventriculai y, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be admhiistered by infusion techniques. The 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 standard pharmaceutical techniques well-known to those skilled in the art.
Foπnulations 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 of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requπing only the addition of the sterile liquid carrier, 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 land previously described.
Generally, in humans, oral or parenteral administration of the compounds of the invention is the prefeπed 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 route of administration which will be most appropriate for a particular animal. The foπnulations of the pharmaceutical compositions of the invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include 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 mgredient with liquid carriers or finely divided solid earners or both, and then, if necessary, shaping the product.
Prefeπed unit dosage foπnulations are those containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of an active mgredient.
A preferred delivery system of the invention may comprise a hydrogel hnpregnated with a compound of the invention, which is preferably earned 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.
It should be understood that in addition to the ingredients particularly mentioned above the foπnulations of this invention may mclude 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. EXAMPLE 3 — Exemplary pharmaceutical 'formulations
Whilst it is possible for a compound of the invention to be administered alone, it is preferable to present it as a pharmaceutical foπnulation, together with one 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.
The following examples illustrate phannaceutical formulations according to the invention in which the active ingredient is a compound of the invention.
Example 3A: Tablet
Active ingredient 100 mg
Lactose 200 mg
Starch 50 mg
Polyvinylpynolidone 5 mg
Magnesium stearate 4 mg
359 mg
Tablets are prepared from the foregoing ingredients by wet granulation followed by compression. Example 3B: Ophthalmic Solution
Active ingredient 0.5 g Sodium chloride, analytical grade 0.9 g Thiomersal 0.001 Purified water to 100 ml pH adjusted to 7.5
Example 3C: Tablet Formulations
The following foπnulations A and B are prepared by wet granulation of the ingredients with a solution of povidone, followed by addition of magnesium stearate and compression.
Formulation A ms/tablet mg/tablet
(a) Active ingredient 250 250
(b) Lactose B. P. 210 26
(c) Povidone B.P. 15 9
(d) Sodium Starch Glycolate 20 12
(e) Magnesium Stearate 5 3
500 300
Formulation B mg/tablet mg/tablet
(a) Active ingredient 250 250
(b) Lactose 150 (c) Avicel PH lOl® 60 26 (d) Povidone B.P. 15 9
(e) Sodium Starch Glycolate 20 12 (f) Magnesium Stearate
500 500
Formulation C mg/tablet Active ingredient 100 Lactose 200 Starch 50
Povidone 5
Magnesium stearate 4
359
The following foπnulations, D and E, are prepared by direct compression of the admixed ingredients. The lactose used in formulation E is of the direction compression type.
Formulation D mg/capsule
Active Ingredient 250
Pregelatmised Starch NF 15 150
400 Formulation E mg/capsule
Active Ingredient 250
Lactose 150
Avicel ® 100
500 Formulation F (Conti'olled Release Formulation)
The foπnulation is prepared by wet granulation of the ingredients (below) with a solution of povidone followed by the addition of magnesium stearate and compression.
Active Ingredient 500 Hydroxypropylmethylcellulose 112 (Methocel K4M Premium)® Lactose B.P. 53 Povidone B.P.C. 28 Magnesium Stearate 7 700
Drug release takes place over a period of about 6-8 hours and was complete after 12 hours.
Example 3D: Capsule Foπnulations
Formulation A
A capsule formulation is prepared by admixing the ingredients of Foπnulation D in Example C above and filling into a two-part hard gelatin capsule. Formulation B (infra) is prepared in a similar manner.
Formulation B mg/capsule
Active ingredient 250
Lactose B.P. 143
Sodium Starch Glycolate 25 Magnesium Stearate
420
Formulation C mg/capsule Active ingredient 250 Macrogol 4000 BP 350 600
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.
Formulation D mg/capsule
Active ingredient 250
Lecithin 100
Arachis Oil 100
450
Capsules are prepared by dispersing the active ingredient in the lecithm and arachis oil and filling the dispersion into soft, elastic gelatin capsules.
Formulation E (Controlled Release Capsule)
