WO2012146936A1 - Pyrrolnitrin derivatives - Google Patents

Pyrrolnitrin derivatives Download PDF

Info

Publication number
WO2012146936A1
WO2012146936A1 PCT/GB2012/050937 GB2012050937W WO2012146936A1 WO 2012146936 A1 WO2012146936 A1 WO 2012146936A1 GB 2012050937 W GB2012050937 W GB 2012050937W WO 2012146936 A1 WO2012146936 A1 WO 2012146936A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
cancer
compound
pyrrolnitrin
salt
Prior art date
Application number
PCT/GB2012/050937
Other languages
French (fr)
Inventor
Charles Roland Wolf
Anna Louise Westwood
Roger Neil Sleigh
Original Assignee
Cxr Biosciences 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
Application filed by Cxr Biosciences Limited filed Critical Cxr Biosciences Limited
Publication of WO2012146936A1 publication Critical patent/WO2012146936A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4025Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil not condensed and containing further heterocyclic rings, e.g. cromakalim
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to the medical use of pyrrolnitrin derivatives and particularly their use in the treatment of cancer.
  • Cancer (or neoplasm) covers a range of diseases in which a group of cells display uncontrolled growth (division beyond the normal limits), invasion (intrusion on and destruction of adjacent tissues), and sometimes metastasis (spread to other locations in the body via lymph or blood). Cancer affects people at all ages with the risk for most types increasing with age and caused about 13% of all human deaths in 2007. Deaths from cancer worldwide are projected to continue rising, with an estimated 12 million deaths in 2030. (WHO, February 2009)
  • cancer treatment is based around surgery, chemotherapy and radiotherapy.
  • the effectiveness of surgery is often limited by the propensity of cancers to invade adjacent tissue or to spread to distant sites by microscopic metastasis.
  • the effectiveness of chemotherapy is often limited by toxicity to other tissues in the body. Radiation can also cause damage to normal tissue.
  • targeted therapy has had a significant impact in the treatment of some types of cancer. This constitutes the use of agents specific for the deregulated proteins of cancer cells.
  • Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell.
  • agents are selected as having a promising activity against a particular biological target thought to be important in disease; however, little will be known about the safety, toxicity, pharmacokinetics and metabolism of this agent in humans. Therefore, it is traditionally necessary to assess all of these parameters prior to human clinical trials in order to be able to recommend a dose and schedule to be used the first time.
  • Pyrrolnitrin is an anti-fungal, secondary metabolite, produced by a range of bacteria. It was originally described as acting by inhibiting the electron transport chain (Lyr, H. 1977). Mutants defective in components of the HOG pathway are resistant to the synthetic derivatives of pyrrolnitrin, fludioxonil and fenpiclonil. These compounds normally induce cell death by activating the HOG pathway resulting in accumulation of intracellular glycerol, cell swelling and ultimately cell death.
  • Pyrrolnitrin continues to be marketed in several countries for the treatment of fungal infections of the skin. For example, Pyrrolnitrin alone is marketed by Pharmacia in Italy as Micutrin and in combination with betametasone valerate as Beta Micutrin.
  • pyrrolnitrin derivatives can be used in the treatment of cancer (such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases) and may also be useful in the treatment of vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma osteoporosis, reperfusion injury, asthma, fibrosis, anti-inflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, and other indications mediated by Rho-kinase, e.g., coronary heart disease.
  • cancer such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases
  • vasospasm such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases
  • the compounds of the invention are useful to treat erectile dysfunction, i.e. erectile dysfunction mediated by Rho-kinase.
  • Erectile dysfunction can be defined as an inability to obtain or sustain an erection adequate for intercourse.
  • Fludioxonil and fenpiclonil are two such derivatives that originated from a lead optimisation programme at Ciba Geigy, who used pyrrolnitrin as the starting structure for a developmental chemistry program, with the aim of identifying new fungicides. This is described in Nyfeler and Ackermann (1992) Phenylpyrroles, a new class of agricultural fungicide related to the natural product pyrrolnitrin and in Baker, D. et al, Synthesis and Chemistry of Agrochemicals III (pp 395 - 404) Washington, D.C. American Chemical Society. Fludioxonil and fenpiclonil were further developed by Ciba Geigy and were initially registered as agricultural fungicides in 1991 and 1988 respectively. They are still used commercially for this purpose today.
  • Fenpiclonil was first registration as agricultural fungicide in 1988, in Switzerland.
  • Commercial products include BERET and GALBAS sold by Syngenta.
  • Fludioxonil was first registration as agricultural fungicide in 1991, in France.
  • Commercial products include SAPHIRE, CELEST and MAXIM sold by Syngenta. These compounds are not known or sold for the treatment of any non-agricultural use.
  • derivatives of pyrrolnitrin may be useful in medicine, particularly in the treatment of cancer (such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases) and may also be useful in the treatment of vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma osteoporosis, reperfusion injury, asthma, fibrosis, antiinflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, and other indications mediated by Rho-kinase, e.g., coronary heart disease.
  • cancer such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases
  • vasospasm such as cancers of the breast, colon, prostate, ovaries, brain and lung and their
  • the compounds of the invention are useful to treat erectile dysfunction, i.e. erectile dysfunction mediated by Rho-kinase.
  • Erectile dysfunction can be defined as an inability to obtain or sustain an erection adequate for intercourse.
  • This invention is not intended to cover pyrrolnitrin itself, but only derivatives of pyrrolnitrin as described in the following aspects of the invention.
  • A aromatic or heteroaromatic substituted with 1-3 N in the ring
  • B aromatic or a partially or fully reduced five or six membered ring.
  • Either A or B rings can be optionally benzo fused.
  • Fludioxonil (CAS number: 131341-86-1) is represented by the following structure:
  • the pyrrolnitrin derivative is fenpiclonil or a salt or an ester thereof.
  • Fenpiclonil (CAS number: 74738-17-3) is represented by the following structure:
  • Fludioxonil and Fenpiclonil are inhibitors of PIM kinase family members (PIM1 , PIM2 and PIM3) and also of ROCKIl kinase.
  • PIM kinases are cytoplasmic serine/threonine kinases that are known to be involved in regulation of apoptosis and cellular metabolism. Certain PIM kinases have been shown to be upregulated in cancers and as such their inhibition represents a mechanism of action by which Fludioxonil and Fenpiclonil can have an anti-tumour effect in conditions such as leukaemia, lymphoma, prostate cancer, colon cancer and pancreatic cancer. The below studies have shown this link:
  • Liver cancer Gong (2009), Fujii (2005) and Wu (2010) have shown PIM-2 to promote humourigenesis and PIM-3 to accelerate hepatocellular carcinoma development when induced by hepatocarcinogen.
  • Colon cancer Popivanova (2007) has shown PIM-3 to be aberrantly expressed in human colon cancer cells but not normal colon mucosa.
  • PIM-3 expression occurs in human pancreatic cancer but not normal cells and PIM-1 blockage using siRNA resensitises pancreatic cancer cells to apoptosis and PIM-1 levels correlate to clinicopathological parameters in pancreatic cancer.
  • PIM-2 expression is increased in leukaemia/lymphoma
  • expression of PIM-1 and PIM-2 is dependent on Abl kinase activity and PIM-1 mediates homing and migration of malignant haematopoietic cells.
  • Oral cancer Chiang (2006) and Choi (2010) have shown PIM-1 expression to be high in squamous cell carcinoma.
  • Prostrate cancer Chen (2005), umenthaler (2009), He (2007), Xu (2005), Dai (2005) and Roh (2008) have shown PIM-1 overexpression in prostatic carcinoma.
  • Adipocyte tumours Nga (2010) has shown benign and malignant adipocytic tumours to have strong PIM-1 expression.
  • PIM kinases are constitutively active and their activity as shown above and in Amaravadi (2005) and Shah (2008) supports in vitro and in vivo human cell growth and survival.
  • the pathology of a number of human and animal diseases including hypertension, erectile dysfunction, coronary cerebral circulatory impairments, neurodegenerative disorders and cancer can be linked directly to changes in the actin cytoskeleton. These diseases pose a serious unmet medical need.
  • the actin cytoskeleton is composed of a meshwork of actin filaments and actin-binding proteins found in all eukaryotic cells. In smooth muscle cells the assembly and disassembly of the actin cytoskeleton is the primary motor force responsible for smooth muscle contraction and relaxation.
  • the actin cytoskeleton is controlled by a family of proteins that are a subset of the Ras superfamily of GTPases. This subset currently consists of RhoA through E and RhoG (refereed to collectively as Rho), Rac 1 and 2, Cdc42Hs and G25K and TC10 isoforms (Mackay, et al. JBiol Chem 1998,273,20685). These proteins are GTP (guanine nucleotide triphosphate) binding proteins with intrinsic GTPase activity. They act as molecular switches and cycles between inactive GDP (guanine nucleotide diphosphate) bound and active GTP bound states. Using biochemical and genetic manipulations, it has been possible to assign functions to each family member.
  • Rho proteins controls the formation of actin stress fibers, thick bundles of actin filaments, and the clustering of integrins at focal adhesion complexes.
  • Rac proteins control the formation of lamellopodia or membrane ruffles on the cell surface and Cdc42 controls filopodia formation.
  • This family of proteins plays a critical part in the control of key cellular functions including cell movement, axonal guidance, cytokinesis, and changes in cell morphology, shape and polarity.
  • Rho proteins can control different biological responses.
  • Rho proteins are responsible for the calcium sensitization during smooth muscle contraction.
  • the Rho GTPases are responsible for the cellular responses to agonist such as lysophosphatidic acid (LPA), thrombin and thromboxane A2 (Fukata, et al. Trends Pharcol Sci 2001 , 22,32).
  • Rho GTPases activate a number of downstream effectors including PIP5-kinase, Rhothekin, Rhophilin, PKN and Rho-Kinase isoforms ROCK-1/ROKbeta and ROCK-2/ROKalpha (Mackay and Hall J Biol Chem 1998,273, 20685; Aspenstrom Curr Opin Cell Biol 1999, 11 , 95; Amano, et al.
  • Rho-kinase was identified as a RhoA interacting protein isolated from bovine brain (Matsui, et al. Embo J 1996,15,2208). It is a member of the myotonic dystrophy family of protein kinase and contains a serine/threonine kinase domain at the amino terminus, a coiled- coil domain in the central region and a Rho interaction domain at the carboxy terminus (Amano, et al. Exp Cell Res 2000,261 ,44). Its kinase activity is enhanced upon binding to GTP-bound RhoA and when introduced into cells, it can reproduce many of the activities of activated RhoA.
  • Rho-Kinase mediates calcium sensitization and smooth muscle contraction and inhibition of Rho-kinase blocks 5-HT and phenylephrine agonist induced muscle contraction.
  • Rho- kinase When introduced into non- smooth muscle cells, Rho- kinase induces stress fiber formation and is required for the cellular transformation mediated by RhoA (Sahai, et al. Curr Biol 1999, 9,136).
  • Rho- kinase regulates a number of downstream proteins through phosphorylation, including myosin light chain (Somlyo, et al. J Physiol (Lond) 2000,522 Pt 2,177), the myosin light chain phosphatase binding subunit (Fukata, et al.
  • ROCK kinases are effectors of the small GTPase Rho and belong to the AGC family of kinases.
  • the Rho-associated kinases (ROCKs) are serine/threonine protein kinases that serve as key downstream effectors of the Rho GTPase, RhoA, and play an important role in cytoskeletal function.
  • the Rho/ROCK signal transduction participates in signalling pathways via rearrangement of the actin cytoskeleton that leads to various cellular reactions.
  • ROCKs impact cytoskeletal function through their ability to phosphorylate a range of proteins that directly regulate cytoskeletal proteins, including myosin light chain kinase, myosin light chain phosphatase, beta-catenin, cofilin, FAK and LIMK, and thus the ROCKs are positioned to play essential roles in the physiological and pathophysiological processes of various cell types.
  • the ROCK enzymes, ROCK1 and ROCK2 share 65% overall homology. Gene knockout and siRNA studies suggest that each isoform plays distinct roles in mammalian biology. Despite having similar kinase domains, ROCK1 and ROCK2 may serve different functions and may have different downstream targets.
  • ROCK1 expression tends to be ubiquitous, while ROCK2 is most highly expressed in cardiac and brain tissues. Clinically, inhibition of the ROCK pathway is believed to contribute to some of the cardiovascular benefits of statin therapy that are independent of lipid lowering (Liao et al 2010, J Cardiovascular Pharmacol 2007, 50(1):17-24).
  • Rho kinase is involved in a wide range of diseases such as glaucoma, vasospasm, pulmonary hypertension multiple sclerosis and nerve injury.
  • Pharmacology studies of ROCK kinase inhibitors have shown that compounds of this activity are effective in animal models of glaucoma (for example at reducing intraocular pressure (IOP) in animal models by improving aqueous hour drainage through the trabecular pathway).
  • IOP intraocular pressure
  • Inhibition of Rho-kinase activity in animal models has demonstrated a number of benefits of Rho-kinase inhibitors for the treatment of human diseases.
  • Rho-kinase activity has benefits for controlling cerebral vasospasms and ischemia following subarachnoid hemorrhage (Pharma Japan 1995,1470, 16).
  • Non-isoform selective ROCK inhibitors such as fasudil have been shown to prevent cerebral vasospasm after subarachnoid hemorrhage.
  • fasudil is the only ROCK inhibitor approved for clinical use. Fasudil was approved in 1995 in Japan and China for prevention and treatment of cerebral vasospasm following surgery for subarachnoid hemorrhage and has since been used in over 124,000 patients in Japan.
  • Many pharmaceutical and biotechnology companies are developing other selective and non-selective ROCK inhibitors (see table below modified and updated from Liao et al 2007).
  • ROCK inhibition will not have any safety limiting factors and that ROCK is a significant and genuine drug target.
  • ROCK inhibitors such as Y27632 have been shown to inhibit additional kinases.
  • Compounds selective for ROCK represent a significant advancement over those described currently as ROCK inhibitors.
  • a compound may have a particular advantage over and above those inhibiting these kinases individually because the PIM kinase and ROCK kinase pathways intersect through p21Cip1/WAF1 (hereafter referred to as p21).
  • p21 the PIM kinase phosphorylates p21 on Thr 145 and thus regulates its stability and cellular localization in cells (Zhang et al., 2007, Mol Cancer Res 5(9): 909-922).
  • Thr 145 When p21 is phosphorylated on Thr 145 it localizes to the nucleus and results in the disruption of the association between proliferating cell nuclear antigen (PCNA) and p21. Furthermore phosphorylation of Thr 145 promotes stabilization of p21. Inhibition of PIM and the consequent reduction in phosphorylation of Thr 145 of p21 would result in reduced stability of p21, and a reduction in cell proliferation. P21 also intersects with the ROCK/LIMK/Cofilin pathway. P21 forms a physical complex with ROCK and inhibits its activity (Lee et al., 2004, Journal Biological Chemistry 279(3): 1885-1891).
  • Ras activates p21 transcription and promotes p21 protein stability and it has been suggested that p21 is an essential factor in the signalling pathways that contribute to Ras-induced actin cytoskeletal re-modelling.
  • the inhibition of PIM and ROCK together may therefore be particularly significant in tumours dependent upon or transformed through Ras mutation.
  • salts include acid addition salts and base addition salts.
  • Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound of formula I with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
  • ester is included those formed with an alcohol of formula R 1 OH, wherein R 1 represents aryl or alkyl; and those formed with a thiol of formula R SH, wherein R 1 is as hereinbefore defined (i.e. a thioester). It is preferred that the ester is not a thioester.
  • R 1 represents C 1-6 alkyl, for example Ci alkyl (eg methyl).
  • Compounds of the invention may contain double bonds and may thus exist as E (entadel) and Z (zusammen) geometric isomers about each individual double bond. All such isomers and mixtures thereof are included within the scope of the invention.
  • Compounds of the .invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism.
  • Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation.
  • the various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques.
  • the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation (i.e.
  • a 'chiral pool' method by reaction of the appropriate starting material with a 'chiral auxiliary' which can subsequently be removed at a suitable stage, by derivatisation (i.e. a resolution, including a dynamic resolution), for example with a homochiral acid followed by separation of the diastereomeric derivatives by conventional means such as chromatography, or by reaction with an appropriate chiral reagent or chiral catalyst all under conditions known to the skilled person. All stereoisomers and mixtures thereof are included within the scope of the invention.
  • C 1-q alkyl groups (where q is the upper limit of the range) defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of two or three, as appropriate) of carbon atoms, be branched-chain, and/or cyclic (so forming a Ca-q-cycloalkyl group).
  • Such cycloalkyl groups may be monocyclic or bicyclic and may further be bridged. Further, when there is a sufficient number (i.e. a minimum of four) of carbon atoms, such groups may also be part cyclic.
  • Such alkyl groups may also be saturated or, when there is a sufficient number (i.e.
  • C 1-q alkyl groups may also be spiro-groups (i.e. two cycloalkyi rings linked together by a single common carbon atom), although they are preferably not so.
  • halo when used herein, includes fluoro, chloro, bromo and iodo.
  • Heterocycloalkyi groups that may be mentioned include non-aromatic monocyclic and bicyclic heterocycloalkyi groups (which groups may further be bridged) in which at least one (e.g. one to four) of the atoms in the ring system is other than carbon (i.e. a heteroatom), and in which the total number of atoms in the ring system is between three and twelve (e.g. between five and ten). Further, such heterocycloalkyi groups may be saturated or unsaturated containing one or more double and/or triple bonds, forming for example a C 2-q heterocycloalkenyl (where q is the upper limit of the range) or a C 7-q heterocycloalkynyl group.
  • C 2-q heterocycloalkyi groups that may be mentioned include 7- azabicyclo-[2.2.1]heptanyl, 6-azabicyck>[3.1.1]heptanyl, 6-azabicyclo[3.2.1]-octanyl, 8- azabicyclo[3.2.1]octanyl, aziridinyl, azetidinyl, dihydropyranyl, dihydropyridyl, dihydropyrrolyl (including 2,5-dihydropyrrolyl), dioxolanyl (including 1 ,3-dioxolanyl), dioxanyl (including 1 ,3-dioxanyl and 1 ,4-dioxanyl), dithianyl (including 1,4-dithianyl), dithiolanyl (including 1 ,3-dithiolanyl), imidazolidinyl, imidazolinyl, morpholinyl, 7- oxabicyclo[2.
  • Substituents on heterocycloalkyi groups may, where appropriate, be located on any atom in the ring system including a heteroatom. Further, in the case where the substituent is another cyclic compound, then the cyclic compound may be attached through a single atom on the heterocycloalkyi group, forming a so-called "spiro"-compound.
  • the point of attachment of heterocycloalkyi groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system.
  • Heterocycloalkyi groups may also be in the N- or S- oxidised form.
  • bicyclic refers to groups in which the second ring of a two-ring system is formed between two adjacent atoms of the first ring.
  • bridged refers to monocyclic or bicyclic groups in which two non-adjacent atoms are linked by either an alkylene or heteroalkylene chain (as appropriate).
  • Aryl groups that may be mentioned include C 6- 14 (such as Ce. ⁇ (e.g. C 6-10 )) aryl groups. Such groups may be monocyclic or bicyclic and have between 6 and 14 ring carbon atoms, in which at least one ring is aromatic.
  • C 6- i 4 aryl groups include phenyl, naphthyl and the like, such as 1,2,3,4-tetrahydronaphthyl, indanyl, indenyl and fluorenyl.
  • the point of attachment of aryl groups may be via any atom of the ring system. However, when aryl groups are bicyclic or tricyclic, they are preferably linked to the rest of the molecule via an aromatic ring.
  • Heteroaryl groups that may be mentioned include those which have between 5 and 14 (e.g. 10) members. Such groups may be monocyclic, bicyclic or tricyclic, provided that at least one of the rings is aromatic and wherein at least one (e.g. one to four) of the atoms in the ring system is other than carbon (i.e. a heteroatom).
  • Heteroaryl groups that may be mentioned include acridinyl, benzimidazolyl, benzodioxanyl, benzodioxepinyl, benzodioxolyl (including 1,3-benzodioxolyl), benzofuranyl, benzofurazanyl, benzothiazolyl, benzoxadiazolyl (including 2,1 ,3-benzoxadiazolyl), benzoxazinyl (including 3,4-dihydro-2H-1,4-benzoxazinyl), benzoxazolyl, benzomorpholinyl, benzoselenadiazolyl (including 2,1 ,3-benzoselenadiazolyl), benzothiadiazolyl (including 2,1 ,3-benzothiadiazolyl), benzothienyl, carbazolyl, chromanyl, cinnolinyl, furanyl, imidazolyl, imidazopyridyl (including
  • heteroaryl groups may, where appropriate, be located on any atom in the ring system including a heteroatom.
  • the point of attachment of heteroaryl groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system.
  • heteroaryl groups when polycyclic, they are preferably linked to the rest of the molecule via an aromatic ring.
  • Heteroaryl groups may also be in the N- or S- oxidised form.
  • Heteroatoms that may be mentioned include phosphorus, silicon, boron, tellurium, selenium and, preferably, oxygen, nitrogen and sulphur.
  • a pharmaceutical composition comprising a pyrrolnitrin derivative of formula I or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier.
  • composition comprising between 10mg and 2000mg of an active ingredient per dosage unit, wherein the active ingredient is a compound of the invention or a derivative, salt or variant thereof.
  • dosage unit we mean the unit of medicament administered to a patient at one time.
  • the dosage unit, or single dose may be administered by a single capsule/tablet, single injection, or single intravenous infusion, a single subcutaneous injection, or by a single procedure using other routes of administration, as discussed below.
  • the single dose may be administered to the patient by two or more capsules/tablets or injections given simultaneously or sequentially to deliver the entire dose to the patient in the continuous, single and defined treatment period; by two or more intravenous infusions given simultaneously or sequentially to deliver the entire dose to the patient in the continuous, single and defined treatment; or by multiple procedures using other routes of administration as discussed below.
  • the single dose to be administered to the patient can be delivered by a combination of routes to deliver the entire dose to the patient in the continuous, single and defined treatment.
  • the dosage unit may then be repeated at intervals of time such as a few hours, days, weeks, or months later.
  • Dosage units can be administered to patients in such a way that the patient receives a loading dose followed by one or more maintenance doses.
  • the loading dose may be a high dose in order to quickly reach a desired plasma concentration and then subsequent maintenance doses are a lower dose than the loading dose in order to maintain the required plasma concentration.
  • active ingredient we mean the molecule having the desired effect.
  • variants and derivatives we mean any molecules of substantially identical chemical structure but including minor modifications that do not alter activity but may offer improved or alternative properties for formulation, such as formation into a salt.
  • the compound of the invention containing composition, and medicaments of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient, diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the compound of the invention containing composition, and medicaments 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 compound of the invention containing composition, and medicaments of the invention may also be administered via intracavernosal injection.
  • Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, 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 glycine
  • disintegrants such as starch (preferably com, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates,
  • 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 compound of the invention containing composition, medicaments and pharmaceutical compositions 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.
  • composition, and medicaments of the invention can also be administered parenterally, for example, intravenously, intra- arterially, intraperitoneally, intra-thecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They 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.
  • suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.
  • Medicaments and pharmaceutical compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain antioxidants, 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 medicaments and pharmaceutical compositions 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 requiring 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 kind previously described.
  • composition, and medicaments of the invention can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134A3 or 1,1,1,2,3,3,3- heptafluoropropane (HFA 227EA3), carbon dioxide or other suitable gas.
  • a suitable propellant e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active agent, e.g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e.g. sorbitan trioleate.
  • a lubricant e.g. sorbitan trioleate.
  • Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the invention containing composition, of the invention and a suitable powder base such as lactose or starch.
  • Aerosol or dry powder formulations are preferably arranged so that each metered dose or "puff contains an effective amount of an agent or polynucleotide of the invention for delivery to the patient. It will be appreciated that the overall daily dose with an aerosol will vary from patient to patient, and may be administered in a single dose or, more usually, in divided doses throughout the day.
  • the compound of the invention containing composition, and medicaments of the invention can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, gel, ointment or dusting powder.
  • the compound of the invention containing composition, and medicaments of the invention may also be transdermal ⁇ administered, for example, by the use of a skin patch. They may also be administered by the ocular route, particularly for treating diseases of the eye.
  • the compound of the invention containing composition, and medicaments of the invention can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride.
  • a preservative such as a benzylalkonium chloride.
  • they may be formulated in an ointment such as petrolatum.
  • the compound of the invention containing composition, and medicaments of the invention can be formulated as a suitable ointment containing the active agent suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene agent, emulsifying wax and water.
  • compositions 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.
  • oral or parenteral administration of the compound of the invention containing composition, medicaments and pharmaceutical compositions of the invention is the preferred route, being the most convenient.
  • the compound of the invention containing composition, and medicaments of the invention are administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
  • composition as defined herein may be formulated as described in the accompanying Examples.
  • the composition may comprise any effective amount of active ingredient, this may be between 10mg and 2000mg of active ingredient per dosage unit, and preferably is between 50mg and lOOOmg.
  • the dosage unit contains an amount of active ingredient per dosage unit selected from 10mg, 20mg, 25mg, 50mg, 100mg, 200mg, 300mg, 400mg, 450mg, 600mg, 750mg, 950mg, 10OOmg and 1200mg.
  • the composition may comprise between 10-50mg, 10-75mg, 10-100mg, 10- 200mg, 10-300mg, 10-400mg, 10-600mg, 10-750mg, 10-950mg, 10-1 OOOmg, 10- 1200mg, 50-75mg, 50-100mg, 50-200mg, 50-300mg, 50-450mg, 50-600mg, 50-750mg, 50-950mg, 50-1 OOOmg, 50-1200mg, 75-1 OOmg, 75-200mg, 75-300mg, 75-450mg, 75- 600mg, 75-750mg, 75-950mg, 75-1000mg, 75-1200mg, 100-200mg, 100-300mg, 100- 450mg, 100-600mg, 100-750mg, 100-950mg, 100-1 OOOmg, 100-1200mg, 200-300mg, 200-450mg, 200-600mg, 200-750mg, 100
  • composition is pharmaceutically acceptable, and may optionally contain a pharmaceutically acceptable excipient, diluent, carrier or filler.
  • a further aspect of the invention provides a method of treating cancer (such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases) comprising administering to the patient an effective amount of a pyrrolnitrin derivative of formula I or a salt or an ester thereof.
  • cancer such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases
  • a further aspect of the invention provides a method of treating diseases and conditions selected from vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma osteoporosis, reperfusion injury, asthma, fibrosis, anti-inflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, and other indications mediated by Rho-kinase, e.g., coronary heart disease.
  • the compounds of the invention are useful to treat erectile dysfunction, i.e. erectile dysfunction mediated by Rho-kinase. Erectile dysfunction can be defined as an inability to obtain or sustain an erection adequate for intercourse.
  • patient includes all animals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, and pigs.
  • patient means an animal having a disorder in need of treatment. Preferably, the patient is selected from humans, cows, dogs, cats, goats, sheep, and pigs. More preferably, the patient is a human.
  • the compounds of the invention may be used in cancer treatment either alone or in combination with well known anti-cancer agents.
  • compositions of the invention may be effective in adult and pediatric oncology including in solid phase tumours/malignancies, locally advanced tumours, human soft tissue sarcomas, metastatic cancer, including lymphatic metastases, blood cell malignancies including multiple myeloma, acute and chronic leukemias, and lymphomas, head and neck cancers including mouth cancer, larynx cancer and thyroid cancer, lung cancers including small cell carcinoma and non-small cell cancers, breast cancers including small cell carcinoma and ductal carcinoma, gastrointestinal cancers including esophageal cancer, stomach cancer, colon cancer, colorectal cancer and polyps associated with colorectal neoplasia, pancreatic cancers, liver cancer, urologic cancers including bladder cancer and prostate cancer, malignancies of the female genital tract including ova
  • Compounds of the invention may be administered to treat cancer.
  • the cancer to be treated is preferably selected from pancreatic cancer, ovarian cancer, breast cancer, prostate cancer, liver cancer, chondrosarcoma, lung cancer, head and neck cancer, colon cancer, sarcoma, leukaemia, myeloma, lymphoma, kidney cancer, thyroid cancer and brain cancers such as glioblastoma.
  • compositions can be administered in therapeutically effective dosages alone or in combination with adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy, and may provide a beneficial effect, e.g. reducing tumour size, slowing rate of tumour growth, inhibiting metastasis, or otherwise improving overall clinical condition, without necessarily eradicating the cancer.
  • adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy
  • composition can also be administered in therapeutically effective amounts as a portion of an anti-cancer cocktail.
  • An anti-cancer cocktail is a mixture of the compound or modulator of the invention with one or more anti-cancer drugs in addition to a pharmaceutically acceptable carrier for delivery. The use of anti-cancer cocktails as a cancer treatment is routine.
  • Anti-cancer drugs that are well known in the art and can be used as a treatment in combination with the compounds of the invention include: 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin
  • therapeutic compositions of the invention may be used for prophylactic treatment of cancer.
  • hereditary conditions and/or environmental situations e.g. exposure to carcinogens
  • In vitro models can be used to determine the effective doses of the compounds of the invention as a potential cancer treatment.
  • These in vitro models include proliferation assays of cultured tumour cells, growth of cultured tumour cells in soft agar (see Freshney, (1987) Culture of Animal Cells: A Manual of Basic Technique, Wily-Liss, New York, NY Ch 18 and Ch 21), tumour systems in nude mice as described in Giovanella et a/., J. Natl. Can.
  • tumour cells in Boyden Chamber assays as described in Pilkington et at., Anticancer Res., 17: 4107-9 (1997), and angiogenesis assays such as induction of vascularization of the chick chorioallantoic membrane or induction of vascular endothelial cell migration as described in Ribatta et a/., Intl. J. Dev. Biol., 40: 1189-97 (1999) and Li ef a/., Clin. Exp. Metastasis, 17:423-9 (1999), respectively.
  • Suitable tumour cells lines are available, e.g. from American Type Tissue Culture Collection catalogues.
  • the method, use or composition of the invention additionally comprises a further chemotherapeutic agent.
  • the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Aisterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3,
  • the compounds may be used to treat patients who may not be amenable to conventional chemotherapeutic agents.
  • Conventional chemotherapies are generally immunosuppressive and so are withdrawn in certain situations, such as for patients who are pre-operative, post-operative, receiving radiation, terminally ill, elderly, or receiving adjuvant or neo-adjuvant thereapy.
  • the compounds of the invention may still be used in such situations.
  • the compounds of the invention are used to treat patients who may not be amenable to conventional chemotherapeutic agents, particularly those from whom conventional chemotherapeutic agents have been withdrawn.
  • a further aspect of the invention provides a kit of parts comprising:
  • the kit of parts additionally comprises a further chemotherapeutic agent.
  • the further chemotherapeutic agent is selected from 17AAG, 17D AG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Ci
  • Figure 1 shows the % inhibition of Pim1, 2 and 3 kinase with increasing concentration ( ⁇ ) of fludioxonil (1A) or fenpiclonil (1B).
  • Figure 2 Kinase specificity of fludioxonil and fenpiclonil
  • Figure 2 shows by western blot PIM 1 kinase and cleaved PARP levels in response to increasing concentrations if fludioxonil and fenpiclonil.
  • Figure 3 shows the % inhibition of Rock I and Rock II kinase with increasing concentration of fludioxonil or fenpiclonil.
  • Figure 4 Bodyweights and liver weights from a PC3 xenograft study in mice
  • Figure 4 shows (a) bodyweight over duration of study (b) terminal bodyweight and (c) liver weight of mice from a PC3 xenograft study with fludioxonil (CXR6032) and fenpiclonil (CXR6069).
  • Figure 5 Tumour volume and weight from a PC3 xenograft study in mice
  • Figure 5 shows (a) ALT, AST and ALP levels, (b) tumour volume and (c) tumour weight over the duration of the PC3 xenograft study with fludioxonil (CXR6032) and fenpiclonil (CXR6069).
  • Figure 6 shows the Log10 of fludioxonil concentration in ng/mL over time, both with a single dose and with 4 daily repeat doses.
  • Figure 7 shows the Log10 of fenpiclonil concentration in ng/mL over time, both with a single dose and with 4 daily repeat doses.
  • Figure 8 Diagram of GM-CSF signalling pathway overlaid with fludioxonil (CXR6032) EC50 signature list
  • FIG. 9 Diagram of GM-CSF signalling pathway overlaid with fenpiclonil (CXR6069) EC50 signature list
  • Figure 10 Diagram of acute myeloid leukaemia signalling pathway overlaid with fludioxonil (CXR6032) EC50 signature list
  • Figure 11 Diagram of acute myeloid leukemia signalling pathway overlaid with fenpiclonil (CXR6069) EC50 signature list
  • Figure 11 shows the fold change data from fenpiclonil (CXR6032) EC50 signature Rst superimposed on the genes shown in the above pathway using IPA software (Ingenuity Systems Inc.).
  • Fold changes black lettering under gene icons
  • Icons in medium grey signify fold changes of less than 1.5.
  • FIG. 12 Diagram of RhoA signalling pathway overlaid with fludioxonil (CXR6032) EC50 signature list
  • FIG 13 Diagram of RhoA signalling pathway overlaid with fenpiclonil (CXR6069) EC50 signature list
  • Figure 14 RNA microarray for ftudioxonil and fenpiclonil experimental structure
  • Figure 14 shows the fludioxonil (CXR6032) and fenpiclonil (CXR6069) RNA microarray experimental structure using triplicates of each sample.
  • Example 1 selection of the compounds of the invention based on specific properties
  • the following method details how the compounds of the invention were selected to arrive at those to be used for the treatment of diseases such as cancer.
  • the compounds of the invention were selected using the following method:
  • Step 1 A database was produced of some 182 compounds registered for use as agro- or industrial chemicals with available dossiers regarding bioactivity and toxicity collated from sources including the UK's Advisory Committee on Pesticides (ACP), the US Environmental Protection Agency (EPA), the European Food Safety Authority (EFSA), the US Department of Health & Human Services' Agency for Toxic Substances and Disease Registry (ATSDR), Canada's Bureau of Chemical Safety and the International Programme on Chemical Safety (IPCS).
  • ACP Advisory Committee on Pesticides
  • EPA US Environmental Protection Agency
  • EFSA European Food Safety Authority
  • ATSDR US Department of Health & Human Services' Agency for Toxic Substances and Disease Registry
  • IPCS International Programme on Chemical Safety
  • Table 13 details the 182 compounds contained in the database.
  • Step 2 A panel of 144 chemically diverse compounds were selected as being readily available from the database produced in (1).
  • Table 14 details the 144 chemically diverse compounds.
  • Step 3 The compounds from (2) were tested against 15 human tumour cell lines for cytotoxicity. Effects on cell survival at 72 hours and at 7 days at a single compound concentration (100 ⁇ ) were measured. Tables 11 and 12 show cell proliferation data for the 144 compounds at 72 hours and 7 day exposure, respectively. The data is expressed as a percentage of untreated control cells, so 100 % means that there are the same number of cells as the control, less than 100 % indicates there are fewer cells than the control and greather than 100 % indicates that more cells than the control are present. Cell assay methods
  • Test compounds were dissolved in DMSO. Cells were seeded at a density of 500 cells per well in 75 ⁇ of DMEM (10% FBS/2 mM Glutamine) media for 7 day incubation in 96- well plates or 5000 cells per well for 72 hrs incubations. The following day, 25 ⁇ of test compounds, diluted 1:25 in DMEM (10% FBS), were added to the wells to give a final compound concentrations of 100 ⁇ . Duplicate plates were assembled and then incubated for either 72 hrs or 7 days at 37°C/5% C0 2 . No media/drug replacement during the incubation period was undertaken.
  • Cell number was estimated using WST-1 reagent from Roche Applied Bioscience according to the manufacturer's instructions. Briefly 10 ⁇ of reagent was added to 100 ⁇ of media and incubated until colour developed. The plates were read at wavelength 450nm on an appropriate absorbance plate reader.
  • Step 4 The cytotoxicity data from (3) was used to rank compounds with respect to (a) global cytotoxicity; (b) selective cytotoxicity; and (c) published literature (i.e. a lack of published literature linking the compounds with cytotoxicity).
  • Step 5 Panel of 48 compounds chosen as the top-ranked compounds using the results of (4).
  • Step 6 Compounds selected under (5) were tested for cytotoxicity with compound dose response (see table 10) and the published toxicology data reviewed for (a) bioavailability (see table 3); (b) rat LD50 (see table 5); (c) Therapeutic Index (NOEL (No Observable Effects Limit): IC50 ratio) (table 2).
  • Step 7 Panel from (6) screened for effects on protein kinase activities. The results are shown in Table 8.
  • Step 8 Compounds selected for further development on basis of meeting at least criteria (1) and (2) (but preferably more) of the 7 criteria listed below:
  • Step 9 (optional) Kinase inhibition (such as PIM-Kinase) by dose response results can be used to select specific compounds for further development (see Examples 3 and 4)
  • Step 10 Compounds selected under (8) and (9) will be tested further in xenograft studies (see Example 6).
  • Toxicological testing is a significant barrier in the pre-clinical development of pharmaceuticals, both in terms of cost and risk. Selection of molecules already tested for toxicity is therefore an attractive alternative.
  • Favourable Therapeutic Index - NOELICsn Ratio must be greater than 5
  • Table 2 shows which of a selection of 48 compounds (as selected according to step 5 of the screening method) have an IC50/NOEL ratio of greater than five.
  • tau-fluvalinate 1 2.0 14.00 0.1
  • Oral bioavailability preferred to be greater than 20% Oral bioavailability is an attractive property for pharmaceutical compounds. Oral dosing will mean patients do not have to travel to the hospital situation regularly during their treatment. This will not only save considerable resources at the treatment centre but also reduce patient comfort by reducing travel and in-patient time. Oral bioavailability is calculated by measuring the plasma concentration of the compound over time after administering the drug both orally (po) and intravenously (IV). The area under the curve (AUC) is measured for both modes of administration and is corrected for different dose levels for the different routes. The following formula is used for calculating oral bioavailability (F):
  • Table 3 shows which of a selection of compounds has an oral bioavailability of >20%:
  • the LPTM be greater than 500 mq/ka
  • the selection approach described is to select compounds which have undergone extensive toxicological testing and have been deemed of low toxicity and safe for release to the environment as either agrochemicals or for use in other industries.
  • LD 50 Lethal Dose 50
  • MTD Maximum Tolerated Dose
  • the toxicity of molecules can be classified according to the table below.
  • the compounds selected are of no greater than "low toxicity", with oral LD50 values in the rat no lower than 500 mg kg.
  • Table 4 details the different measures of toxicity of compounds.
  • Table 5 details which of a selection of compounds have an oral LD50 in the rat of over 500mg/kg.
  • Ethoprophos 33 Preferred that in vivo half-life greater than 4 hours
  • Compounds are selected from a library partly on the basis of in vivo half-life measured during development of the compounds as agro-industrial chemicals.
  • In vivo half-life is a measure of the rate at which a compound is excreted or metabolised.
  • the change in plasma concentration of a compound following dosing is described by the equation:
  • tumour types may be best targeted with the molecule.
  • Some of this information can be learned from testing the compound for effects on cellular proliferation on a panel of tumour ceH lines.
  • An alternative approach is to test the molecules for activity, inhibitory or activatory, on to use in vitro assays of molecular targets such as protein kinases, GPCRs, enzymes or other cellular machinery. It is preferable that the compounds have a known mode of action.
  • One aspect of this could be inhibitory activity of a compound against a member(s) of a panel of protein kinases known to be implicated in cancer biology.
  • Such a panel could be comprised of the protein kinases shown in table 7: Table 7: A panel of kinases known to be implicated in cancer biology
  • MAPK2/ERK2 assay MAPK/ERK2 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na 3 V0 4 , 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against MBP in a final volume of 25.5 ⁇ in 25 mM Tris pH 7.5, 0.1 mM EGTA, 0.33 mg/ml MBP, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ](500 -1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. MKK1 assay
  • inactive MAPK (0.06 mg/ml) is activated by MKK1 (diluted in 25 mM Tris, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 0.01% Brij35, 1 mg/ml BSA) in 25.5 ⁇ containing 25 mM Tris, 0.1 mM EGTA, 0.01% Brij35, 10 mM magnesium acetate and 0.005 mM ATP.