The following controlled release capsule foπnulation is prepared by extruding ingredients a, b, and c using an extruder, followed by spheronisation of the extradate and drymg. The dried pellets are then coated with release- controlling membrane (d) and filled into a two-piece, hard gelatin capsule. mg/capsule Active ingredient 250 Microci stalline Cellulose 125 Lactose BP 125 Ethyl Cellulose 13
51.
Example 3E: Injectable Formulation
Active ingredient 0.200 g Sterile, pyrogen free phosphate buffer (pH7.0) to 10 ml
The active ingredient is dissolved in most of the phosphate buffer (35-40°C), then made up to volume and filtered through a sterile micropore filter mto a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals.
Example 3F: Intramuscular injection
Active ingredient 0.20 g
Benzyl Alcohol 0.10 g
Glucofurol 75® 1.45 g
Water for Inj ection q.s. to 3.00 ml
The active ingredient is dissolved i 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). Example 3G: Svrup Suspension
Active ingredient 0.2500 g Sorbitol Solution 1.5000 g Glycerol 2.0000 g Dispersible Cellulose 0.0750 g Sodium Benzoate 0.0050 g Flavour, Peach 17.42.3 .169 0.0125 ml Purified Water q.s. to 5.0000 ml
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. Further thickening is achieved as required by extra shearing of the suspension.
Example 3H: Suppository mg/suppository Active ingredient (63 μm)* 250 Hard Fat, BP (Witepsol HI 5 - Dynamit Nobel) 1770
2020
*The active ingredient is used as a powder wherein at least 90% of the particles are of 63 μm diameter or less.
One fifth of the Witepsol HI 5 is melted hi a steam-jacketed pan at 45 °C maximum. The active mgredient is sifted through a 200 μm sieve and added to the molten base with mixing, usmg a silverson fitted with a cutting head, until a smooth dispersion is achieved. Maintaining the mixture at 45 °C, the remaining Witepsol HI 5 is added to the suspension and stirred to ensure a homogenous mix. The entire suspension is passed through a 250 μm stainless steel screen and, with continuous stirring, is allowed to cool to 40 °C. At a temperature of 38°C to 40°C 2.02 g of the mixture is filled into suitable plastic moulds. The suppositories are allowed to cool to room temperature.
Example 31: Pessaries mg/pessary Active ingredient 250 Anhydrate Dextrose 380 Potato Starch 30J Magnesium Stearate 7
1000
The above ingredients are mixed directly and pessaries prepared by direct compression of the resulting mixture.
Example 3J: Creams and ointments
Described in Remington.
Example 3K: Microsphere foπnulations
The compounds of the invention may also be delivered using microsphere foπnulations, such as those described in Cleland (1997; 2001).
Example 3L: Dry Powder Inhalation
The compounds of the invention may be delivered by inhalation, with the aid of a dry powder inhaler delivering micronised particles in metered quantities as described in Ansel (1999). Example 3M: Aerosol Inhalation
The compounds of the invention may be delivered by inhalation, with the aid of a suitable inhaler delivering micronised paiticles in metered quantities employing a non CFC propellant as described in Ansel (1999).
References
Allport VC, Pieber D, Slater DM, Newton R, White JO, Bennett PR. Human labour is associated with nuclear factor-icB activity which mediates cyclo-ox3'genase-2 expression and is involved with the functional progesterone withdrawal, Mol Hum Reprod 2001; 7: 581-586.
Ansel. Pharmaceutical Dosage Forms and Drug Delivery Systems, 1999, Lippincott Williams and Wilkins.
Bendixen AC, Shevde NK, Dienger KM, Willson TM, Funk CD, Pike JW. E -4 inhibits osteoclast formation through a direct action on osteoclast precursors via peroxisome proliferator-activated receptor γl. Proc Natl Acad Sci USA 2001; 98: 2443-2448.
Bennett PR, Rose MP, Myatt L, Elder MG. Preterm labor: stimulation of arachidonic acid metabolism in human amnion cells by bacterial products. Am J Obstet Gynecol. 1987, 156:649-5.
Bennett, P. and Slater, D. The role of cyclo-oxygenases in the onset of labour., in improved non-steroid anti-inflaimnatory drugs: COX-2 enzyme hihibitors. J. Vane, R. Botting, and G. Bottmg, Editors. 1996, Kluwer Academic: London. P. 112-118.
Brown, NX., Alvi, S.A., Elder, M.G., Beimett, P.R. and Sullivan, M.H. A spontaneous induction of fetal membrane prostaglandin production precedes clinical labour. J Endocrinol 1998; 157: R1-R6.
Canon et al, Obstet Gynecol, 1992. 85(5), 769-774. Chinetti G, Griglio S, Antonucci M, Torra IP, Delerive P, Majd Z, Fmchart J-C, Chapman J, Najib J, Staels B. Activation of proliferator-activated receptors α and γ induces apopotosis of human monocyte-derived macrophages. JBiol Chem 1998; 273: 25573-25580.
Cleland, J.L. (1991) Pharm. Biotechnol 10:1-43.
Cleland et al. (2001) J. Control Release 72:13-24.
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 human lower uterine segment in vitro. Can J Physiol Pharmacol, 1994. 72(8): p. 870-4.