  • PKCa (5-20 mU diluted in 20 mM Hepes pH 7.4, 0.03% Triton X-100) is assayed against Histone H1 in the presence of PtdSerine and DAG (0.1 mg/ml. and 10 pg/ml) and 0.1 mM CaCI2.
  • the assay is carried out in a final volume of 25.5 ⁇ containing 20 mM Hepes pH 7.4, 0.03% Triton X-100, 0.1 mg/ml Histone H1 , 10 mM magnesium acetate and 0.02 ⁇ ) ⁇ [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature.
  • Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • PDK1 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQ-EMFRDFDYIADWC) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 100 ⁇ substrate peptide, 10mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • PDKtide KTFCGTPEYLAPEVRREPRILSEEEQ-EMFRDFDYIADWC
  • APH-PKBa-S473D (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ - mercaptoethanol, 1 mg/ml BSA) is assayed against a modified Crosstide peptide GRPRTSSFAEGKK in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ - mercaptoethanol, 30 ⁇ substrate peptide, 10 mM magnesium acetate and 0.005 mM
  • SGK (5-20mU diluted in 20 mM MOPS pH 7.5, 1mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against a modified Crosstide peptide GRPRTSSFAEGKK in a final volume of 25.5 ⁇ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 30 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 p -y-ATPJ (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • S6K1/P70 S6K (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against substrate peptide (KKRNRTLTV) in a final volume of 25.5 ⁇ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 0.1 mM substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 p -v- ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • ROCK-II (ROKa) (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ - mercaptoethanol, 1 mg/ml BSA) is assayed against Long S6 substrate peptide (KEAKEKRQEQIAKRRRLSSLRASTSKSGGSQK) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 30 ⁇ Long S6 substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • CHK1 assay CHK1 (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.1% ⁇ -mercaptoethanol, 0.01% Brij-35, 5% glycerol, 1 mg/ml BSA) is assayed against CHKtide substrate peptide (KKKVSRSGLYRSPSMPENLNRPR) in a final volume of 25.5 ⁇ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 ⁇ CHKtide, 10 mM magnesium acetate and 0.02 mM [33 p - ⁇ - ⁇ ](50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • CHKtide substrate peptide KKKVSRSGLYRSPSMPENLNRPR
  • LCK assay LCK (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% B-mercaptoethanol, 1 mg/ml BSA) is assayed against Cdc2 peptide (KVEKIGEGTYGWYK) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na3Vo4, Cdc2 peptide (0.25 mM), 10 mM magnesium acetate and 0.05mM [33 ⁇ - ⁇ - ⁇ ](500-1000 cpm/pmole) and incubated for 15 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • Cdc2 peptide KVEKIGEGTYGWYK
  • CDK2/cyclin A (5-20 mU diluted in 50 mM Hepes pH 7.5, 1 mM DTT, 0.02% Brij35, 100 mM NaCI) is assayed against Histone H1 in a final volume of 25.5 ⁇ containing 50 mM Hepes pH7.5, 1 mM DTT, 0.02% Brij35, 100 mM NaCI, Histone H1 (1 mg/ml), 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ](500-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. NEK2a assay
  • NEK2a 5-20mU of NEK2a (diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1mg ml BSA, 0.1%, ⁇ - mercaptoethanol) is assayed against NEK2a peptide (RFRRSRRMI) in a final volume of 25.5 ⁇ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.01% Brij, 0.1%, ⁇ - mercaptoethanol, 300 ⁇ NEK2a peptide, 10 mM magnesium acetate and 0.05 mM [33 p - ⁇ - ⁇ ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid.
  • MAPKAP-K1b (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% ⁇ -mercaptoethanol, 1mg/ml BSA) is assayed against substrate peptide (KKLNRTLSVA) in a final volume of 25.51 containing 50 mM Na- ⁇ -glycerophosphate (pH 7.5), 0.5 mM EDTA, 30 ⁇ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • IKKb 5-20mU of IKKb (diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1mg/ml BSA, 0.1% ⁇ - mercaptoethanol) is assayed against substrate peptide (LDDRHDSGLDSMKDEEY) in a final volume of 25.5 ⁇ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.1%, ⁇ mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33 ⁇ - ⁇ - ⁇ ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid.
  • Aurora B (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ - mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (LRRLSLGLRRLSLGLRRLSLGLRRLSLG) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • LRRLSLGLRRLSLGLRRLSLGLRRLSLG substrate peptide
  • CHK2 (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.1% ⁇ -mercaptoethanol, 0.01% Brij-35, 5% glycerol, 1 mg/ml BSA) is assayed against CHKtide substrate peptide (KKKVSRSGLYRSPSMPENLNRPR) in a final volume of 25.5 ⁇ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 ⁇ CHKtide, 10 mM magnesium acetate and 0.02 mM [33 p - ⁇ - ⁇ ](50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • CHKtide substrate peptide KKKVSRSGLYRSPSMPENLNRPR
  • Src (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (KVEKIGEGTYGWYK) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • KVEKIGEGTYGWYK substrate peptide
  • PLK1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA, 100 ⁇ Vanadate) is assayed against a substrate peptide (ISDELMDATFADQEAKKK) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 10 ⁇ Vanadate, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid
  • PIM1 assay PIM1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (RSRHSSYPAGT) in a final volume of 25.5 pi containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. PIM3 assay
  • PIM3 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (RSRHSSYPAGT) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • FGF-R1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • substrate peptide Poly Glut Tyr
  • EPH-A2 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.05 mM [33 p - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • IGF-1R (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKSPGEYVNIEFG) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. VEG-FR assay
  • VEG-FR (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKSPGEYVNIEFG) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • TBK1 (DU12569) (5-20mll diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKKERLLDDRHDSGLDSMKDEE) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • IKKepsilon (DU14231)(diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1 mg/ml BSA) is assayed against MBP in a final volume of 25.5 ⁇ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.33mg/ml MBP, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid HER4 assay
  • HER4 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against Poly Glut Tyr in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 1mg/ml Poly Glut Tyr, 10 mM magnesium acetate and 0.005mM [33 p -y-ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • Aurora A (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ - mercaptoethanol, 1mg/ml BSA) is assayed against LRRLSLGLRRLSLGLRRLSLGLRRLSLG in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 0.3mM LRRLSLGLRRLSLGLRRLSLGLRRLSLG, 10 mM magnesium acetate and 0.005mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • JAK2 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.05% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQ- EMFRDFDYIADWC) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.05% ⁇ -mercaptoethanol, 100 ⁇ substrate peptide, 10mM magnesium acetate and 0.005 mM [33 p -y-ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • PDKtide KTFCGTPEYLAPEVRREPRILSEEEQ- EMFRDFDYIAD
  • TrkA assay TrkA (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 10mM MnCI, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped- by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.-
  • NEK2a NEK2a
  • TBK1 DU12569
  • IKKepsilon DU14231
  • Aurora B VEGFR
  • PKCa PKCa
  • PKBAph SGK, KK1, Aurora B, flocoumafen S6K1, CHK1 VEGFR, PKCa, SRC, TRKA, RSK2,
  • Compounds must exhibit broad activity against a panel of cell lines from tumour types of varying origins. Effects on cellular proliferation were measured against 14 cell lines representing carcinomas of the lung, prostate, breast, liver, colon, pancreas as well as sarcomas. Cells were treated with 8 concentrations of compound 300, 100, 30, 10, 3, 1 , 0.3, 0.1, 0.03 and 0.01 ⁇ and cell number estimated at each concentration. Cell number was calculated as a percentage survival relative to untreated cells. EC 50 was calculated by fitting the data to the 4-parameter logistic model:
  • Table 9 details the transformed cell lines.
  • Table 9 Table 10 shows the cytotoxicity data ( ⁇ EC50s) for selected compounds against 14 tumour cell lines over a 7 day exposure
  • Table 15 shows a summary of the kinase screening data for fludioxonil and fenpiclonil. Results are represented as % inhibition relative to a positive control. Negative values represent compounds which activate kinase activity. Where (-) is shown there was no activity of the compound. Inhibition of the kinases SGK, ROCKII, PIM1 , TRKA, RSK2, PIM3 and FGFR1 was common to both fludioxonil and fenpiclonil.
  • Table15 Summary table of kinase inhibition for selected compounds
  • Example 3 Inhibition ofPIM Kinase activity PIM kinase inhibition has been investigated for each of PIM-1 , PIM-2 and PIM-3 kinase molecules.
  • the PIM-1 assay is performed using the Upstate IC 50 Profiler ExpressTM service.
  • human recombinant PIM-1 (5-1 OmU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 100 ⁇ KKRNRTLTV, 10mM MgAcetate and [v- 33 P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required).
  • the reaction is stopped by the addition of 3% phosphoric acid solution.
  • 10 ⁇ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
  • the PIM2 assay is performed using the Upstate IC 50 Profiler ExpressTM service.
  • human recombinant PIM-2 (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 300 ⁇ RSRHSSYPAGT, 10 mM MgAcetate and [?- 33 P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required).
  • the reaction is stopped by the addition of 3% phosphoric acid solution. 10 ⁇ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in menthanol prior to drying and scintillation counting.
  • the PIM-3 assay is performed using the Upstate IC 50 Profiler ExpressTM service.
  • human recombinant PIM-3 (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 300 ⁇ RSRHSSYPAGT, 10 mM MgAcetate and p- ⁇ P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required).
  • the reaction is stopped by the addition of 3% phosphoric acid solution.
  • 10 ⁇ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in menthanol prior to drying and scintillation counting.
  • FIG. 1a and 1b shows the inhibition of PI 1, PIM2 and PIM3 for fludioxonil and fenpiclonil respectively.
  • the plots show % inhibition in an in vitro kinase assay.
  • Table 16 details the IC50 and EC50 data for each compound against the PIM kinases.
  • K562 cells express high levels of PIM1 protein. It is known that use of a PIM inhibitor can result in downregulation of PIM1 protein as shown in Swords et al. (SGI-1776: A novel PIM kinase inhibitor with potent preclinical activity against Acute Myeloid Leukemia (AML), Swords, R et al, Abstracts of the AACR Annual Meeting, 2009, April 18-22, Denver, CO, USA, Abstract umber 3743).
  • proteins were transferred to a PVDF membrane and incubated for 1 hour at room temperature in blocking solution. Then the membranes were incubated overnight at 4°C in antibody solution containing anti-PIM1 (12H8) antibody (Santa Cruz Biotechnology) or containing anti-cleaved PARP antibody (Abeam pic). After washing, the membrans were incubated for 1 hour at room temperature in solution containing appropriate dilution of HRP-conjugated secondary antibody (GE Healthcare, UK).
  • ECL plus (GE Healthcare, UK) was used to detect PI 1 and cleaved PARP protein levels.
  • ROCK-II (ROKa) (2.6 - 10.5 ng active Rock-ll diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against Long S6 substrate peptide (KEAKEKRQEQIAKRRRLSSLRASTSKSGGSQK) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 30 ⁇ Long S6 substrate peptide, 10 mM magnesium acetate and 0.1 mM ATP (spiked with sufficient [33 p -v- ATP] to give approximately 500-800 cpm/pmole) and incubated for 10 min at 30°C. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then 10 ⁇ harvested onto P30 filter-mat and washed 3 times with 75 mM orthophosphoric acid, and once with methanol.
  • ROCK-I (ROK) (72.7 - 200.5ng active Rock-I diluted in 20mM MOPS/NaOH pH 7.0, 1 mM EDTA, 0.01% Brij-35, 5% Glycerol, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against 30 ⁇ Long S6 substrate peptide (KEAKEKRQEQIAKRRRLSSLRASTSKSGGSQK) in a final volume of 25 ⁇ containing 8mM MOPS/NaOH pH 7.0, 0.2 mM EDTA, 30 ⁇ Long S6 substrate peptide, 10 mM magnesium acetate and 0.1 mM ATP (spiked with sufficient [33 p - ⁇ - ATP] to give approximately 500-800 cpm/pmole) and incubated for 10 min at 30°C. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then 10 ⁇ harvested onto P30 filter-mat and washed 3
  • Figure 3 show the increase in % inhibition of Rock I and Rock II with increasing does of fludioxonil or fenpiclonil.
  • Table 17 details the IC50 and EC50 data for ROCK-I and ROCK-II using fludioxonil, fenpiclonil and pyrollnitrin. Table 17
  • Table 18 shows the 7 day IC50 ( ⁇ ) of fludioxonil and fenpiclonal on a selection of cancer cell models.
  • Test compounds were dissolved in DMSO and dilution series performed. Cells were seeded at a density of 500 cells per well in 75 ⁇ of DMEM (10% FBS/2 mM
  • Glutamine media for 7 day incubation in 96-well plates. The following day, 25 ⁇ of test compounds, diluted 1 :25 in DMEM (10% FBS), were added to the wells to give final compound concentrations as detailed above. Duplicate plates were assembled and then incubated for 7 days at 37°C/5% C0 2 . No media/drug replacement during the 7 day period was undertaken.
  • Cell number was estimated using WST-1 reagent from Roche Applied Bioscience according to the manufacturer's instructions. Briefly 10 ⁇ of reagent was added to 100 ⁇ of media and incubated until colour developed. The plates were read at wavelength 450nm on an appropriate absorbance plate reader.
  • the aim of this study was to assess the efficacy of fludioxonil and fenpiclonil in vivo in nude mouse xenografts using mice harbouring tumours derived from human prostate (PC3).
  • PC3 cell line was grown in culture and implanted into female nu-nu mice; dosing was initiated 72 hours later and tumour growth was monitored by calliper measurements.
  • Mice harbouring PC3 tumour cells were dosed with ethanol/PEG200/water only or CXR6032 (Fludioxonil), or CXR6069 (Fenpiclonil). Compounds were to be administered daily for 28 days by oral gavage.
  • PC3 ECACC catalogue number 90112714
  • mice Animals Adult (6 - 8 weeks) female, athymic nude (nu/nu) mice were obtained from Charles River, UK.
  • mice On arrival in the animal unit the mice were housed, up to 8 per cage, on sterile sawdust in sterile, solid-bottom, polypropylene cages.
  • the cages were individually vented units attached to a Techniplast Slimline Air Handling Unit. This unit maintains 70-80 air changes per cage, per hour, through HEPA air filters. Bedding was changed once weekly in a laminar flow unit. The temperature was maintained within a target range of 19-23° C and relative humidity of the IVC within a range of 40-70%. Twelve-hour periods of light were cycled with twelve-hour periods of darkness.
  • Sterile RM1 diet (Special Diet Services Ltd., Stepfield, Witham, Essex, UK) was used. The animal unit held the specification of the diet. Deionised water was autoclaved prior to use and changed at least once a week. Mice were allowed water and diet ad libitum and were acclimatised for at least 5 days prior to the study start.
  • mice have specific pathogen free (SPF)-status and the housing and changing system assured that the SPF-status was preserved during the study. Trained personnel under supervision handled the mice.
  • SPF pathogen free
  • mice were randomly allocated to groups on arrival.
  • the mice were numbered appropriately and weighed prior to the start of the experiment.
  • the first mouse assigned to a cage was individually identified by tail tattooing with the lowest number for that cage; the second mouse was assigned the second (lowest) number and so on.
  • An experimental card was placed on each cage and showed the project licence code, test group, study number, sex and individual numbers of the mice within, and identified the Home Office Licensee. In addition, these cards were colour coded to correlate with the coding for the group.
  • PC3 cells were cultured in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% heat inactivated foetal calf serum (FCS) supplemented with 2mM L-glutamine, penicillin (50 lU/ml) and streptomycin (50 ⁇ 9/ ⁇ ). Cultures were incubated in a humidified incubator at 37°C, 5% C0 2 , until sufficient cells were available to implant the mice. Cells were harvested, pooled, centrifuged, and re-suspended in cold medium. This wwe mixed with an equal volume of cold Matrigel, so that the tumour cell injection solution was a 50:50 mixture of tumour cells/medium and Matrigel.
  • DMEM Dulbecco's Modified Eagle's Medium
  • FCS heat inactivated foetal calf serum
  • Tumour cells were injected in a volume of 100 ⁇ in a single flank only.
  • PC3 cells were injected at a density of 2.5 x 10 6 cells per flank.
  • the cell/Matrigel suspension was aliquoted into pre-chilled Eppendorf tubes and kept on ice prior to injection. The period between preparation of the cell/Matrigel suspension and injection of tumour cells did not exceed two hours.
  • mice were administered Test Compounds 72 hours following implantation of tumour cells. Mice harbouring PC3 tumour cells were dosed with CXR6032 (Fludioxonil), or CXR6069 (Fenpiclonil) in an ethanol/PEG200/water vehicle. Refer to Table 1 for the experimental design.
  • Compounds were administered daily for 28 days by oral gavage in a dose volume of 10ml/kg.
  • mice Prior to the start of the study, all mice were observed to ensure that they were physically normal and that they exhibited normal activity. Only normal mice were allocated to the study. Following cell inoculation, each mouse was observed twice weekly and a general assessment of condition recorded in the study diary. Animals were terminated if tumours became ulcerated or if the Home Office Project License moderate severity limit was exceeded.
  • Tumour growth was measured twice weekly for the duration of the study following cell implantation once tumours became palpable. Tumour diameters were measured at four different sites - two lengths and two widths - using a digital slide gauge. This was increased to three lengths and three widths if the tumour was an irregular shape.
  • Tumour volumes were calculated using the formula: 4/ 3 ⁇
  • tumour was removed and the weights and gross morphology recorded. • The tumour was cut in half and one half placed in 10 % neutral buffered formalin (NBF) then prepared for paraffin embedding for possible future histochemical analysis.
  • NBF neutral buffered formalin
  • cryovial 1 was labeled microarray and cryovial 2 was labeled MS. These samples were stored at approximately - 80°C for possible future analysis.
  • Cryovial 1 was labeled 'Microarray' and cryovial 2 was labeled 'MS'. These samples were stored at approximately - 80°C for possible future analysis.
  • venous blood samples were mixed on a roller for 10 minutes. Red blood cells were removed by centrifugation at 2,000 - 3000 rpm for 10 minutes at 8 - 10 °C. The supernatant (plasma) was transferred to a second tube and stored at approximately -70 °C until required for analysis.
  • Tumour and liver samples were placed immediately into NBF for 36-45 hours. Tissues were processed according to internal standard operating procedures. The tissues were embedded in paraffin wax for possible future histochemical analysis.
  • Figure 4 (a) shows the bodyweight over the duration of study in PC3 xenograft mice treated with fludioxonil (CXR6032), fenpiclonil (CXR6069) and vehicle control.
  • Figure 4 (b) and (c) shows the terminal body and liver weights respectively from the same study.
  • Figure 5 shows (a) the Alanine transaminase (ALT), Aspartate transaminase (AST) and Alkaline phosphatase (ALP) activity levels and (c) tumour weight from the same study.
  • Figure 5 (b) shows the tumour volume over time.
  • Example 7 Metabolism and pharmacokinetic properties of fludioxonil and fenpiclonil
  • Microsomal incubation mixtures (500pL) were prepared in HEPES buffer (50mM HEPES, 15mM MgCI 2 , 0.1 mM EDTA, pH 7.6) containing a NADPH regenerating system (1 mM NADPH, 10mM glucose-6-phosphate 1 IU glucose-6-phosphate dehydrogenase), pooled human liver microsomes (0.5mg protein), and 5pL of 7 concentrations and positive control inhibitors (see Table 21).
  • HEPES buffer 50mM HEPES, 15mM MgCI 2 , 0.1 mM EDTA, pH 7.6
  • NADPH regenerating system (1 mM NADPH, 10mM glucose-6-phosphate 1 IU glucose-6-phosphate dehydrogenase
  • pooled human liver microsomes 0.5mg protein
  • 5pL 7 concentrations and positive control inhibitors
  • CYP1A2 activity was monitored by phenacetin O-deethylation, CYP2C9 by tolbutamide hydroxylation, CYP2C19 by omeprazole hydroxylation, CYP2D6 by bufuralol l'-hydroxylation, and CYP3A4 by midazolam hydroxylation.
  • Table 21 Positive inhibitor controls and Test Item concentrations
  • test Items or positive control inhibitors After addition of the Test Items or positive control inhibitors, the samples were pre- incubated at 37°C for approximately 5 minutes. Subsequently, the NADPH regenerating system was added to the reaction mixture and incubated for 20 minutes at 37°C. Reactions were terminated by the addition of 250 ⁇ _ of methanol containing 5 ⁇ dextrorphan as an internal standard.
  • the aim of this study was to examine the pharmacokinetics of fenpiclonil and fludioxonil administered by oral dosing to nude mice.
  • sampling took place over a 1 day period after a single oral dose of compound to obtain a 24hr post dose profile.
  • the mice were repeat dosed, daily for 4 days, prior to PK sampling.
  • mice Sufficient adult (6 - 8 weeks) female, athymic nude (nu/nu) mice were obtained from Charles River, UK. The mice were housed, up to 7 per cage, on sterile sawdust in sterile, solid-bottom, polypropylene cages. The cages were individually vented units attached to a Techniplast Slimline Air Handling Unit. This unit maintains 70-80 air changes per cage, per hour, through HEPA air filters. Bedding was changed once weekly in a laminar flow unit. The temperature was maintained within a target range of 19-23° C and relative humidity of the IVC within a range of 40-70%. Twelve-hour periods of light were cycled with twelve-hour periods of darkness.
  • mice were fed with sterile RM1 diet (Special Diet Services Ltd., Stepfield, Witham, Essex, UK). Deionised water was autoclaved prior to use and changed at least once a week. Mice were allowed water and diet ad libitum and were acclimatised for at least 5 days prior to the study start.
  • mice were randomly allocated to groups on arrival.
  • the mice were numbered appropriately and weighed prior to the start of the experiment.
  • the first mouse assigned to a cage was individually identified by tail tattooing with the lowest number for that cage; the second mouse will be assigned the second (lowest) number and so on.
  • An experimental card was placed on each cage and showed the project licence code, test group, study number, sex and individual numbers of the mice within, ans identified the Home Office Licensee. In addition, these cards were colour coded to correlate with the coding for the group.
  • Oral dosing solutions of the Test Items were prepared at concentrations of 12mg/mL (for Fludioxonil and Fenpiclonil) in the dosing vehicle ethanol:PEG-200:water in the ratio 3:5:2 v/v.
  • the Test Items were orally administered at a dose of 120mg/kg (for Fludioxonil and Fenpiclonil) all with a dosing volume of lOmlJkg. The bodyweight of each mouse was recorded immediately before each daily dose.
  • Oral pharmacokinetic samples from the 4 daily repeat dosed animals were taken pre-dose (on day 4) and at 0.5, 1, 1.5, 2, 3, 4, 6 and 24 hours after administration of the final daily dose.
  • Oral pharmacokinetic samples from the single dosed animals were taken at 0.5, 1 , 1.5, 2, 3, 4, 6 and 24 hours after administration of the dose.
  • mice in the repeat dose groups were bled at approximately 0.5, 1 , 1.5, 2, 3, 4, 6 and 24 hours.
  • Whole blood (0.020ml_) was taken from the tail vein of each animal and added to plastic sample vials containing milli Q water (0.020ml_) at each of the 9 timepoints. The whole blood:water samples were collected onto dry ice and stored at approximately -70°C prior to analysis.
  • mice were bled at approximately 0.5, 1 , 1.5, 2, 3, 4, 6 and 24 hours.
  • Whole blood (0.020mL) was taken from the tail vein of each animal and added to plastic sample vials containing milli Q water (0.020ml_) at each of the 8 timepoints.
  • the whole blood:water samples were collected onto dry ice and stored at approximately -70°C prior to analysis.
  • mice were transferred to the post mortem room or designated Laboratory.
  • the animals were killed by exposure to a rising concentration of C0 2 .
  • Blood was harvested by cardiac puncture and transferred to heparinised tubes for plasma preparation.
  • venous blood samples were mixed on a roller for 10 minutes. Red blood cells were removed by centrifugation at 2,000 - 3000 rpm for 10 minutes at 8 - 10 °C. The supernatant (plasma) was transferred to a second tube and stored at approximately -70 °C until required for analysis.
  • Blood concentration time data was analysed, where possible, using Pharsight WinNonLin software Version 5.2, for appropriate pharmacokinetic parameters such as half-life, bioavailability, Cmax, Tmax, AUC and clearance.
  • Table 24 Plasma concentrations of fludioxonil (ng/ml) per animal for repeat dose and single dose groups
  • Figure 6 shows graphically the changes in fludioxonil concentration over time for both single dose and for four daily repeat doses.
  • Table 27 Plasma concentrations of fenpiclonil (ng/ml) per animal for repeat dose and single dose groups
  • Figure 7 shows graphically the changes in fenpiclonil concentration over time for both single dose and for four daily repeat doses.
  • Table 30 details the metabolism and pharmacokinetic properties of fludioxonil and fenpiclonil.
  • PC3 had been chosen for this experiment because in xenograft studies, PC3 tumours showed a delayed growth response to CXR6032 and CXR6069.
  • DMEM Dulbecco's Modified Eagle's Medium
  • FCS heat inactivated foetal calf serum
  • tissue culture medium was changed as follows:
  • tissue culture medium in all 6 wells on plate 1 was replaced with tissue culture medium containing 0.1% DMSO.
  • CXR6032 and CXR6069 at the EC 20 in tissue culture medium was added to 2 wells.
  • CXR6032 and CXR6069 at the EC 50 was added to the remaining 2 wells on each plate.
  • the cells were cultured at 37°C and 5% C0 2 for a further 24 hours.
  • Table 31 IC 20 and IC 50 concentrations for test items
  • RNA microarray analysis involved labelled (one colour) RNA from triplicate samples of PC3 cell line treated vehicle, CXR6032, CXR6069 at EC 20 and CXR6032, CXR6069 at EC 5 o as shown in figure 14.
  • RNA integrity was checked using the Agilent bioanalyser and the RNA nano lab chip kit according to the CXR method sheet 'Set-up and Running of Nanochip Assay for RNA Quality Control'.
  • RNA was labelled prior to microarray hybridisation using the Quick Amp Labelling Kit - Two Color (Agilent* 5190-0442), according to the CXR method sheet entitled "Transcriptional Profiling using Standard Agilent 1 Colour Protocol” v2.
  • Table 32 Details signature lists generated. Denominator (D) Test (T) Number of lists
  • IPA Ingenuity pathways analysis
  • Table 33 Data table showing the number of 'signature' genes changes for each treatment
  • Fold change data from the signature lists was overlaid on two PIM-associated pathways: granulocyte-macrophage colony stimulating factor signalling pathway (GM-CSF), Figures 8 and 9; Table 34, and acute myeloid leukemia signalling pathway (AML), Figures 10 and 11 ; Table 35. Fold change data was also overlaid on the RhoA signalling pathway, Figures 12 and 13; Table 36.
  • GM-CSF granulocyte-macrophage colony stimulating factor signalling pathway
  • AML acute myeloid leukemia signalling pathway
  • ROCK Rho associated kinase
  • ARP2/3 actin-related protein complex
  • RhoGAP Rho guanine nucleotide exchange factor GEF 12
  • PLD1 phospholipase D1
  • it serine/threonine kinase 21
  • MLC myosin light chain
  • FEN Treatment with FEN led to down-regulation of lysophosphatidic acid receptor (LPAR), focal adhesion kinase (FAK), actin binding protein (ANLN) and ezrin (EZR).
  • LPAR lysophosphatidic acid receptor
  • FK focal adhesion kinase
  • ANK actin binding protein
  • ezrin ezrin
  • FLU and FEN both caused the down regulation of multiple genes in the RhoA signalling pathway. All of the genes down-regulated are down-stream effectors of RhoA signalling, consistent with inhibition of Rho associated kinase.
  • microarray data suggests that FLU and FEN have a similar mode of action.
  • Table 34 Data table showing gene changes in GM-CSF signalling pathway in CXR6032 and CXR6069 EC50 and EC20 signature lists.
  • Table 35 Data table showing gene changes in acute myeloid leukaemia signalling pathway in CXR6032 and CXR6069 EC50 and EC20 signature lists.
  • Table 36 Data table showing gene changes in RhoA signalling pathway in CXR6032 and CXR6069 EC50 and EC20 signature lists.
  • Example 9 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 directly to the required site. For example, Vitrasert releases ganciclovir directly into the eye to treat CMV retinitis. The direct application of this toxic agent to the site of disease 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 permeability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drug delivery.
  • Compounds of the invention 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.
  • ReGel injectable system that is thermosensitive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into known, safe, biodegradable polymers. The active drug is delivered over time as the biopolymers dissolve.
  • Trojan peptides 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 compounds to the cytoplasm and nucleus, and may be non-cell type specific and highly efficient (Derossi et al., 1998, Trends Cell Biol., 8, 84-87).
  • the pharmaceutical formulation of the present invention is a unit dosage containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of the active ingredient.
  • the compounds of the invention can be administered by any parenteral route, in the form of a pharmaceutical formulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form.
  • a parenteral route in the form of a pharmaceutical formulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form.
  • the compositions may be administered at varying doses.
  • the compounds of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical exipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the compounds of the invention can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They 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.
  • suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.
  • Formulations 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 requiring 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 kind previously described.
  • the compounds of the invention can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
  • the compounds of the invention can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • oral or parenteral administration of the compounds of the invention is the preferred 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 determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
  • 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 ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Preferred unit dosage formulations are those containing a daily dose or unit, daily sub- dose or an appropriate fraction thereof, of an active ingredient.
  • a preferred delivery system of the invention may comprise a hydrogel impregnated with a polypeptides, polynucleotides and antibodies of the invention, which is preferably carried on a tampon which can be inserted into the cervix and withdrawn once an appropriate cervical ripening or other desirable affect on the female reproductive system ⁇ ias been produced.
  • formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question.
  • compounds of the invention Whilst it is possible for compounds of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, 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.
  • 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 into a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals.
  • Example 10B Intramuscular injection
  • Tablets are prepared from the foregoing ingredients by wet granulation followed by compression.
  • Vanbreuseghem R De Vroey C. n-vitro testing of fungistatic activity of pyrrolnitrin on pathogenic toadstools
  • Farmaco Prat. 32(12),617-33, 1977.
  • Seidenari S Di Nardo A, Motolese A, Pincelli C. "Erythema multiforme associated with contact sensitization. Description of 6 clinical cases", G Ital Dermatol Venereol., 125(1- 2), 35-40, 1990.

Abstract

There is provided pyrrolnitrin derivatives of formula I for use in medicine, preferably for the treatment of cancer.

Description

Pyrrolnitrin derivatives
The present invention relates to the medical use of pyrrolnitrin derivatives and particularly their use in the treatment of cancer.
Background
Cancer (or neoplasm) covers a range of diseases in which a group of cells display uncontrolled growth (division beyond the normal limits), invasion (intrusion on and destruction of adjacent tissues), and sometimes metastasis (spread to other locations in the body via lymph or blood). Cancer affects people at all ages with the risk for most types increasing with age and caused about 13% of all human deaths in 2007. Deaths from cancer worldwide are projected to continue rising, with an estimated 12 million deaths in 2030. (WHO, February 2009)
Traditionally cancer treatment is based around surgery, chemotherapy and radiotherapy. However, the effectiveness of surgery is often limited by the propensity of cancers to invade adjacent tissue or to spread to distant sites by microscopic metastasis. The effectiveness of chemotherapy is often limited by toxicity to other tissues in the body. Radiation can also cause damage to normal tissue.
More recently, targeted therapy has had a significant impact in the treatment of some types of cancer. This constitutes the use of agents specific for the deregulated proteins of cancer cells. Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell.
The discovery and development of new agents for cancer treatment is both a very costly and very time consuming process. Traditionally agents are selected as having a promising activity against a particular biological target thought to be important in disease; however, little will be known about the safety, toxicity, pharmacokinetics and metabolism of this agent in humans. Therefore, it is traditionally necessary to assess all of these parameters prior to human clinical trials in order to be able to recommend a dose and schedule to be used the first time.
In addition, much drug development work is required to establish the physicochemical properties of a new agent, such as its chemical makeup, stability, solubility. The process l by which the chemical is made will be optimized and it will be further examined for its suitability to be made into capsules, tablets, aerosol, intramuscular injectable, subcuteneous injectable, or intravenous formulations. Many aspects of drug development are focused on satisfying the regulatory requirements of drug licensing authorities. These generally constitute a number of tests designed to determine the major toxicities of a novel compound prior to first use in man. It is a legal requirement that an assessment of major organ toxicity be performed (effects on the heart and lungs, brain, kidney, liver and digestive system), as well as effects on other parts of the body that might be affected by the drug (e.g. the skin if the new drug is to be delivered through the skin). While, increasingly, these tests can be made using in vitro methods (e.g. with isolated cells), many tests can only be made by using experimental animals, since it is only in an intact organism that the complex interplay of metabolism and drug exposure on toxicity can be examined.
The process of drug development does not stop once a novel agent begins human clinical trials. In addition to the tests required to move a novel drug into the clinic for the first time it is also important to ensure that long-term or chronic toxicities are determined, as well as effects on systems not previously monitored (fertility, reproduction, immune system, etc).
If a compound emerges from these tests with an acceptable toxicity and safety profile, and it can further be demonstrated to have the desired effect in clinical trials, then it can be submitted for marketing approval in the various countries where it will be sold. However, most new agents fail during drug development, often because they have some unacceptable toxicity.
Existing anti-cancer agents have considerable problems. A significant problem, which affects the majority of traditional anti-cancer agents, is the severe associated toxicities resulting in side effects such as depletion of the immune system, vomiting, malnutrition, hair loss, fatigue and anemia. Another significant problem is lack of efficacy. Current anti-cancer agents all have limited efficacy and cannot be guaranteed to cure a patient of disease. In some cancers a cure or halt of the disease can actually be very unlikely with the current anti-cancer drugs. Therefore, there remains a clear need to discover and develop new agents for use in the treatment of cancer which reduce the problems associated with current anti-cancer agents. To circumnavigate many of the developmental issues described above, the applicants have developed a method of screening compounds for potential pharmaceutical agents (preferably anti-cancer compounds) based on compounds for which bioactivity and toxicity data is already available. For example, such compounds might have previously been tested for use in unrelated fields, such as agrochemicals.
Here the applicants provide compounds that have been identified by this new screening method for use in medicine, and in particular, cancer treatment.
Pyrrolnitrin is an anti-fungal, secondary metabolite, produced by a range of bacteria. It was originally described as acting by inhibiting the electron transport chain (Lyr, H. 1977). Mutants defective in components of the HOG pathway are resistant to the synthetic derivatives of pyrrolnitrin, fludioxonil and fenpiclonil. These compounds normally induce cell death by activating the HOG pathway resulting in accumulation of intracellular glycerol, cell swelling and ultimately cell death.
There was an active research program on antifungal activity of pyrrolnitrin in- the early 1970's. Several papers arose describing the biosynthesis, metabolism, mechanism of action and effectiveness of pyrrolnitrin as an antibiotic (see Nishida 1965, Wente 1977, Yamada 1968, Macotela 1974, Vanbreuseghem 1977, Seidenari 1990, Uchida 1999, Tripathi 1969, Arima 1965).
However, ultimately the conclusion was drawn that the compound was less effective than rivals "amphotericin, hamcyin, 5-fluorocytosine and saramycetin". In addition, there were reports of cases of contact dermatitis induced by pyrrolnitrin reported from 1975 onwards (Meneghini 1975, Romaguera 1980, Valsecchi 1981 , Meneghini 1982, Balato 1983).
Pyrrolnitrin continues to be marketed in several countries for the treatment of fungal infections of the skin. For example, Pyrrolnitrin alone is marketed by Pharmacia in Italy as Micutrin and in combination with betametasone valerate as Beta Micutrin.
The applicant has now shown that pyrrolnitrin derivatives can be used in the treatment of cancer (such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases) and may also be useful in the treatment of vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma osteoporosis, reperfusion injury, asthma, fibrosis, anti-inflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, and other indications mediated by Rho-kinase, e.g., coronary heart disease. In addition, the compounds of the invention are useful to treat erectile dysfunction, i.e. erectile dysfunction mediated by Rho-kinase. Erectile dysfunction can be defined as an inability to obtain or sustain an erection adequate for intercourse.
Fludioxonil and fenpiclonil are two such derivatives that originated from a lead optimisation programme at Ciba Geigy, who used pyrrolnitrin as the starting structure for a developmental chemistry program, with the aim of identifying new fungicides. This is described in Nyfeler and Ackermann (1992) Phenylpyrroles, a new class of agricultural fungicide related to the natural product pyrrolnitrin and in Baker, D. et al, Synthesis and Chemistry of Agrochemicals III (pp 395 - 404) Washington, D.C. American Chemical Society. Fludioxonil and fenpiclonil were further developed by Ciba Geigy and were initially registered as agricultural fungicides in 1991 and 1988 respectively. They are still used commercially for this purpose today.
Fenpiclonil was first registration as agricultural fungicide in 1988, in Switzerland. Commercial products include BERET and GALBAS sold by Syngenta.
Fludioxonil was first registration as agricultural fungicide in 1991, in France. Commercial products include SAPHIRE, CELEST and MAXIM sold by Syngenta. These compounds are not known or sold for the treatment of any non-agricultural use.
It has now been found that derivatives of pyrrolnitrin may be useful in medicine, particularly in the treatment of cancer (such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases) and may also be useful in the treatment of vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma osteoporosis, reperfusion injury, asthma, fibrosis, antiinflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, and other indications mediated by Rho-kinase, e.g., coronary heart disease. In addition, the compounds of the invention are useful to treat erectile dysfunction, i.e. erectile dysfunction mediated by Rho-kinase. Erectile dysfunction can be defined as an inability to obtain or sustain an erection adequate for intercourse.
This invention is not intended to cover pyrrolnitrin itself, but only derivatives of pyrrolnitrin as described in the following aspects of the invention.
In a first aspect of the invention there is provided a pyrrolnitrin derivative of formula I or a salt or ester thereof for use in medicine. Formula I:
Figure imgf000007_0001
A = aromatic or heteroaromatic substituted with 1-3 N in the ring
B = aromatic or a partially or fully reduced five or six membered ring.
Either A or B rings can be optionally benzo fused. Q = bond, C1-4 alky, O, -(CH2)mO-(CH2)n- (m and n = 1, 2), S, -(CH2)mS-(CH2)n- (m and n = 1 , 2), NR (R = H or C1-4 alkyl), or -(pH2)mNR-(CH2)n- (m and n = 1 , 2; R = H or C1-4 alkyl) -
Y = (CX)2, CX2, NH, NR (R = H or C1-4 alkyl), S, or O. X = one or more substituents on ring A selected from: H, Halo (from F, CI, Br, I), C1-6 alkyl (linear and branched), C1-6 alkoxy, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyi (halo is 1-3 atoms from F, CI, Br, I), C1-4 haloalkoxy (halo is 1-3 atoms from F, CI, Br, I), OH, SR (R = H or C1-4 alkyl), NR2 (R = H or C1-4 alkyl), NRS02R (R = H or C1-4 alkyl), NRCOR (R = H or C1-4 alkyl), N02, CN, C02R (R = H or C1-4 alkyl), CONR2 (R = H or C1-4 alkyl), S02R (R = H or C1-4 alkyl), SOR (R = H or C1-4 alkyl), S02NR2 (R = H or C1-4 alkyl), OCOR (R = H or C1-4 alkyl), and COR (R = C1-4 alkyl).
Ring A may also be optionally substituted by 2 O atoms that when are adjacent on the ring may together represent the moiety -O-CR'2-O- where R' = H, C1-4 alkyl or F.
Z = one or more substituents on ring B selected from: H, Halo (from F, CI, Br, I), C1-6 alkyl, C1-6 alkoxy, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyi (halo is 1-3 atoms from F, CI, Br, I), C1-4 haloalkoxy (halo is 1-3 atoms from F, CI, Br, I), OH, SR (R = H or C1-4 alkyl), NR2 (R = H or C1-4 alkyl), NRS02R (R = H or C1-4 alkyl), NRCOR (R = H or C1- 4 alkyl), N02, CN, C02R (R = H or C1-4 alkyl), CONR2 (R = H or C1-4 alkyl), S02R (R = H or C1-4 alkyl), SOR (R = H or C1-4 alkyl), S02NR2 (R = H or C1-4 alkyl), OCOR (R = H or C1-4 alkyl), and COR (R = C1-4 alkyl). In one embodiment of the invention the pyrrolnitrin derivative is fludioxonil or a salt or an ester thereof.