Dammann O and Leviton A. Brain damage in preterm newborns: Might enhancement of developmentally regulated endogenous protection open a 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 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 Gynecol 1988; 158: 281-285. 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.
Feπy G, Bruneau V, Beauverger P, et al. 2001 Binding of prostaglandins to human PPARgamma: tool assessment and new natural ligands. Eur J Pharmacol, 417:77-89.
Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM. 15-deoxy-Δ12"14-prostaglandin J2 is a ligand for the adipocyte determination factor PPARγ. Cell 1995; 83: 803-812.
Ganstrom et al, Acta Obstet Gynecol cand, 1987. 66:429-431. Garfϊeld RE and Hertzberg EL. Cell-to-cell coupling in the myometrium: Emil Bozler'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 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 γ suppresses nuclear factor icB-mediated apoptosis hiduced by Hehcobacter 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 vaginal delivery based on multivariate analysis of pelvic and other obstetrical factors. JPerinat Med. 1993, 21: 117-24. Huang JT, Welch JS, Ricote M. Binder CJ, Willson TM. Kelly C, Witztum JL, Funk CD, Comad D, Glass CK. Interleukin-4-deρendent production of PPAR-γ ligands in macrophages by 12/15-lipox3'genase. Nature 1999; 400: 378-382.
Ikawa H, Kameda H, Kamitani H, Baek SJ, Nixon JB, His LC, Eling TE. Effect of PPAR activators on cytoldne-stimulated cyclooxygenase-2 expression in human colorectal carcinoma cells. Exp Cell Res 2001; 267: 73-80.
Johnson C, Van Antwerp D, Hope TJ. An N-teπninal nuclear export signal is required for the nucleoc3^toplasmic shuttling of IicBα. EMBO J 1999; 23: 6682-6693.
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 tenn and preterm parturition. Am J Obstet Gynecol 1999; 181 : 1530-1536.
Keirse (1995) "Indomethacin tocolysis in pre-term labour" in Pregnancy and Childbirth Module (Eds. Enkin, M.W., Keirse, M.J.N.C, Renfrew, M.J., Neilson, J.P.) Cochrane Database of Systematic Reviews, No 04383, Oxford).
Kelly RW. Inflammatory mediators and cervical ripening. / Reprod Immunol 2002; 57: 217-224.
Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, Lelimam JM 1995 A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor gamma and promotes adipoc3^te differentiation. Cell 83:813-9. Kunsch C, Ruben SM, Rosen CA. Selection of optimal kappa B/Rel DNA- binding motifs: interaction of both subunits of NF-kappaB with DNA is required for transcriptional activation. Mol Cell Biol 1992; 12: 4412-4421.
D 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 factor0 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, Stimmel 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. Li Q and Verma IM. NF-icB regulation in the iimnune system. Nature Reviews, 2002. 2:725-735.
Lin R, Gewert D, Hiscott J. Differential transcriptional activation in vitro by NF-icB/Rel proteins. JBiol Chem 1995; 270: 3123-3131.
Maul H, Nagel S, Welsch G, Schafer A, Winkler M, Rath W. Messenger ribonucleic acid levels of interleukin- 1 beta, inteι eukin-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 remained unchanged. Eur J Obstet Gynecol Reprod Biol 2002; 102: 143-7. Meade EA, Mclntyre TM, Zimmerman GA, Prescott SM. Peroxisome proliferators enhance cycloox3'genase-2 expression in epithelial cells. J Biol Chem 1999; 274: 8328-8334. Mitchell MD, Edwin SS, Lundin-Schiller S, Silver RM, Smotkiα 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. 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.
Moise et al. Effect of advancing gestational age on the frequency of fetal ductal constriction in association with maternal indomethacin use" Am. J. Obstet. Gynecol, 1995, 170(45), 1204-5.
Narumiya S. Structures, properties and distributions of prostanoid receptors. A dv Prost Thromb Leuk Res 1995; 23 : 17-22.
Narumiya S, Ohno K, Fukushima M, Fujiwara M. Site and mechanism of growth inliibition by prostaglandins. III. Distribution and binding of prostaglandin A2 and delta 12-prostaglandin J2 in nuclei. J Pharmacol Exp Ther 1987; 242: 306-11.
Nasuhura et al, JBC, 1999. 274: 19965.
Pang L, Nie M, Corbett L, Knox AJ. Cycloox3'genase-2 expression by nonsteroidal anti-hifiaimnatory drugs in human airway smooth muscle cells: Role of peroxisome proliferator-activated receptors. J Immunol 2003; 170: 1043-1051.
Phelps CB, Sengchanthalangsy LL, Malek S, Ghosh G. Mechanism of DB DNA binding by Rel/NF-icB dimers. JBiol Chem 2000; 275 : 24392-24399.
Pieber D, AUport VC, Hills F, Johnson M, Bennett PR. Interactions between progesterone receptor isofoπns in myometrial cells in human labour. Mol Hum Reprod. 2001, 7:875-9.