Fludioxonil (CAS number: 131341-86-1) is represented by the following structure:
Figure imgf000008_0001
Synonyms for Fludioxonil
1 ) 4-(2,2-difluoro-1 ,3-benzdioxol-4-yl)-1 h-pyrrole-3-carbonitrile
2) 4-(2,2-difluoro-1 ,3-benzodioxol-4-yl)-1h-pyrrole-3-carbonitril
3) 4-(2,2-difluoro-1 ,3-benzodioxol-4-yl)-1h-pyrrole-3-carbonitrile
4) cgal 73506
5) CELEST 6) FLUDIOXONIL
7) MAXIM
8) 1 H-Pyrrole-3-carbonitrile, 4-(2,2-difluoro-1 ,3-benzodioxol-4-yl)-
9) fludioxonil (bsi, pa e-iso)
10) 4-(2,2-difluoro-1 ,3-benzodioxol-4-vl) pyrrole-3-carbonitrile
11) Celest Saphire
12) Fludioxinil
In a further embodiment of the invention, the pyrrolnitrin derivative is fenpiclonil or a salt or an ester thereof. Fenpiclonil (CAS number: 74738-17-3) is represented by the following structure:
Figure imgf000009_0001
Synonyms for Fenpiclonil
1) GAMBIT
2) BERET
3) 3-(2,3-dichlorophenyl)-4-cyanopyrrole
4) 4-(2,3-dichlorophenyl)-1 h-pyrrole-3-carbonitril
5) 4-(2,3-dichlorophenyl)-1 H-pyrrole-3- carbonitrile
6) 4-cyano-3-(2,3-dichlorophenyl)pyrrole
7) cga142705
It has now been found that Fludioxonil and Fenpiclonil are inhibitors of PIM kinase family members (PIM1 , PIM2 and PIM3) and also of ROCKIl kinase.
PIM Kinase activity
PIM kinases are cytoplasmic serine/threonine kinases that are known to be involved in regulation of apoptosis and cellular metabolism. Certain PIM kinases have been shown to be upregulated in cancers and as such their inhibition represents a mechanism of action by which Fludioxonil and Fenpiclonil can have an anti-tumour effect in conditions such as leukaemia, lymphoma, prostate cancer, colon cancer and pancreatic cancer. The below studies have shown this link:
Liver cancer: Gong (2009), Fujii (2005) and Wu (2010) have shown PIM-2 to promote humourigenesis and PIM-3 to accelerate hepatocellular carcinoma development when induced by hepatocarcinogen.
Gastric cancer: Zhen (2008) and Warnecke-Eberz (2009) have shown overexpression of PIM-1 in gastric glands to be associated with lymph node metastases.
Head and neck cancer: Beier (2007) has shown PIM-1 overexpression in head and neck squamous cell carcinomas.
Colon cancer: Popivanova (2007) has shown PIM-3 to be aberrantly expressed in human colon cancer cells but not normal colon mucosa.
Pancreatic cancer: Li (2006), Chen (2009) and Reiser-Erkan (2008) have shown
PIM-3 expression occurs in human pancreatic cancer but not normal cells and PIM-1 blockage using siRNA resensitises pancreatic cancer cells to apoptosis and PIM-1 levels correlate to clinicopathological parameters in pancreatic cancer.
Leukaemia/ly m phoma : Adam (2006), Hammerman (2005), Cohen (2004), Hogan
(2008), Lin (2010), Kim (2005), Chen (2008) and Brault (2010) have shown PIM-2 expression is increased in leukaemia/lymphoma, expression of PIM-1 and PIM-2 is dependent on Abl kinase activity and PIM-1 mediates homing and migration of malignant haematopoietic cells.
Oral cancer: Chiang (2006) and Choi (2010) have shown PIM-1 expression to be high in squamous cell carcinoma. Prostrate cancer: Chen (2005), umenthaler (2009), He (2007), Xu (2005), Dai (2005) and Roh (2008) have shown PIM-1 overexpression in prostatic carcinoma.
Breast cancer: Roh (2008) has shown PIM-1 overexpression to convert mammary epithelia cells to become tumourgenic.
Adipocyte tumours: Nga (2010) has shown benign and malignant adipocytic tumours to have strong PIM-1 expression.
PIM kinases are constitutively active and their activity as shown above and in Amaravadi (2005) and Shah (2008) supports in vitro and in vivo human cell growth and survival. ROCK II kinase
The pathology of a number of human and animal diseases including hypertension, erectile dysfunction, coronary cerebral circulatory impairments, neurodegenerative disorders and cancer can be linked directly to changes in the actin cytoskeleton. These diseases pose a serious unmet medical need. The actin cytoskeleton is composed of a meshwork of actin filaments and actin-binding proteins found in all eukaryotic cells. In smooth muscle cells the assembly and disassembly of the actin cytoskeleton is the primary motor force responsible for smooth muscle contraction and relaxation. In non- muscle cells, dynamic rearrangements of the actin cytoskeleton are responsible for regulating cell morphology, cell motility, actin stress fiber formation, cell adhesion and specialized cellular functions such as neurite retraction, phagocytosis or cytokinesis (Van Aelst, et al. Genes Dev 1997,11 ,2295).
The actin cytoskeleton is controlled by a family of proteins that are a subset of the Ras superfamily of GTPases. This subset currently consists of RhoA through E and RhoG (refereed to collectively as Rho), Rac 1 and 2, Cdc42Hs and G25K and TC10 isoforms (Mackay, et al. JBiol Chem 1998,273,20685). These proteins are GTP (guanine nucleotide triphosphate) binding proteins with intrinsic GTPase activity. They act as molecular switches and cycles between inactive GDP (guanine nucleotide diphosphate) bound and active GTP bound states. Using biochemical and genetic manipulations, it has been possible to assign functions to each family member. Upon activation the Rho proteins controls the formation of actin stress fibers, thick bundles of actin filaments, and the clustering of integrins at focal adhesion complexes. When activated the Rac proteins control the formation of lamellopodia or membrane ruffles on the cell surface and Cdc42 controls filopodia formation. Together this family of proteins plays a critical part in the control of key cellular functions including cell movement, axonal guidance, cytokinesis, and changes in cell morphology, shape and polarity.
Depending on the cell type and the activating receptor, the Rho proteins can control different biological responses. In smooth muscle cells, Rho proteins are responsible for the calcium sensitization during smooth muscle contraction. In non-smooth muscle cells the Rho GTPases are responsible for the cellular responses to agonist such as lysophosphatidic acid (LPA), thrombin and thromboxane A2 (Fukata, et al. Trends Pharcol Sci 2001 , 22,32).
Agonist response is coupled through heterotrimeric G proteins Galphal2 or Galphal3 (Goetzl, et al. Cancer Res 1999,59,4732; Buhl, et al. J Biol Chem 1995,270,24631) though other receptors may be involved. Upon activation Rho GTPases activate a number of downstream effectors including PIP5-kinase, Rhothekin, Rhophilin, PKN and Rho-Kinase isoforms ROCK-1/ROKbeta and ROCK-2/ROKalpha (Mackay and Hall J Biol Chem 1998,273, 20685; Aspenstrom Curr Opin Cell Biol 1999, 11 , 95; Amano, et al. Exp Cell Res 2000,261 , 44). Rho-kinase was identified as a RhoA interacting protein isolated from bovine brain (Matsui, et al. Embo J 1996,15,2208). It is a member of the myotonic dystrophy family of protein kinase and contains a serine/threonine kinase domain at the amino terminus, a coiled- coil domain in the central region and a Rho interaction domain at the carboxy terminus (Amano, et al. Exp Cell Res 2000,261 ,44). Its kinase activity is enhanced upon binding to GTP-bound RhoA and when introduced into cells, it can reproduce many of the activities of activated RhoA. In smooth muscle cells Rho-Kinase mediates calcium sensitization and smooth muscle contraction and inhibition of Rho-kinase blocks 5-HT and phenylephrine agonist induced muscle contraction. When introduced into non- smooth muscle cells, Rho- kinase induces stress fiber formation and is required for the cellular transformation mediated by RhoA (Sahai, et al. Curr Biol 1999, 9,136). Rho- kinase regulates a number of downstream proteins through phosphorylation, including myosin light chain (Somlyo, et al. J Physiol (Lond) 2000,522 Pt 2,177), the myosin light chain phosphatase binding subunit (Fukata, et al. J Cell Biol 1998, 141 , 409) and LIM- kinase 2 (Sumi, et al. J Bio Chem 2001 , 276,670). ROCK kinases are effectors of the small GTPase Rho and belong to the AGC family of kinases. The Rho-associated kinases (ROCKs) are serine/threonine protein kinases that serve as key downstream effectors of the Rho GTPase, RhoA, and play an important role in cytoskeletal function. The Rho/ROCK signal transduction participates in signalling pathways via rearrangement of the actin cytoskeleton that leads to various cellular reactions. The ROCKs impact cytoskeletal function through their ability to phosphorylate a range of proteins that directly regulate cytoskeletal proteins, including myosin light chain kinase, myosin light chain phosphatase, beta-catenin, cofilin, FAK and LIMK, and thus the ROCKs are positioned to play essential roles in the physiological and pathophysiological processes of various cell types. The ROCK enzymes, ROCK1 and ROCK2, share 65% overall homology. Gene knockout and siRNA studies suggest that each isoform plays distinct roles in mammalian biology. Despite having similar kinase domains, ROCK1 and ROCK2 may serve different functions and may have different downstream targets. ROCK1 expression tends to be ubiquitous, while ROCK2 is most highly expressed in cardiac and brain tissues. Clinically, inhibition of the ROCK pathway is believed to contribute to some of the cardiovascular benefits of statin therapy that are independent of lipid lowering (Liao et al 2010, J Cardiovascular Pharmacol 2007, 50(1):17-24).
Rho kinase is involved in a wide range of diseases such as glaucoma, vasospasm, pulmonary hypertension multiple sclerosis and nerve injury. Pharmacology studies of ROCK kinase inhibitors have shown that compounds of this activity are effective in animal models of glaucoma (for example at reducing intraocular pressure (IOP) in animal models by improving aqueous hour drainage through the trabecular pathway). Inhibition of Rho-kinase activity in animal models has demonstrated a number of benefits of Rho-kinase inhibitors for the treatment of human diseases. Several patents have appeared claiming (+)-trans-4- (1-aminoethyl)-1- (pyridin-4-ylaminocarbonyl) cyclohexane dihydrochloride monohydrate (WO-00078351 , WO-00057913) and substituted isoquinolinesulfonyl (EP-00187371) compounds as Rho-kinase inhibitors with activity in animal models. These include models of cardiovascular diseases such as hypertension (Uehata, et al. Nature 1997,389,990), atherosclerosis (Retzer, et al. FEBS Lett 2000, 466, 70), restenosis (Eto, et al. Am JPhysiol Heart Circ Physiol 2000, 278, H1744 ; Negoro, et al.
Biochem Biophys Res Commun 1999,262,211), cerebral ischemia (Uehata, et al. Nature 1997,389,990; Seasholtz, et al. Circ Res 1999, 84, 1186; Hitomi, et al. Life Sci 2000,67, 1929; Yamamoto, et al. J Cardiovasc Pharmacol 2000, 35, 203), cerebral vasospasm (Sato, et al. Circ Res 2000,87,195; Kim, et al. Neurosurgery 2000,46,440), penile erectile dysfunction (Chitaley, et al. Nat Med 2001,7,119), central nervous system disorders such as neuronal degeneration and spinal cord injury (Hara, et al. J Neurosurg 2000,93,94; Toshima, et al. Stroke 2000,31 ,2245) and in neoplasias where inhibition of Rho-kinase has been shown to inhibit tumor cell growth and metastasis (Itoh, et al. Nat Med 1999,5,221; Somlyo, et al. Biochem Biophys Res Commun 2000,269,652), angiogenesis (Uchida, et al. Biochem Biophys Res Commun 2000,269,633; Gingras, et al. Biochem J 2000,348 Pt 2, 273), arterial thrombotic disorders such as platelet aggregation (Klages, et al. I Cell Biol 1999,144,745; Retzer, et al. Cell Signal 2000, 12, 645) and leukocyte aggregation (Kawaguchi, et al. Eur JPharmacol 2000, 403, 203; Sanchez-Madrid, et al. Embo J 1999, 18, 501), asthma (Setoguchi, et al. Br J Pharmacol 2001, 132, 111; Nakahara, et al. Eur J Pharmacol 2000, 389, 103), regulation of intraoccular pressure (Honjo, et al. Invest Ophthalmol Vis Sci 2001, 42, 137) and bone resorption (Chellaiah, et al. JBiol Chem 2000, 275, 11993; Zhang, et al. J Cell Sci 1995, 108, 2285).
The inhibition of Rho-kinase activity in patients has benefits for controlling cerebral vasospasms and ischemia following subarachnoid hemorrhage (Pharma Japan 1995,1470, 16).
Non-isoform selective ROCK inhibitors such as fasudil have been shown to prevent cerebral vasospasm after subarachnoid hemorrhage. Despite the potential clinical importance of ROCK inhibition, fasudil is the only ROCK inhibitor approved for clinical use. Fasudil was approved in 1995 in Japan and China for prevention and treatment of cerebral vasospasm following surgery for subarachnoid hemorrhage and has since been used in over 124,000 patients in Japan. Many pharmaceutical and biotechnology companies are developing other selective and non-selective ROCK inhibitors (see table below modified and updated from Liao et al 2007). The proven safety and efficacy of fasudil suggests that ROCK inhibition will not have any safety limiting factors and that ROCK is a significant and genuine drug target. However even the relatively selective ROCK inhibitors such as Y27632 have been shown to inhibit additional kinases. Compounds selective for ROCK represent a significant advancement over those described currently as ROCK inhibitors.
ROCK inhibitors in development
Figure imgf000014_0001
Compound Therapeutic Area Company Development status
(intravenous and vasospasm, acute Pharmaceuticals for cerebral oral formulation) stroke, angina, Inc (licensed to vasospasm and (also known as HA- , pulmonary CoTherix in USA ischemia. Phase 2 1077) hypertension and Europe for use for other
in pulmonary indications hypertension)
Y27632 Reperfusion injury, Mitsubishi Discontinued, hypertension, Pharmaceuticals research tool stroke, asthma,
cancer
Y39983 Glaucoma Mitsubishi Phase 1
(Ophthalmic liquid Pharmaceuticals,
formulation) Senju
Pharmaceuticals,
Novartis AG
Wf-536 Cancer Mitsubishi Discovery Phase
Pharmaceuticals
SLx-2119 Artherosclerosis, Surface Logix Inc Discovery Phase fibrosis, solid
tumours
Azabenzimidazole- Hypertension antiGlaxo Smithkline Discovery Phase aminofurazans inflammatory
DE-104, Olefins, Glaucoma Santern Discovery Phase
Isoquinolines, Pharmaceuticals,
Indazoles, UBE Industries
pyridinealkene
derivatives
H-1152P Glaucoma Kowa Discovery Phase
Pharmaceuticals
ROCK inhibitor Asthma, BioFocus pic Discovery Phase cardiovascular
disease, erectile
dysfunction,
glaucoma, HIV
infection,
osteoporosis
XD-400 Atherosclerosis, Xcellsyz Ltd Discovery Phase fibrosis, solid
tumours
HMN-1152 Hearing loss, Nagoya University Discovery Phase tinnitus,
hypertension,
cerebrovascular
disease
AR-12286 Glaucoma Aerie Phase 2
Pharmaceuticals
Rhostatin Solid tumours, BioAxone Discovery Phase brain tumours Therapeutics
BA-210 Macular BioAxone Phase 2
degeneration, Therapeutics
spinal cord injury, Compound Therapeutic Area Company Development
status
glaucoma, cancer
Ki-23095 Cancer VasGene Discovery Phase
Therapeutics
ROCK inhibitor Cancer Bayer AG Discovery Phase (quinazoline)
Advantages of a combination PIM kinase inhibitor and ROCK kinase inhibitor
In some situations it may be advantageous for a compound to inhibit more than one or multiple protein kinases for maximal biological effect. Compounds that inhibit both PIM kinase and ROCK kinase may have a particular advantage over and above those inhibiting these kinases individually because the PIM kinase and ROCK kinase pathways intersect through p21Cip1/WAF1 (hereafter referred to as p21). In particular PIM kinase phosphorylates p21 on Thr 145 and thus regulates its stability and cellular localization in cells (Zhang et al., 2007, Mol Cancer Res 5(9): 909-922). When p21 is phosphorylated on Thr 145 it localizes to the nucleus and results in the disruption of the association between proliferating cell nuclear antigen (PCNA) and p21. Furthermore phosphorylation of Thr 145 promotes stabilization of p21. Inhibition of PIM and the consequent reduction in phosphorylation of Thr 145 of p21 would result in reduced stability of p21, and a reduction in cell proliferation. P21 also intersects with the ROCK/LIMK/Cofilin pathway. P21 forms a physical complex with ROCK and inhibits its activity (Lee et al., 2004, Journal Biological Chemistry 279(3): 1885-1891).
Ras activates p21 transcription and promotes p21 protein stability and it has been suggested that p21 is an essential factor in the signalling pathways that contribute to Ras-induced actin cytoskeletal re-modelling. The inhibition of PIM and ROCK together may therefore be particularly significant in tumours dependent upon or transformed through Ras mutation.
Pyrrolnitrin derivatives of formula I and salts thereof are referred to herein as "the compounds of the invention":
Pharmaceutically-acceptable salts include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound of formula I with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
By the term "ester" is included those formed with an alcohol of formula R1OH, wherein R1 represents aryl or alkyl; and those formed with a thiol of formula R SH, wherein R1 is as hereinbefore defined (i.e. a thioester). It is preferred that the ester is not a thioester. In embodiments R1 represents C1-6 alkyl, for example Ci alkyl (eg methyl).
Compounds of the invention may contain double bonds and may thus exist as E (entgegen) and Z (zusammen) geometric isomers about each individual double bond. All such isomers and mixtures thereof are included within the scope of the invention.
Compounds of the invention may also exhibit tautomerism. All tautomeric forms and mixtures thereof are included within the scope of the invention.
Compounds of the .invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism. Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation. The various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques. Alternatively the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation (i.e. a 'chiral pool' method), by reaction of the appropriate starting material with a 'chiral auxiliary' which can subsequently be removed at a suitable stage, by derivatisation (i.e. a resolution, including a dynamic resolution), for example with a homochiral acid followed by separation of the diastereomeric derivatives by conventional means such as chromatography, or by reaction with an appropriate chiral reagent or chiral catalyst all under conditions known to the skilled person. All stereoisomers and mixtures thereof are included within the scope of the invention.
Unless otherwise specified, C1-q alkyl groups (where q is the upper limit of the range) defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of two or three, as appropriate) of carbon atoms, be branched-chain, and/or cyclic (so forming a Ca-q-cycloalkyl group). Such cycloalkyl groups may be monocyclic or bicyclic and may further be bridged. Further, when there is a sufficient number (i.e. a minimum of four) of carbon atoms, such groups may also be part cyclic. Such alkyl groups may also be saturated or, when there is a sufficient number (i.e. a minimum of two) of carbon atoms, be unsaturated (forming, for example, a C2.q alkenyl or a C2-q alkynyl group). Where the number of carbon atoms permits, C1-q alkyl groups may also be spiro-groups (i.e. two cycloalkyi rings linked together by a single common carbon atom), although they are preferably not so.
The term "halo", when used herein, includes fluoro, chloro, bromo and iodo.
Heterocycloalkyi groups that may be mentioned include non-aromatic monocyclic and bicyclic heterocycloalkyi groups (which groups may further be bridged) in which at least one (e.g. one to four) of the atoms in the ring system is other than carbon (i.e. a heteroatom), and in which the total number of atoms in the ring system is between three and twelve (e.g. between five and ten). Further, such heterocycloalkyi groups may be saturated or unsaturated containing one or more double and/or triple bonds, forming for example a C2-q heterocycloalkenyl (where q is the upper limit of the range) or a C7-q heterocycloalkynyl group. C2-q heterocycloalkyi groups that may be mentioned include 7- azabicyclo-[2.2.1]heptanyl, 6-azabicyck>[3.1.1]heptanyl, 6-azabicyclo[3.2.1]-octanyl, 8- azabicyclo[3.2.1]octanyl, aziridinyl, azetidinyl, dihydropyranyl, dihydropyridyl, dihydropyrrolyl (including 2,5-dihydropyrrolyl), dioxolanyl (including 1 ,3-dioxolanyl), dioxanyl (including 1 ,3-dioxanyl and 1 ,4-dioxanyl), dithianyl (including 1,4-dithianyl), dithiolanyl (including 1 ,3-dithiolanyl), imidazolidinyl, imidazolinyl, morpholinyl, 7- oxabicyclo[2.2.1]heptanyl, 6-oxabicyclo[3.2.1]-octanyl, oxetanyl, oxiranyl, piperazinyl, piperidinyl, pyranyl, pyrazolidinyl, pyrrolidinonyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, sulfolanyl, 3-sulfolenyl, tetrahydropyranyl, tetrahydrofuranyl, tetrahydropyridyl (such as 1 ,2,3,4-tetrahydropyridyl and 1 ,2,3,6-tetrahydropyridyl), thietanyl, thiiranyl, thiolanyl, thiomorpholinyl, trithianyl (including 1 ,3,5-trithianyl), tropanyl and the like. Substituents on heterocycloalkyi groups may, where appropriate, be located on any atom in the ring system including a heteroatom. Further, in the case where the substituent is another cyclic compound, then the cyclic compound may be attached through a single atom on the heterocycloalkyi group, forming a so-called "spiro"-compound. The point of attachment of heterocycloalkyi groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system. Heterocycloalkyi groups may also be in the N- or S- oxidised form.
For the avoidance of doubt, the term "bicyclic" (e.g. when employed in the context of heterocycloalkyi groups) refers to groups in which the second ring of a two-ring system is formed between two adjacent atoms of the first ring. The term "bridged" (e.g. when employed in the context of heterocycloalkyi groups) refers to monocyclic or bicyclic groups in which two non-adjacent atoms are linked by either an alkylene or heteroalkylene chain (as appropriate).
Aryl groups that may be mentioned include C6-14 (such as Ce.^ (e.g. C6-10)) aryl groups. Such groups may be monocyclic or bicyclic and have between 6 and 14 ring carbon atoms, in which at least one ring is aromatic. C6-i4 aryl groups include phenyl, naphthyl and the like, such as 1,2,3,4-tetrahydronaphthyl, indanyl, indenyl and fluorenyl. The point of attachment of aryl groups may be via any atom of the ring system. However, when aryl groups are bicyclic or tricyclic, they are preferably linked to the rest of the molecule via an aromatic ring.
Heteroaryl groups that may be mentioned include those which have between 5 and 14 (e.g. 10) members. Such groups may be monocyclic, bicyclic or tricyclic, provided that at least one of the rings is aromatic and wherein at least one (e.g. one to four) of the atoms in the ring system is other than carbon (i.e. a heteroatom). Heteroaryl groups that may be mentioned include acridinyl, benzimidazolyl, benzodioxanyl, benzodioxepinyl, benzodioxolyl (including 1,3-benzodioxolyl), benzofuranyl, benzofurazanyl, benzothiazolyl, benzoxadiazolyl (including 2,1 ,3-benzoxadiazolyl), benzoxazinyl (including 3,4-dihydro-2H-1,4-benzoxazinyl), benzoxazolyl, benzomorpholinyl, benzoselenadiazolyl (including 2,1 ,3-benzoselenadiazolyl), benzothiadiazolyl (including 2,1 ,3-benzothiadiazolyl), benzothienyl, carbazolyl, chromanyl, cinnolinyl, furanyl, imidazolyl, imidazopyridyl (including imidazo[4,5-fo]pyridyl, imidazo[5,4- ?]pyridyl and imidazo[1 ,2-a]pyridyl), indazolyl, indolinyl, indolyl, isobenzofuranyl, isochromanyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiaziolyl, isothiochromanyl, isoxazolyl, naphthyridinyl (including 1 ,6-naphthyridinyl or, preferably, 1 ,5-naphthyridinyl and 1 ,8- naphthyridinyl), oxadiazolyl (including 1,3,4-oxadiazolyl), oxazolyl, phenazinyl, phenothiazinyl, phthalazinyl, pteridinyl, purinyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinolizinyl, quinoxalinyl, tetrahydroiso- quinolinyl (including 1,2,3,4-tetrahydroisoquinolinyl and 5,6,7,8-tetrahydroisoquinolinyl), tetrahydroquinolinyl (including 1,2,3,4-tetrahydroquinolinyl and 5,6,7,8- tetrahydroquinolinyl), tetrazolyl, thiadiazolyl (including 1 ,3,4-thiadiazolyl), thiazolyl, oxazolopyridyl (including oxazolo[4,5-0]pyridyl, oxazolo[5,4- )]pyridyl and, in particular, oxazolo[4,5-c]pyridyl and oxazolo[5,4-c]pyridyl), thiazolopyridyl (including thiazolo[4,5- b] pyridyl, thiazolo[5,4-/>]pyridyl and, in particular, thiazolo[4,5-c]pyridyl and thiazolo[5,4- c] pyridyl), thiochromanyl, thienyl, triazolyl (including 1 ,2,3-triazolyl and 1 ,2,4-triazolyl) and the like. Substituents on heteroaryl groups may, where appropriate, be located on any atom in the ring system including a heteroatom. The point of attachment of heteroaryl groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system. However, when heteroaryl groups are polycyclic, they are preferably linked to the rest of the molecule via an aromatic ring. Heteroaryl groups may also be in the N- or S- oxidised form.
Heteroatoms that may be mentioned include phosphorus, silicon, boron, tellurium, selenium and, preferably, oxygen, nitrogen and sulphur.
For the avoidance of doubt, in cases in which the identity of two or more substituents in a compound of the invention may be the same, the actual identities of the respective substituents are not in any way interdependent. For example, in the situation in which two X1 groups are present, which both represent R5a, i.e. a C1-6 alkyl group optionally substituted as hereinbefore defined, the alkyl groups in question may be the same or different. Similarly, when groups are substituted by more than one substituent as defined herein, the identities of those individual substituents are not to be regarded as being interdependent. For example, when there are two X1 substituents present, which represent -R5a and -C(0)R5b in which R5b represents R5a, then the identities of the two R53 groups are not to be regarded as being interdependent. Likewise, when Y2 or Y3 represent e.g. an aryl group substituted by G1 in addition to, for example, C1-8 alkyl, which latter group is substituted by G , the identities of the two G groups are not to be regarded as being interdependent. In a second aspect of the invention, there is provided a pharmaceutical composition comprising a pyrrolnitrin derivative of formula I or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier.
In one embodiment of the invention there is provided a composition comprising between 10mg and 2000mg of an active ingredient per dosage unit, wherein the active ingredient is a compound of the invention or a derivative, salt or variant thereof.
By dosage unit we mean the unit of medicament administered to a patient at one time. For example, the dosage unit, or single dose may be administered by a single capsule/tablet, single injection, or single intravenous infusion, a single subcutaneous injection, or by a single procedure using other routes of administration, as discussed below. Alternatively, the single dose may be administered to the patient by two or more capsules/tablets or injections given simultaneously or sequentially to deliver the entire dose to the patient in the continuous, single and defined treatment period; by two or more intravenous infusions given simultaneously or sequentially to deliver the entire dose to the patient in the continuous, single and defined treatment; or by multiple procedures using other routes of administration as discussed below.
Alternatively, the single dose to be administered to the patient can be delivered by a combination of routes to deliver the entire dose to the patient in the continuous, single and defined treatment.
The dosage unit may then be repeated at intervals of time such as a few hours, days, weeks, or months later.
Dosage units can be administered to patients in such a way that the patient receives a loading dose followed by one or more maintenance doses. For example the loading dose may be a high dose in order to quickly reach a desired plasma concentration and then subsequent maintenance doses are a lower dose than the loading dose in order to maintain the required plasma concentration. By active ingredient we mean the molecule having the desired effect. In this case of this invention we primarily mean the compounds of the invention and derivatives, salts or variants thereof.
By variants and derivatives we mean any molecules of substantially identical chemical structure but including minor modifications that do not alter activity but may offer improved or alternative properties for formulation, such as formation into a salt.
In human therapy, the compound of the invention containing composition, and medicaments of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient, diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
For example, the compound of the invention containing composition, and medicaments 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 compound of the invention containing composition, and medicaments of the invention may also be administered via intracavernosal injection.
Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, 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 compound of the invention containing composition, medicaments and pharmaceutical compositions 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 compound of the invention containing composition, and medicaments of the invention can also be administered parenterally, for example, intravenously, intra- arterially, intraperitoneally, intra-thecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They 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.
Medicaments and pharmaceutical compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain antioxidants, 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 medicaments and pharmaceutical compositions 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 requiring 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 kind previously described.
The compound of the invention containing composition, and medicaments of the invention can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134A3 or 1,1,1,2,3,3,3- heptafluoropropane (HFA 227EA3), carbon dioxide or other suitable gas. In the case of a pressurised aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active agent, e.g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant,, e.g. sorbitan trioleate. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the invention containing composition, of the invention and a suitable powder base such as lactose or starch.
Aerosol or dry powder formulations are preferably arranged so that each metered dose or "puff contains an effective amount of an agent or polynucleotide of the invention for delivery to the patient. It will be appreciated that the overall daily dose with an aerosol will vary from patient to patient, and may be administered in a single dose or, more usually, in divided doses throughout the day.
Alternatively, the compound of the invention containing composition, and medicaments of the invention can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, gel, ointment or dusting powder. The compound of the invention containing composition, and medicaments of the invention may also be transdermal^ administered, for example, by the use of a skin patch. They may also be administered by the ocular route, particularly for treating diseases of the eye.
For ophthalmic use, the compound of the invention containing composition, and medicaments of the invention can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum. For application topically to the skin, the compound of the invention containing composition, and medicaments of the invention can be formulated as a suitable ointment containing the active agent suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene agent, emulsifying wax and water. Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water. 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.
Generally, in humans, oral or parenteral administration of the compound of the invention containing composition, medicaments and pharmaceutical compositions of the invention is the preferred route, being the most convenient. For veterinary use, the compound of the invention containing composition, and medicaments of the invention are administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
The compound of the invention containing composition, as defined herein may be formulated as described in the accompanying Examples.
The composition may comprise any effective amount of active ingredient, this may be between 10mg and 2000mg of active ingredient per dosage unit, and preferably is between 50mg and lOOOmg. Advantageously it is lOOOmg. Conveniently, the dosage unit contains an amount of active ingredient per dosage unit selected from 10mg, 20mg, 25mg, 50mg, 100mg, 200mg, 300mg, 400mg, 450mg, 600mg, 750mg, 950mg, 10OOmg and 1200mg.
Alternatively, the composition may comprise between 10-50mg, 10-75mg, 10-100mg, 10- 200mg, 10-300mg, 10-400mg, 10-600mg, 10-750mg, 10-950mg, 10-1 OOOmg, 10- 1200mg, 50-75mg, 50-100mg, 50-200mg, 50-300mg, 50-450mg, 50-600mg, 50-750mg, 50-950mg, 50-1 OOOmg, 50-1200mg, 75-1 OOmg, 75-200mg, 75-300mg, 75-450mg, 75- 600mg, 75-750mg, 75-950mg, 75-1000mg, 75-1200mg, 100-200mg, 100-300mg, 100- 450mg, 100-600mg, 100-750mg, 100-950mg, 100-1 OOOmg, 100-1200mg, 200-300mg, 200-450mg, 200-600mg, 200-750mg, 200-950mg, 200-1 OOOmg, 200-1200mg, 300- 450mg, 300-600mg, 300-750mg, 300-950mg, 300-1 OOOmg, 300-1200mg, 400-600 mg, 400-750mg, 400-950mg, 400-1 OOOmg, 400-1200mg, 450-600mg, 450-750mg, 450- 950mg, 450-1 OOOmg, 450-1200mg, 600-750mg, 600-950mg, 600-1 OOOmg, 600-1200mg, 700-950mg, 700-IOOOmg, 700-1200mg, 950-1000mg, 950-1200mg and 1000-1200mg
Preferably there is 400-600mg of active ingredient. More preferably there is 400-1200mg of active ingredient. Most preferably there is 10OOmg of active ingredient.
Conveniently, the composition is pharmaceutically acceptable, and may optionally contain a pharmaceutically acceptable excipient, diluent, carrier or filler.
The examples describe some methods of producing pharmaceutical formulations, however the skilled person will appreciate that the most appropriate formulation will depend on a number of factors including route of administration, patient type (e.g. patient age, weight/size).
A further aspect of the invention provides a method of treating cancer (such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases) comprising administering to the patient an effective amount of a pyrrolnitrin derivative of formula I or a salt or an ester thereof.
A further aspect of the invention provides a method of treating diseases and conditions selected from vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma osteoporosis, reperfusion injury, asthma, fibrosis, anti-inflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, and other indications mediated by Rho-kinase, e.g., coronary heart disease. In addition, the compounds of the invention are useful to treat erectile dysfunction, i.e. erectile dysfunction mediated by Rho-kinase. Erectile dysfunction can be defined as an inability to obtain or sustain an erection adequate for intercourse.
The term "patient" includes all animals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, and pigs. The term "patient" means an animal having a disorder in need of treatment. Preferably, the patient is selected from humans, cows, dogs, cats, goats, sheep, and pigs. More preferably, the patient is a human.
The compounds of the invention may be used in cancer treatment either alone or in combination with well known anti-cancer agents.
Cancer treatments promote tumour regression by inhibiting tumour cell proliferation, inhibiting angiogenesis (growth of new blood vessels that is necessary to support tumour growth) and/or prohibiting- metastasis by reducing tumour cell motility or invasiveness. Therapeutic compositions of the invention may be effective in adult and pediatric oncology including in solid phase tumours/malignancies, locally advanced tumours, human soft tissue sarcomas, metastatic cancer, including lymphatic metastases, blood cell malignancies including multiple myeloma, acute and chronic leukemias, and lymphomas, head and neck cancers including mouth cancer, larynx cancer and thyroid cancer, lung cancers including small cell carcinoma and non-small cell cancers, breast cancers including small cell carcinoma and ductal carcinoma, gastrointestinal cancers including esophageal cancer, stomach cancer, colon cancer, colorectal cancer and polyps associated with colorectal neoplasia, pancreatic cancers, liver cancer, urologic cancers including bladder cancer and prostate cancer, malignancies of the female genital tract including ovarian carcinoma, uterine (including endometrial) cancers, and solid tumour in the ovarian follicle, kidney cancers including renal cell carcinoma, brain cancers including intrinsic brain tumours, neuroblastoma, astrocytic brain tumours, gliomas, metastatic tumour cell invasion in the central nervous system, bone cancers including osteomas, skin cancers including malignant melanoma, tumour progression of human skin keratinocytes, squamous cell carcinoma, basal cell carcinoma, hemangiopericytoma and Karposi's sarcoma. Compounds of the invention may be administered to treat cancer. The cancer to be treated is preferably selected from pancreatic cancer, ovarian cancer, breast cancer, prostate cancer, liver cancer, chondrosarcoma, lung cancer, head and neck cancer, colon cancer, sarcoma, leukaemia, myeloma, lymphoma, kidney cancer, thyroid cancer and brain cancers such as glioblastoma.
Therapeutic compositions can be administered in therapeutically effective dosages alone or in combination with adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy, and may provide a beneficial effect, e.g. reducing tumour size, slowing rate of tumour growth, inhibiting metastasis, or otherwise improving overall clinical condition, without necessarily eradicating the cancer.
The composition can also be administered in therapeutically effective amounts as a portion of an anti-cancer cocktail. An anti-cancer cocktail is a mixture of the compound or modulator of the invention with one or more anti-cancer drugs in addition to a pharmaceutically acceptable carrier for delivery. The use of anti-cancer cocktails as a cancer treatment is routine. Anti-cancer drugs that are well known in the art and can be used as a treatment in combination with the compounds of the invention include: 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine, Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16- 213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1 , Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha- 2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541 , NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474,4HC,6-[4-(2-Piperidin-1-ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1 ,5- ajpyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat.
In addition, therapeutic compositions of the invention may be used for prophylactic treatment of cancer. There are hereditary conditions and/or environmental situations (e.g. exposure to carcinogens) known in the art that predispose an individual to developing cancers. Under these circumstances, it may be beneficial to treat these individuals with therapeutically effective doses of the compounds of the invention to reduce the risk of developing cancers.
In vitro models can be used to determine the effective doses of the compounds of the invention as a potential cancer treatment. These in vitro models include proliferation assays of cultured tumour cells, growth of cultured tumour cells in soft agar (see Freshney, (1987) Culture of Animal Cells: A Manual of Basic Technique, Wily-Liss, New York, NY Ch 18 and Ch 21), tumour systems in nude mice as described in Giovanella et a/., J. Natl. Can. Inst, 52: 921-30 (1974), mobility and invasive potential of tumour cells in Boyden Chamber assays as described in Pilkington et at., Anticancer Res., 17: 4107-9 (1997), and angiogenesis assays such as induction of vascularization of the chick chorioallantoic membrane or induction of vascular endothelial cell migration as described in Ribatta et a/., Intl. J. Dev. Biol., 40: 1189-97 (1999) and Li ef a/., Clin. Exp. Metastasis, 17:423-9 (1999), respectively. Suitable tumour cells lines are available, e.g. from American Type Tissue Culture Collection catalogues.
In one embodiment, the method, use or composition of the invention additionally comprises a further chemotherapeutic agent. Preferably, the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Aisterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine , Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16-213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1 , Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha-2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541 , NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474,4HC,6-[4-(2-Piperidin-1- ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1 ,5-a]pyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat.
Due to the favourable toxicity and safety profiles of the compounds of the invention, the compounds may be used to treat patients who may not be amenable to conventional chemotherapeutic agents. Conventional chemotherapies are generally immunosuppressive and so are withdrawn in certain situations, such as for patients who are pre-operative, post-operative, receiving radiation, terminally ill, elderly, or receiving adjuvant or neo-adjuvant thereapy. However, the compounds of the invention may still be used in such situations.
Therefore, in a further aspect of the invention the compounds of the invention are used to treat patients who may not be amenable to conventional chemotherapeutic agents, particularly those from whom conventional chemotherapeutic agents have been withdrawn.
A further aspect of the invention provides a kit of parts comprising:
(i) pyrrolnitrin derivative of formula I or a salt or ester thereof or a pharmaceutical composition comprising a pyrrolnitrin derivative of formula I or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier;
(ii) apparatus for administering the compound or pharmaceutical composition; and
(iii) instructions for use.
In one embodiment, the kit of parts additionally comprises a further chemotherapeutic agent. Preferably the further chemotherapeutic agent is selected from 17AAG, 17D AG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine, Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16- 213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1 , Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha- 2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541 , NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474,4HC,6-[4-(2-Piperidin-1-ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1 ,5- a]pyrimidine (Dorsomorphin),Etoposide,Gemcitabine,Mitoxanthrone and Vorinostat.
EXAMPLES The following examples embody various aspects of the invention. It will be appreciated that the specific compounds used in the examples serve to illustrate the principles of the invention and are not intended to limit its scope.
The following examples are described with reference to the accompanying figures in which:
Figure 1 - Kinase specificity of fludioxonil and fenpiclonil
Figure 1 shows the % inhibition of Pim1, 2 and 3 kinase with increasing concentration (μΜ) of fludioxonil (1A) or fenpiclonil (1B). Figure 2 - Kinase specificity of fludioxonil and fenpiclonil
Figure 2 shows by western blot PIM 1 kinase and cleaved PARP levels in response to increasing concentrations if fludioxonil and fenpiclonil.
Figure 3 - Rock kinase specificity of fludioxonil and fenpiclonil
Figure 3 shows the % inhibition of Rock I and Rock II kinase with increasing concentration of fludioxonil or fenpiclonil. Figure 4 - Bodyweights and liver weights from a PC3 xenograft study in mice
Figure 4 shows (a) bodyweight over duration of study (b) terminal bodyweight and (c) liver weight of mice from a PC3 xenograft study with fludioxonil (CXR6032) and fenpiclonil (CXR6069). Figure 5 - Tumour volume and weight from a PC3 xenograft study in mice
Figure 5 shows (a) ALT, AST and ALP levels, (b) tumour volume and (c) tumour weight over the duration of the PC3 xenograft study with fludioxonil (CXR6032) and fenpiclonil (CXR6069).