Prusty D, Park B-H, Davis KE, Farmer SR. Activation of MEK/ERK signaling promotes adipogenesis by enhancing peroxisome proliferators- activated receptor γ (PPARγ) and C/EBP α gene expression during the differentiation of 3T3-L1 preadipocytes. J Biol Chem 2002; 277: 46226- 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.
Remington: The Science and Practise of Pharmacy, 19th ed., The Philadelphia College of Pharmacy and Science, ISBN 0-912734-04-3.
Respondek et al, Fetal echocardiography during indomethacin treatment. Ultrasound Obstet Gynecol, 1995. 5, 86-89.
Romero R, Parvizi ST, Oyarzun E, Mazor M, Wu YK, Avila C, Afnanassiadis AP, Mitchell MD. Amniotic fluid interleukin- 1 in spontaneous labour at tenn. J Reprod Med 1990; 35: 235-238. Romero R, Espinoza J, Chaiworapongsa T, Kalache K. Infection and prematurity and the role of preventive strategies. Semin Neonatol. 2002; 7:259-74. Ruan H, Pownall HJ, Lodish HF. Troglitazone antagonizes TNF-α-induced reprogramming of adipocyte gene expression by inhibiting the transcriptional regulatory functions of NF-icB. J Biol Chem 2003; Manuscript M303141200. Rush RW, Keirse MJNC, Howat P, Baum JD, Anderson AB, Tumbull AC. Contribution of preterm delivery to perinatal mortality. Br Med J 1916; 2: 965.
Satoh K, Yasumizu T, Fukuoka H, Kinoshita K, Kaneko Y, Tsuchiya M, Sakamoto 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 Spring Harb our pub .
Schreiber et al, Rapid detection of octomer binding proteins with mini- extracts prepared foπn a small number of cells. Nucl. Acids Res, 1989. 17:6419.
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. 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 J Obstet Gynecol 1995; 172: 77-82. Staels B, Koenig W, Habib A, Merval R, Lebret M, Toπa IP, Delerive P, Fadel A, Chinetti G, Fmchart JC, Najib J, Maclouf J, Tedgui A. Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators. Nature 1998; 393 : 790-793. Straus D. S. and Glass C. K. Cyclopentenone prostaglandins: new insights on biological activities and cellular targets. Med Res Rev, 2001. 1:185- 210.
Subbaramaiah K, Lin DT, Hart JC, Dannenberg AJ. Peroxisome proliferator-activated receptor γ ligands suppress the transcriptional activation of cyclooxygenase-2. JBiol Chem 2001; 276: 12440-12448.
Suyang H, Phillips R, Douglas I, Ghosh S. Role of unphosphoitylated, newly synthesized IicBβ in persistent activation of NF-icB. Mol Cell Biol 1996; 16: 5444-5449.
Suzawa M, Takada I, Yanagisawa J, Ohtake F, Ogawa S, Yamauchi T, Kadowaki T, Takeuchi Y, Shibuya H, Gotoh Y, Matsumoto K, Kato S. C3^tokines suppress adipogenesis and PPAR-γ function through the TAK1/TAB 1/NIK cascade. Nature Cell Biol 2003; 5: 224-230.
Suzuki et al, Three component coupling synthesis of prostaglandins. A simplified, general procedure. Tetrahedron, 1990, 46:4809-4822.
Takata Y, Kitami Y, Yang Z-H, Nakamura M, Okura T, Hiwada K. Vascular inflammation is negatively autoregulated by interaction between CCAAT/enhancer-binding protein-δ and peroxisome proliferator-activated receptor-γ. Circ Res 2002; 91: 427-433.
Takeuchi et al. Adv. Drug. Delic. Rev., 2001, 47:39-54.
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 inflammatory cytokines and its reversal by thiazolidinediones. Diabetologia 1999; 42: 702-710.
Toledano MB, 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.
Tontonoz P, Nagy L, Alvarez J, Thomazy V, Evans R. PPARγ promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell 1998; 93: 241-252.
Tulzer et al, Doppler- -echocardiography of fetal ductus arteriosus constriction versus increased right ventricular output. JACC, 1991. 18(2), 532-36.
Turnbull, A. The fetus and birth, in Elsevier, London. 1977.
Urakubo A, Jarskog LF, Lieberman JA, Gilmore JH. Prenatal exposure to maternal infection alters C3>tokine expression in the placenta, amniotic fluid, and fetal brain. Schizophrenia research 2001; 47: 27-36. Van Meir, C.A., et al, Chorionic prostaglandin catabolism is decreased in the lower uterine segment with tenn labour. Placenta, 1991. 18(2-3): p.109- 14. Van Meir, C.A., et al, Immunoreactive 15-hydroxyprostaglandin dehydrogenase (PGDH) is reduced in fetal membranes from patients at preterm delivery in the presence of infection. Placenta, 1996. 17(5-6): p.291-7.
Volpe JJ. Neurobiology of periventricular leukomalacia in the premature infant. Pediatr Res 2001;50:553-62.
Ward C, Dransfield I, Munay J, Farrow SN, Haslett C, Rossi AG. Prostaglandin D2 and its metabolites induce caspase-dependent granulocyte apoptosis that is mediated via inhibition of IicBα degradation using a peroxisome proliferator-activated receptor-γ-independent mechanism. J Immunol 2002; 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 / 1997(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 α (PPARα) signaling pathways. J Biol Chem 1999; 274: 2672-2681.