Figure 6 - Fludioxonil whole blood pK results
Figure 6 shows the Log10 of fludioxonil concentration in ng/mL over time, both with a single dose and with 4 daily repeat doses.
Figure 7 - Fenpiclonil whole blood pK results
Figure 7 shows the Log10 of fenpiclonil concentration in ng/mL over time, both with a single dose and with 4 daily repeat doses. Figure 8 - Diagram of GM-CSF signalling pathway overlaid with fludioxonil (CXR6032) EC50 signature list
Figure 8 shows the fold change data from fluduioxonil (CXR6032) EC50 signature list superimposed on the genes shown in the above pathway using IPA software (Ingenuity Systems Inc.). Fold changes (black lettering under gene icons) greater than 1.5 fold causes in the gene icons to be coloured either dark grey = up-regulated or light grey = down-regulated. Icons in medium grey signify fold changes of less than 1.5. Icons in white signify genes were un-changed by the treatment or not significantly changed (p>0.01).
Figure 9 - Diagram of GM-CSF signalling pathway overlaid with fenpiclonil (CXR6069) EC50 signature list
Figure 9 shows the fold change data from fenpiclonil (CXR6032) EC50 signature list superimposed on the genes shown in the above pathway using IPA software (Ingenuity Systems Inc.). Fold changes (black lettering under gene icons) greater than 1.5 fold causes in the gene icons to be coloured either dark grey = up-regulated or light grey = down-regulated. Icons in medium grey signify fold changes of less than 1.5. Icons in white signify genes were un-changed by the treatment or not significantly changed (p>0.01). Figure 10 - Diagram of acute myeloid leukaemia signalling pathway overlaid with fludioxonil (CXR6032) EC50 signature list
Figure 10 shoes the fold change data from fluduioxonil (CXR6032) EC50 signature list superimposed on the genes shown in the above pathway using I PA software (Ingenuity Systems Inc.). Fold changes (black lettering under gene icons) greater than 1.5 fold causes in the gene icons to be coloured either dark grey = up-regulated or light grey = down-regulated. Icons in medium grey signify fold changes of less than 1.5. Icons in white signify genes were un-changed by the treatment or not significantly changed (p>0.01). Figure 11 - Diagram of acute myeloid leukemia signalling pathway overlaid with fenpiclonil (CXR6069) EC50 signature list
Figure 11 shows the fold change data from fenpiclonil (CXR6032) EC50 signature Rst superimposed on the genes shown in the above pathway using IPA software (Ingenuity Systems Inc.). Fold changes (black lettering under gene icons) greater than 1.5 fold causes in the gene icons to be coloured either dark grey = up-regulated or light grey = down-regulated. Icons in medium grey signify fold changes of less than 1.5. Icons in white signify genes were un-changed by the treatment or not significantly changed (p>0.01).
Figure 12 - Diagram of RhoA signalling pathway overlaid with fludioxonil (CXR6032) EC50 signature list
Figure 12 shows the fold change data from fludioxonil (CXR6032) EC50 signature list superimposed on the genes shown in the above pathway using IPA software (Ingenuity Systems Inc.). Fold changes (black lettering under gene icons) greater than 1.5 fold causes in the gene icons to be coloured either dark grey = up-regulated or light grey = down-regulated. Icons in medium grey signify fold changes of less than 1.5. Icons in white signify genes were un-changed by the treatment or not significantly changed (p>0.01).
Figure 13 - Diagram of RhoA signalling pathway overlaid with fenpiclonil (CXR6069) EC50 signature list Figure 13 shows fold change data from fenpiclonil (CXR6032) EC50 signature list superimposed on the genes shown in the above pathway using IPA software (Ingenuity Systems Inc.). Fold changes (black lettering under gene icons) greater than 1.5 fold causes in the gene icons to be coloured either dark grey = up-regulated or light grey = down-regulated. Icons in medium grey signify fold changes of less than 1.5. Icons in white signify genes were un-changed by the treatment or not significantly changed (p>0.01).
Figure 14 - RNA microarray for ftudioxonil and fenpiclonil experimental structure Figure 14 shows the fludioxonil (CXR6032) and fenpiclonil (CXR6069) RNA microarray experimental structure using triplicates of each sample.
Example 1: selection of the compounds of the invention based on specific properties
The following method details how the compounds of the invention were selected to arrive at those to be used for the treatment of diseases such as cancer.
The compounds of the invention were selected using the following method:
Step 1 : A database was produced of some 182 compounds registered for use as agro- or industrial chemicals with available dossiers regarding bioactivity and toxicity collated from sources including the UK's Advisory Committee on Pesticides (ACP), the US Environmental Protection Agency (EPA), the European Food Safety Authority (EFSA), the US Department of Health & Human Services' Agency for Toxic Substances and Disease Registry (ATSDR), Canada's Bureau of Chemical Safety and the International Programme on Chemical Safety (IPCS).
Table 13 details the 182 compounds contained in the database.
Step 2: A panel of 144 chemically diverse compounds were selected as being readily available from the database produced in (1).
Table 14 details the 144 chemically diverse compounds.
Step 3: The compounds from (2) were tested against 15 human tumour cell lines for cytotoxicity. Effects on cell survival at 72 hours and at 7 days at a single compound concentration (100μΜ) were measured. Tables 11 and 12 show cell proliferation data for the 144 compounds at 72 hours and 7 day exposure, respectively. The data is expressed as a percentage of untreated control cells, so 100 % means that there are the same number of cells as the control, less than 100 % indicates there are fewer cells than the control and greather than 100 % indicates that more cells than the control are present. Cell assay methods
Cell Culture
Test compounds were dissolved in DMSO. Cells were seeded at a density of 500 cells per well in 75 μΙ of DMEM (10% FBS/2 mM Glutamine) media for 7 day incubation in 96- well plates or 5000 cells per well for 72 hrs incubations. The following day, 25 μΙ of test compounds, diluted 1:25 in DMEM (10% FBS), were added to the wells to give a final compound concentrations of 100μΜ. Duplicate plates were assembled and then incubated for either 72 hrs or 7 days at 37°C/5% C02. No media/drug replacement during the incubation period was undertaken.
WST-1 Assay
Cell number was estimated using WST-1 reagent from Roche Applied Bioscience according to the manufacturer's instructions. Briefly 10μΙ of reagent was added to 100 μΙ of media and incubated until colour developed. The plates were read at wavelength 450nm on an appropriate absorbance plate reader.
Data Analysis
Data from the plate reader in text format was entered into an excel spreadsheet. The data was converted into a percentage value of the untreated control wells using the following formula:
Figure imgf000035_0001
Step 4: The cytotoxicity data from (3) was used to rank compounds with respect to (a) global cytotoxicity; (b) selective cytotoxicity; and (c) published literature (i.e. a lack of published literature linking the compounds with cytotoxicity).
Step 5: Panel of 48 compounds chosen as the top-ranked compounds using the results of (4). Step 6: Compounds selected under (5) were tested for cytotoxicity with compound dose response (see table 10) and the published toxicology data reviewed for (a) bioavailability (see table 3); (b) rat LD50 (see table 5); (c) Therapeutic Index (NOEL (No Observable Effects Limit): IC50 ratio) (table 2).
Step 7: Panel from (6) screened for effects on protein kinase activities. The results are shown in Table 8.
Step 8: Compounds selected for further development on basis of meeting at least criteria (1) and (2) (but preferably more) of the 7 criteria listed below:
Step 9: (optional) Kinase inhibition (such as PIM-Kinase) by dose response results can be used to select specific compounds for further development (see Examples 3 and 4) Step 10: Compounds selected under (8) and (9) will be tested further in xenograft studies (see Example 6).
The seven criteria for compound selection 1. Available Toxicoloqv/Toxicitv Data must be available
Toxicological testing is a significant barrier in the pre-clinical development of pharmaceuticals, both in terms of cost and risk. Selection of molecules already tested for toxicity is therefore an attractive alternative.
Sufficient publically available safety pharmacology, toxicology and toxicity data should be available, as outlined in the ICH Safety Guidelines S9 available from http:/ www.ich.org/products/guidelines/safety/article/safety-guidelines, to support the compound's use in clinical trials.
2. Favourable Therapeutic Index - NOELICsn Ratio must be greater than 5
Current anticancer drugs give rise to significant side-effects at the dose levels given to patients. This is due to the fact that these traditional molecules function as cellular toxins, their anti-cancer properties arising from the toxic nature. The methodology described herein of selecting compounds shown to be of low toxicity will overcome this limitation and give rise to anti-cancer agents with lower toxicology liabilities. An ideal pharmaceutical would exhibit efficacy in the absence of any toxicological or pharmacological side-effects at the therapeutic dose. A compound with these desirable properties is described as having a favourable therapeutic index or therapeutic window.
As an indicator of compounds with a potentially favourable Therapeutic Index we use the ratio of the rat "No Observed Effect Level" (NOEL) from toxicology studies and the lowest IC50 value of the compound on our cell line panel as a proxy. Compounds which have a larger ratio are more likely to inhibit the growth of tumour cells whilst having no observable side-effects on the animal.
Table 2 shows which of a selection of 48 compounds (as selected according to step 5 of the screening method) have an IC50/NOEL ratio of greater than five. Table 2
Figure imgf000037_0001
Rat Rat
Min IC50 Ratio
Compound NOEL/NOAEL NOEL/NOAEL
(M) IC50/NOEL
(mg/kg) (M)
Benfuracarb 1.3 3.2 10.00 0.3
Flusilazole 1.5 4.8 10.00 0.5
fenpropathrin 1.5 4.3 15.00 0.3
Fenbuconazol 5.2 15.4 15.00 1.0
Transfluthrin 15 40.4 22.00 1.8
Fenazaquin 5 16.3 0.26 62.8
Fenoxycarb 80 265.8 10.20 26.1
Metconazole 2.5 7.8 17.00 0.5
Folicur 10.8 35.1 26.00 1.3
ethiocarb 1.3 5.8 2.60 2.2
Myclobutanil 15 51.9 25.00 2.1
cyfluthrin 10 23.1 17.00 1.4
Bromuconazol 14 37.1 30.00 1.2
Dimethomorph 3.75 9.7 15.00 0.6
Triazophos 0.29 0.9 22.00 0.0
phorate 0.05 0.2 26.00 0.0
irgarol 9.7 38.3 22.00 1.7
Pyriproxifen 2 74.8 15.70 4.8
tau-fluvalinate 1 2.0 14.00 0.1
Fluoroglycofen-ethyl 84 200.0 10.00 20.0
Esfenvalerate 15 35.7 9.58 3.7
Fenoxaprop-ethyl 6 16.6 43.00 0.4
cyhalofop-butyl 3 8.4 35.00 0.2
Flufenoxuron 12.5 25.6 8.00 3.2
Dimefuron 20 59.0 27.00 2.2
Isoproturon 50 242.7 20.00 12.1
Ethoprophos 0.1 0.4 43.31 0.0
3. Oral bioavailability preferred to be greater than 20% Oral bioavailability is an attractive property for pharmaceutical compounds. Oral dosing will mean patients do not have to travel to the hospital situation regularly during their treatment. This will not only save considerable resources at the treatment centre but also reduce patient comfort by reducing travel and in-patient time. Oral bioavailability is calculated by measuring the plasma concentration of the compound over time after administering the drug both orally (po) and intravenously (IV). The area under the curve (AUC) is measured for both modes of administration and is corrected for different dose levels for the different routes. The following formula is used for calculating oral bioavailability (F):
[AUC]po * Dose,
(AUC]IV * Dose,
Table 3 shows which of a selection of compounds has an oral bioavailability of >20%:
Table 3
Figure imgf000039_0001
Compound Bioavailability
Metconazole >80%
Folicur 90%
Methiocarb 0%
Myclobutanil -50%
cyfluthrin >60%
Bromuconazole -25%
Dimethomorph 90%
Triazophos -75%
Phorate 30%
irgarol 0%
Pyriproxifen 7%
tau-fluvalinate 20-40%
Fluoroglycofen-ethyl 20%
Esfenvalerate -50%
Fenoxaprop-P-ethyl -50%
cyhalofop-butyl -90%
Flufenoxuron 50%
Dimefuron 30%
Isoproturon 85%
Ethoprophos 95%
4. When data is available it is preferred that the LP™ be greater than 500 mq/ka
The selection approach described is to select compounds which have undergone extensive toxicological testing and have been deemed of low toxicity and safe for release to the environment as either agrochemicals or for use in other industries.
One measure of toxicity is the Lethal Dose 50 (LD50). This is the concentration of compounds which, when dosed to a test population, causes the death of 50 % of the test population within a set time period. LD50 was introduced in the 1920's by Trevan (The error of determination of toxicity. Proc Roy Soc 1927;101 B:483-514) as a measure of toxicity but has been largely replace the Maximum Tolerated Dose or MTD. MTD is the highest concentration at which the death rate in a dosed population is no greater than the death rate in a control population
According to the classification of Hodge and Sterner (J Ind Hyg Toxicol. 1949 Mar;31(2):79-92), the toxicity of molecules can be classified according to the table below. In contrast with current anti-cancer agents which are known toxic agents, the compounds selected are of no greater than "low toxicity", with oral LD50 values in the rat no lower than 500 mg kg. Table 4 details the different measures of toxicity of compounds. Table 5 details which of a selection of compounds have an oral LD50 in the rat of over 500mg/kg. Table 4
Figure imgf000041_0001
Table 5
Figure imgf000041_0002
Fenpiclonil 5000
Cyprodinil >2000
brodifacoum 1
flocoumafen 0
Difenoconazole 1450
kresoxim-methyl 5000
Azamethiphos 830
Benfuracarb 120 (rats),250 (dogs)
Flusilazole 674
fenpropathrin 870
Fenbuconazole 2000
Transfluthrin 590 - 870
Fenazaquin 134
Fenoxycarb 10000
Metconazole 595
Folicur 1700
Methiocarb 22
Myclobutanil 2000
cyfluthrin 600-1200 (r), >100 (d)
Bromuconazole 300-400
Dimethomorph 3900
Triazophos 65
Phorate 3
irgarol 2000
Pyriproxifen >5000
tau-fluvalinate >3000
Fluoroglycofen-ethyl 1480
Esfenvalerate -200
Fenoxaprop-P-ethyl -2500
cyhalofop-butyl >5000
Flufenoxuron 3000
Dimefuron 2000
Isoproturon 1826
Ethoprophos 33 5. Preferred that in vivo half-life greater than 4 hours
For a compound to exert its biological effect, it is clear that the compound must be exposed to its molecular target. Should a compound be metabolised or excreted rapidly then compound concentration and so pharmacological effect would naturally fall too. In order to circumvent these issues, medicinal chemistry manipulates the chemical structure of the molecule in order to improve resistance to metabolism and excretion.
Compounds are selected from a library partly on the basis of in vivo half-life measured during development of the compounds as agro-industrial chemicals. In vivo half-life is a measure of the rate at which a compound is excreted or metabolised. The change in plasma concentration of a compound following dosing is described by the equation:
Where Ct is the concentration at a time t, C0 represents the concentration at t = 0, Ke is the rate constant of elimination and t is the chosen time point. The half-life can then be calculated using the relationship between rate constant and half-life using the formula:
Figure imgf000043_0001
It is clearly desirable not to have the compound eliminated rapidly from the plasma in order to increase the likely efficacy of the compound in vivo. Our threshold of 4 hours means that if peak dosing were 4 fold of the IC level, then compound levels would be at least the IC50 level for 8 hours out of 24 and therefore dosing would be a convenient 3 times per day. Table 6 details which of a selection of compounds have a half-life of >4 hours.
Table 6
Figure imgf000043_0002
Compound Half-Life
Thiodicarb Rapid
Buprofezin Rats 13hrs and 60 hrs
Dinocap 3 hrs
Thiophanate-methyl < 24hrs
Tolylfluanid 3hrs, 3 days
Triazoxid 2.6 - 5.2 hrs + slow phase
Tebufenpyrad <72hrs
Triflusulfuron-methyl 15 hrs
Fenpiclonil -160
Cyprodinil Rats ~24hrs
brodifacoum Rat 24days, Dog 120 days flocoumafen > 7 days
Difenoconazol <72hrs
kresoxim-methyl 16.9-30.5
Azamethiphos <6hrs
Benfuracarb ~24hrs
Flusilazole 30hrs
fenpropathrin 24hrs
Fenbuconazol 15 hrs
Transfluthrin < 48 hrs
Fenazaquin 25 hrs
Fenoxycarb ~24hrs
Metconazole ND
Folicur 40hrs
Methiocarb ND
Myclobutanil 5.3hrs, 25.7 hrs cyfluthrin <24hrs
Bromuconazol 9 - 90 hrs
Dimethomorph 3 hrs
Triazophos ND
phorate >7 days
irgarol ND
Pyriproxifen 2-8hrs, 23-35hrs tau-fluvalinate ~24hrs
Fluoroglycofen-ethyl 12-37hrs
Esfenvalerate ~12hrs
Fenoxaprop-ethyl 10 hrs, 3 days
cyhalofop-butyl ~3h
Flufenoxuron ND
Dimefuron < 72hrs
Isoproturon 8h
Ethoprophos < 6 hrs 6. Preferred that compounds exhibit activity on Kinase or other molecular target
In order to direct pre-clinical development it is useful to have an idea of which tumour types may be best targeted with the molecule. Some of this information can be learned from testing the compound for effects on cellular proliferation on a panel of tumour ceH lines. An alternative approach is to test the molecules for activity, inhibitory or activatory, on to use in vitro assays of molecular targets such as protein kinases, GPCRs, enzymes or other cellular machinery. It is preferable that the compounds have a known mode of action. One aspect of this could be inhibitory activity of a compound against a member(s) of a panel of protein kinases known to be implicated in cancer biology. Such a panel could be comprised of the protein kinases shown in table 7: Table 7: A panel of kinases known to be implicated in cancer biology
Figure imgf000045_0001
Kinase assay methods
Kinase assays were performed as described below for each individual kinase except the test compound was dissolved in DMSO and added to reaction mix to give final concentrations of 10μΜ and 100μ
MAPK2/ERK2 assay MAPK/ERK2 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na3V04, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against MBP in a final volume of 25.5 μΙ in 25 mM Tris pH 7.5, 0.1 mM EGTA, 0.33 mg/ml MBP, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ](500 -1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. MKK1 assay
This is a two-step assay where inactive MAPK (0.06 mg/ml) is activated by MKK1 (diluted in 25 mM Tris, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 0.01% Brij35, 1 mg/ml BSA) in 25.5 μΙ containing 25 mM Tris, 0.1 mM EGTA, 0.01% Brij35, 10 mM magnesium acetate and 0.005 mM ATP. After incubating at room temperature for 30 min, 5 μΙ from the first reaction is pipetted into 20 μΙ of the second reaction mix containing (final concentration) 25 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na3V04, 0.66 mg/ml myelin basic protein (MBP), 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ] (500 -1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
PKCa assay
PKCa (5-20 mU diluted in 20 mM Hepes pH 7.4, 0.03% Triton X-100) is assayed against Histone H1 in the presence of PtdSerine and DAG (0.1 mg/ml. and 10 pg/ml) and 0.1 mM CaCI2. The assay is carried out in a final volume of 25.5 μΙ containing 20 mM Hepes pH 7.4, 0.03% Triton X-100, 0.1 mg/ml Histone H1 , 10 mM magnesium acetate and 0.02 ητ)Μ[33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
PtdSer/DAG preparation:- PtdSer stock = 10 mg/ml in MeOH/Chloroform (1 :2). Dry down required amount. Resuspend in appropriate volume of 10 mM Hepes pH 7.4. Vortex and briefly sonicate. (2 x 10-15 seconds at 10-15 seconds apart). DAG stock = 10 mg/ml in MeOH/chloroform (1 :2). Dry down required amount. Add sonicated PtdSer solution. Vortex and sonicate. PDK1 assay
PDK1 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.05% β-mercaptoethanol, 1 mg/ml BSA) is assayed against PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQ-EMFRDFDYIADWC) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.05% β-mercaptoethanol, 100 μΜ substrate peptide, 10mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
APH-PKBa-S473D assay
APH-PKBa-S473D (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β- mercaptoethanol, 1 mg/ml BSA) is assayed against a modified Crosstide peptide GRPRTSSFAEGKK in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β- mercaptoethanol, 30 μΜ substrate peptide, 10 mM magnesium acetate and 0.005 mM
[33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
SGK assay
SGK (5-20mU diluted in 20 mM MOPS pH 7.5, 1mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against a modified Crosstide peptide GRPRTSSFAEGKK in a final volume of 25.5 μΙ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 30 μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33p-y-ATPJ (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
S6K1/ P70 S6K assay
S6K1/P70 S6K (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against substrate peptide (KKRNRTLTV) in a final volume of 25.5 μΙ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 0.1 mM substrate peptide, 10 mM magnesium acetate and 0.02 mM [33p-v- ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
ROCK-II (ROKa) assay
ROCK-II (ROKa) (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β- mercaptoethanol, 1 mg/ml BSA) is assayed against Long S6 substrate peptide (KEAKEKRQEQIAKRRRLSSLRASTSKSGGSQK) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 30 μΜ Long S6 substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
CHK1 assay CHK1 (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.1% β-mercaptoethanol, 0.01% Brij-35, 5% glycerol, 1 mg/ml BSA) is assayed against CHKtide substrate peptide (KKKVSRSGLYRSPSMPENLNRPR) in a final volume of 25.5 μΙ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 μΜ CHKtide, 10 mM magnesium acetate and 0.02 mM [33p- γ-ΑΤΡ](50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
LCK assay LCK (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% B-mercaptoethanol, 1 mg/ml BSA) is assayed against Cdc2 peptide (KVEKIGEGTYGWYK) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na3Vo4, Cdc2 peptide (0.25 mM), 10 mM magnesium acetate and 0.05mM [33ρ-γ-ΑΤΡ](500-1000 cpm/pmole) and incubated for 15 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
CDK2/cyclin A assay
CDK2/cyclin A (5-20 mU diluted in 50 mM Hepes pH 7.5, 1 mM DTT, 0.02% Brij35, 100 mM NaCI) is assayed against Histone H1 in a final volume of 25.5 μΙ containing 50 mM Hepes pH7.5, 1 mM DTT, 0.02% Brij35, 100 mM NaCI, Histone H1 (1 mg/ml), 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ](500-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. NEK2a assay
5-20mU of NEK2a (diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1mg ml BSA, 0.1%, β- mercaptoethanol) is assayed against NEK2a peptide (RFRRSRRMI) in a final volume of 25.5μΙ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.01% Brij, 0.1%, β- mercaptoethanol, 300μΜ NEK2a peptide, 10 mM magnesium acetate and 0.05 mM [33p- γ-ΑΤΡ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5μΙ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid.