Claims

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 inflaimnatory 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 inflaimnatory 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 inflaimnatory 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.
9. A use according to Claim 6 wherein the inflammatory 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: preterm 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 C3'stic change (periventricular leucomalacia, PVL).
14. A use according to any preceding clahn wherein the cyclopentenone prostaglandin is 15-deoxy-Δ12;1 -prostaglandin J2 and/or prostaglandin Aι and/or is a prodrug of 15-deoxy-Δ12'14-prostaglandin J2 and/or prostaglandin A^
15. A use according to Claim 14 wherein the prodrug is PGD2 or PGEi .
16. A use according to any preceding claim wherein the medicament further comprises a phaπnaceutically acceptable excipient, diluent or canier.
17. A use according to an ' preceding claim wherein the medicament is in a foπn adapted for delivery by mouth.
18. A use according to an3' 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 foπn 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 Clahn 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 tocobytic agent.
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 sympathomimetic is Ritodrine.
31. A use according to Clahn 27 wherein the nitric oxide donor is glyceryl trinitrate.
32. A use according to any preceding claim wherein the inflaimnatory response is mediated by NFkB 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.
35. A use according to Claim 34 wherein the cyclopentenone prostaglandin is capable of inhibiting and/or reducing NFicB activity by preventing and/or reducing NFKB production in uterine cells.
36. A pharmaceutical composition comprising a cyclopentenone prostaglandin and a pharmaceutically acceptable earner 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-γ independent mamier.
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 Clahn 38 and combining it with a pharmaceutically acceptable earner.
PCT/GB2004/005087 2003-12-02 2004-12-02 Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour WO2005053706A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP04805915A EP1827451A1 (en) 2003-12-02 2004-12-02 Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour
US10/581,532 US20070282004A1 (en) 2003-12-02 2004-12-02 Use of a Cyclopentenone Prostaglandin for Delaying for the Onset and/or Preventing the Continuation of Labour
JP2006542016A JP2007513133A (en) 2003-12-02 2004-12-02 Use of cyclopentenone prostaglandins to delay the onset of labor and / or prevent continued labor
CA002589908A CA2589908A1 (en) 2003-12-02 2004-12-02 Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour
AU2004294800A 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

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB0327975.9 2003-12-02
GBGB0327975.9A GB0327975D0 (en) 2003-12-02 2003-12-02 Methods of treatment
GBPCT/GB2004/001380 2004-03-29
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

Publications (1)

Publication Number Publication Date
WO2005053706A1 true WO2005053706A1 (en) 2005-06-16

Family

ID=34655231

Family Applications (1)

Application Number Title Priority Date Filing Date
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

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)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3019186A4 (en) * 2013-07-12 2017-03-29 Patrick J. Casey 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

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2293101A (en) * 1994-09-14 1996-03-20 British Tech Group Butaprost compositions for preventing 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

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2293101A (en) * 1994-09-14 1996-03-20 British Tech Group Butaprost compositions for preventing 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

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
GROSS G ET AL: "INHIBITION OF CYCLOOXYGENASE-2 PREVENTS INFLAMMATION-MEDIATED PRETERM LABOR IN THE MOUSE", AMERICAN JOURNAL OF PHYSIOLOGY, AMERICAN PHYSIOLOGICAL SOCIETY, BETHESDA, MD, US, vol. 278, no. 6, PART 2, June 2000 (2000-06-01), pages R1415 - R1423, XP008008804, ISSN: 0002-9513 *
LAPPAS M ET AL: "Regulation of proinflammatory cytokines in human gestational tissues by peroxisome proliferator-activated receptor-[gamma]: Effect of 15-deoxy-[delta]<12,14>-PGJ2 and troglitazone", JOURNAL OF CLINICAL ENDOCRINOLOGY AND METABOLISM 01 OCT 2002 UNITED STATES, vol. 87, no. 10, 1 October 2002 (2002-10-01), pages 4667 - 4672, XP002291977, ISSN: 0021-972X *
LAPPAS MARTHA ET AL: "Nuclear factor kappa B regulation of proinflammatory cytokines in human gestational tissues in vitro", BIOLOGY OF REPRODUCTION, vol. 67, no. 2, August 2002 (2002-08-01), pages 668 - 673, XP002291976, ISSN: 0006-3363 *
STRAUS D S ET AL: "CYCLOPENTENONE PROSTAGLANDINS: NEW INSIGHTS ON BIOLOGICAL ACTIVITIES AND CELLULAR TARGETS", MEDICINAL RESEARCH REVIEWS, NEW YORK, NY, US, vol. 21, no. 3, May 2001 (2001-05-01), pages 185 - 210, XP009035137, ISSN: 0198-6325 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3019186A4 (en) * 2013-07-12 2017-03-29 Patrick J. Casey 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

Also Published As

Publication number Publication date
JP2007513133A (en) 2007-05-24
EP1827451A1 (en) 2007-09-05
AU2004294800A1 (en) 2005-06-16
CA2589908A1 (en) 2005-06-16

Similar Documents

Publication Publication Date Title
Olson The role of prostaglandins in the initiation of parturition
Hertelendy et al. Regulation of myometrial smooth muscle functions
WO1995022345A1 (en) A method and agents for control and management of labor during pregnancy
US20080206309A1 (en) Antagonists of prostaglandin receptors ep2 and/or ep4 for the treatment of dysmenorrhea and menorrhagia
JP6273072B2 (en) Compositions and methods of treating cardiac fibrosis with ifetroban
TW202137980A (en) Lasofoxifene treatment of breast cancer
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
EP1827451A1 (en) Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour
JP2005537225A (en) FP receptor antagonist or PGF2α antagonist for treatment of uterine pathology
Gibb et al. Parturition
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
JP2006517576A (en) IP receptor antagonist for the treatment of pathological uterine symptoms
Sunita et al. Histone deacetylase: A potential therapeutic target for ovarian dysfunction
US11464831B2 (en) Compositions and methods using IL-8 for improving health of mammals
JP2005532295A (en) FP receptor antagonist or PGF2α antagonist for treating menorrhagia
M. COLE RF LAMONT Current perspectives on drug treatment for preterm labour
Neuman et al. The prostaglandin E2 EP3 receptor has disparate effects on islet insulin secretion and content in β-cells in a high-fat diet-induced mouse model of obesity
Seymour Examining the Influence of Muscle Fiber Type on Protein Turnover Signaling in Growing Pigs
Yu et al. Myometrium infection decreases TREK1 through NHE1 and increases contraction in pregnant mice
Zhao et al. Na+-Leak Channel, Non-Selective (NALCN) Regulates Myometrial Excitability and Facilitates Successful Parturition.
Hooper et al. Selective serotonin reuptake inhibitor (SSRI) exposure 1 constricts the mouse ductus arteriosus in utero 2
JP2024020797A (en) Treatment for muscle atrophy

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2006542016

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

WWE Wipo information: entry into national phase

Ref document number: 2004294800

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004805915

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2004294800

Country of ref document: AU

Date of ref document: 20041202

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004294800

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2589908

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 10581532

Country of ref document: US

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWP Wipo information: published in national office

Ref document number: 2004805915

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 10581532

Country of ref document: US