MAPKAP-K1 b/RSK2 assay
MAPKAP-K1b (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% β-mercaptoethanol, 1mg/ml BSA) is assayed against substrate peptide (KKLNRTLSVA) in a final volume of 25.51 containing 50 mM Na- β-glycerophosphate (pH 7.5), 0.5 mM EDTA, 30 μΜ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
IKKb assay
5-20mU of IKKb (diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1mg/ml BSA, 0.1% β- mercaptoethanol) is assayed against substrate peptide (LDDRHDSGLDSMKDEEY) in a final volume of 25.5μΙ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.1%, ^mercaptoethanol, 300μΜ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33ρ-γ-ΑΤΡ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5μΙ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid.
Aurora B assay
Aurora B (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β- mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (LRRLSLGLRRLSLGLRRLSLGLRRLSLG) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
CHK2 assay
CHK2 (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.1% β-mercaptoethanol, 0.01% Brij-35, 5% glycerol, 1 mg/ml BSA) is assayed against CHKtide substrate peptide (KKKVSRSGLYRSPSMPENLNRPR) in a final volume of 25.5 μΙ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 μΜ CHKtide, 10 mM magnesium acetate and 0.02 mM [33p- γ-ΑΤΡ](50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
Src assay
Src (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (KVEKIGEGTYGWYK) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
PLK1 assay
PLK1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA, 100μΜ Vanadate) is assayed against a substrate peptide (ISDELMDATFADQEAKKK) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 10μΜ Vanadate, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid
PIM1 assay PIM1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (RSRHSSYPAGT) in a final volume of 25.5 pi containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. PIM3 assay
PIM3 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (RSRHSSYPAGT) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. FGF-R1 assay
FGF-R1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ- ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
EPH-A2 assay
EPH-A2 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.05 mM [33p- γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. IGF-1R assay
IGF-1R (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKSPGEYVNIEFG) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 300μΜ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. VEG-FR assay
VEG-FR (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKSPGEYVNIEFG) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 300μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
TBK1 (DU12569) assay
TBK1 (DU12569) (5-20mll diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKKERLLDDRHDSGLDSMKDEE) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 300μΜ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
IKKepsilon (DU14231) assay
5-20mU of IKKepsilon (DU14231)(diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1 mg/ml BSA) is assayed against MBP in a final volume of 25.5μΙ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.33mg/ml MBP, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5μΙ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid HER4 assay
HER4 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against Poly Glut Tyr in a final volume of 25.5μΙ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 1mg/ml Poly Glut Tyr, 10 mM magnesium acetate and 0.005mM [33p-y-ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
Aurora A assay
Aurora A (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β- mercaptoethanol, 1mg/ml BSA) is assayed against LRRLSLGLRRLSLGLRRLSLGLRRLSLG in a final volume of 25.5μΙ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 0.3mM LRRLSLGLRRLSLGLRRLSLGLRRLSLG, 10 mM magnesium acetate and 0.005mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
JAK2 assay
JAK2 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.05% β-mercaptoethanol, 1 mg/ml BSA) is assayed against PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQ- EMFRDFDYIADWC) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.05% β-mercaptoethanol, 100 μΜ substrate peptide, 10mM magnesium acetate and 0.005 mM [33p-y-ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
TrkA assay TrkA (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 10mM MnCI, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped- by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.-
Kinase Data Analysis
Raw data was converted into percent inhibition by using the following equation:
( Pmtest background
Percent Inhibition = 100 - (— — *
CPmcontrol ~ CPmb ckgro nd
Figure imgf000054_0001
The percent inhibition data was then modeled to the 4-parameter logistic equation:
Figure imgf000054_0002
{Compound Concentration
The values of Bottom, Top, Hill and EC50 were changed in order to give the best values to fit the experimentally obtained data.
Compounds were tested for activity at concentrations of 100μΜ and 10μΜ. Compounds were declared active if the activity at 100 μΜ exceeded a threshold of 50% above or below control level. Table 8 indicates which kinases are activated or inhibited by certain compounds.
Table 8
Figure imgf000054_0003
Compound Activated Kinases Inhibited Kinases
SGK, S6K1, CHK1,
Thiodicarb VEGFR, FGF R1
Buprofezin
PKBAph, Aurora B, PIM1 , SRC, TRKA,
Dinocap CHK1
RSK2, IKKb, PIM3, FGF R1
Thiop anate-methyl RSK2
NEK2a, TBK1 (DU12569), IKKepsilon (DU14231), Aurora B, VEGFR, PKCa,
Tolylfluanid Aurora A, TRKA, RSK2, IKKb, PLK1
(okadaic acid), FGF R1, HER4
Triazoxid
Tebufenpyrad S6K1
Triflusulfuron-methyl SGK, S6K1
SGK, ROCK-II, CDK2/cyclin A, IKKepsilon (DU14231), Aurora B, PIM1,
Fenpiclonil IGF-1R, TRKA, RSK2, IKKb, PIM3,
PLK1 (okadaic acid)
Cyprodinil PIM3
PDK1, PKBAph, MKK1, Aurora B, EPH brodifacoum S6K1, CHK1 A2 VEGFR, SRC, JAK2, TRKA, RSK2,
IKKb PLK1 (okadaic acid), HER4
PKBAph, SGK, KK1, Aurora B, flocoumafen S6K1, CHK1 VEGFR, PKCa, SRC, TRKA, RSK2,
IKKb, PLK1 (okadaic acid), HER4
Difenoconazol Aurora B, RSK2
kresoxim-methyl
Azamethiphos
Benfuracarb SGK, S6K1
Flusilazole
CBK1, VEGFR, PIM3,
fenpropathrin FGF R1
Fenbuconazol S6K1
Transfluthrin CHK1 MKK1
Fenazaquin
Fenoxycarb MKK1
Metconazole
Folicur
Methiocarb
yclobutanil
cyfluthrin CHK1, CHK2
Bromuconazol MKK1
Dimethomorph
Triazophos SGK VEGFR
phorate SGK RSK2
irgarol Compound Activated Kinases Inhibited Kinases
CHK1, VEGFR,
Pyriproxifen FGF R1
tau-fluvalinate CHK1, VEGFR
Fluoroglycofen-ethyl CHK1 , VEGFR
CHK1,
Esfenvalerate
FGF R1
Fenoxaprop-ethyl
PDK1 , SGK, S6K1,
cyhalofop-butyl CHK1, Aurora B,
PKCa, RSK2, PIM3
Flufenoxuron VEGFR
Dimefuron
Isoproturon
Ethoprophos SGK
7. In vitro Cytotoxic/Cytostatic Activity - Preferred that Potency be less than 200 uM on at least one cell line
Compounds must exhibit broad activity against a panel of cell lines from tumour types of varying origins. Effects on cellular proliferation were measured against 14 cell lines representing carcinomas of the lung, prostate, breast, liver, colon, pancreas as well as sarcomas. Cells were treated with 8 concentrations of compound 300, 100, 30, 10, 3, 1 , 0.3, 0.1, 0.03 and 0.01 μΜ and cell number estimated at each concentration. Cell number was calculated as a percentage survival relative to untreated cells. EC50 was calculated by fitting the data to the 4-parameter logistic model:
Figure imgf000056_0001
Table 9 details the transformed cell lines.
Table 9
Figure imgf000056_0002
Table 10 shows the cytotoxicity data (μΜ EC50s) for selected compounds against 14 tumour cell lines over a 7 day exposure
Table 10: Cytotoxicity data (μΜ) for selected compounds against 14 tumour cell lines
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Table 11- 72 hour Cell Proliferation Data at 100 μΜ Compound Concentration (data shown as cell number as % of untreated control)
Figure imgf000061_0001
ο
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
•J\
Figure imgf000067_0001
Figure imgf000068_0001
-J
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Table 12 - 7 day Cell Proliferation Data at 100 μΜ Compound Concentration (data shown as cell number as % of untreated control)
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
•J\
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
00
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
o
Table 13-182 compounds contained in the database according to screening step 1
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Table 14 - A panel of 144 compounds (chemically diverse) from screening step 2
Figure imgf000086_0001
Figure imgf000087_0001
Example 2 - Kinase screening
Table 15 shows a summary of the kinase screening data for fludioxonil and fenpiclonil. Results are represented as % inhibition relative to a positive control. Negative values represent compounds which activate kinase activity. Where (-) is shown there was no activity of the compound. Inhibition of the kinases SGK, ROCKII, PIM1 , TRKA, RSK2, PIM3 and FGFR1 was common to both fludioxonil and fenpiclonil.
Table15: Summary table of kinase inhibition for selected compounds
Figure imgf000088_0001
Example 3: Inhibition ofPIM Kinase activity PIM kinase inhibition has been investigated for each of PIM-1 , PIM-2 and PIM-3 kinase molecules.
PIM 1 (hr
The PIM-1 assay is performed using the Upstate IC50 Profiler Express™ service. In a final reaction volume of 25 μΙ, human recombinant PIM-1 (5-1 OmU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 100 μΜ KKRNRTLTV, 10mM MgAcetate and [v- 33P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required). After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 μΙ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
PIM-2 (h)
The PIM2 assay is performed using the Upstate IC50 Profiler Express™ service. In a final reaction volume of 25 μΙ, human recombinant PIM-2 (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 300 μΜ RSRHSSYPAGT, 10 mM MgAcetate and [?-33P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required). After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 μΙ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in menthanol prior to drying and scintillation counting.
PIM-3 (h)
The PIM-3 assay is performed using the Upstate IC50 Profiler Express™ service. In a final reaction volume of 25 μΙ, human recombinant PIM-3 (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 300 μΜ RSRHSSYPAGT, 10 mM MgAcetate and p-^P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required). After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 μΙ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in menthanol prior to drying and scintillation counting.
Results
Dose response against PIM kinase family members gave low μΜ IC50 (see figure 1). Figure 1a and 1b shows the inhibition of PI 1, PIM2 and PIM3 for fludioxonil and fenpiclonil respectively. The plots show % inhibition in an in vitro kinase assay. Table 16 details the IC50 and EC50 data for each compound against the PIM kinases.
Table 16
Figure imgf000090_0001
K562 cells express high levels of PIM1 protein. It is known that use of a PIM inhibitor can result in downregulation of PIM1 protein as shown in Swords et al. (SGI-1776: A novel PIM kinase inhibitor with potent preclinical activity against Acute Myeloid Leukemia (AML), Swords, R et al, Abstracts of the AACR Annual Meeting, 2009, April 18-22, Denver, CO, USA, Abstract umber 3743).
To test whether fenpiclonil and fludioxonil could down-regulate PIM, an assay was established using K562 cells. Test compounds are dissolved in 100% dimethylsulfoxide, diluted to the appropriated concentration and added to the culture medium of K562 cells. DMSO treated cells were used as a negative control. Compound CXR6099 was used as a positive control. The results are shown in figure 2 where fludioxonil and fenpiclonil can be seen to cause a reduction in PIM1 levels with increasing concentration.
Precise Method for Western Blot:
After trypsinisation and dilution in RPMI1640 medium, 1.4 x 106 K562 cells were transferred to a 60 mm culture dish, and cultured overnight. Following 24 h of treatment with different concentrations of compound, cells were collected and lysed. Protein level of PIM1 was evaluated by immunoblot analysis.
Briefly, proteins were transferred to a PVDF membrane and incubated for 1 hour at room temperature in blocking solution. Then the membranes were incubated overnight at 4°C in antibody solution containing anti-PIM1 (12H8) antibody (Santa Cruz Biotechnology) or containing anti-cleaved PARP antibody (Abeam pic). After washing, the membrans were incubated for 1 hour at room temperature in solution containing appropriate dilution of HRP-conjugated secondary antibody (GE Healthcare, UK).
ECL plus (GE Healthcare, UK) was used to detect PI 1 and cleaved PARP protein levels.
Example 4: Inhibition of ROCK activity
ROCK-II (ROKa) assay
ROCK-II (ROKa) (2.6 - 10.5 ng active Rock-ll diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against Long S6 substrate peptide (KEAKEKRQEQIAKRRRLSSLRASTSKSGGSQK) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 30 μΜ Long S6 substrate peptide, 10 mM magnesium acetate and 0.1 mM ATP (spiked with sufficient [33p-v- ATP] to give approximately 500-800 cpm/pmole) and incubated for 10 min at 30°C. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then 10 μΙ harvested onto P30 filter-mat and washed 3 times with 75 mM orthophosphoric acid, and once with methanol.
ROCK-I (ROKb) assay
ROCK-I (ROK) (72.7 - 200.5ng active Rock-I diluted in 20mM MOPS/NaOH pH 7.0, 1 mM EDTA, 0.01% Brij-35, 5% Glycerol, 0.1% β -mercaptoethanol, 1 mg/ml BSA) is assayed against 30μΜ Long S6 substrate peptide (KEAKEKRQEQIAKRRRLSSLRASTSKSGGSQK) in a final volume of 25 μΙ containing 8mM MOPS/NaOH pH 7.0, 0.2 mM EDTA, 30 μΜ Long S6 substrate peptide, 10 mM magnesium acetate and 0.1 mM ATP (spiked with sufficient [33p- γ - ATP] to give approximately 500-800 cpm/pmole) and incubated for 10 min at 30°C. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then 10 μΙ harvested onto P30 filter-mat and washed 3 times with 75 mM orthophosphoric acid, and once with methanol.
Results
Figure 3 show the increase in % inhibition of Rock I and Rock II with increasing does of fludioxonil or fenpiclonil. Table 17 details the IC50 and EC50 data for ROCK-I and ROCK-II using fludioxonil, fenpiclonil and pyrollnitrin. Table 17
Figure imgf000092_0001
Example 5: Cytotoxic effects of fludioxonil and fenpiclonil
Table 18 shows the 7 day IC50 (μΜ) of fludioxonil and fenpiclonal on a selection of cancer cell models.
Table 18
Figure imgf000092_0002
Cytotoxicity assay methods Cell Culture
Test compounds were dissolved in DMSO and dilution series performed. Cells were seeded at a density of 500 cells per well in 75 μΙ of DMEM (10% FBS/2 mM
Glutamine) media for 7 day incubation in 96-well plates. The following day, 25 μΙ of test compounds, diluted 1 :25 in DMEM (10% FBS), were added to the wells to give final compound concentrations as detailed above. Duplicate plates were assembled and then incubated for 7 days at 37°C/5% C02. No media/drug replacement during the 7 day period was undertaken.
For drug combination experiments and 72 hour cytoxicity testing on day one cells were seeded at a density of 5000 cells per well in 75 μΙ of DMEM (10% FBS/2 mM Glutamine) media in 96-well plates. The following day, 12.5 μΙ of each test compound, diluted 1 :50 in DMEM (10% FBS), was added to the wells to give final compound concentrations. Duplicate plates were assembled and then incubated for 3 days at 37°C/5% C02.
WST-1 Assay
Cell number was estimated using WST-1 reagent from Roche Applied Bioscience according to the manufacturer's instructions. Briefly 10μΙ of reagent was added to 100 μΙ of media and incubated until colour developed. The plates were read at wavelength 450nm on an appropriate absorbance plate reader.
Data Analysis
Data from the plate reader in text format was entered into an excel spreadsheet. The data was converted into a percentage value of the untreated control wells using the following formula:
Figure imgf000093_0001
The %Survival data was then modelled to the 4-parameter logistic equation:
Figure imgf000094_0001
Compound Concentration1
The values of Bottom, Top, Hill and EC50 were changed manually in order to give the best values to fit the experimentally obtained data.
Example 6 - Data from Xenograft study CXR0929
The aim of this study was to assess the efficacy of fludioxonil and fenpiclonil in vivo in nude mouse xenografts using mice harbouring tumours derived from human prostate (PC3).
The PC3 cell line was grown in culture and implanted into female nu-nu mice; dosing was initiated 72 hours later and tumour growth was monitored by calliper measurements. Mice harbouring PC3 tumour cells were dosed with ethanol/PEG200/water only or CXR6032 (Fludioxonil), or CXR6069 (Fenpiclonil). Compounds were to be administered daily for 28 days by oral gavage.
At termination, blood was harvested by cardiac puncture and mouse tissues and tumour pieces were fixed in NBF and also flash frozen.
Materials and methods Cell Line
The PC3 (ECACC catalogue number 90112714) cell line was resuscitated from stocks maintained at CXR Biosciences.
Safety Precautions
The normal safety precautions as detailed in the relevant SOPs and COSHH assessments applied, no additional precautions were considered necessary.
Animals Adult (6 - 8 weeks) female, athymic nude (nu/nu) mice were obtained from Charles River, UK.
Animal Accommodation and Husbandry
On arrival in the animal unit the mice were housed, up to 8 per cage, on sterile sawdust in sterile, solid-bottom, polypropylene cages.
The cages were individually vented units attached to a Techniplast Slimline Air Handling Unit. This unit maintains 70-80 air changes per cage, per hour, through HEPA air filters. Bedding was changed once weekly in a laminar flow unit. The temperature was maintained within a target range of 19-23° C and relative humidity of the IVC within a range of 40-70%. Twelve-hour periods of light were cycled with twelve-hour periods of darkness.
Diet
Sterile RM1 diet (Special Diet Services Ltd., Stepfield, Witham, Essex, UK) was used. The animal unit held the specification of the diet. Deionised water was autoclaved prior to use and changed at least once a week. Mice were allowed water and diet ad libitum and were acclimatised for at least 5 days prior to the study start.
Animal Health and Welfare
The mice have specific pathogen free (SPF)-status and the housing and changing system assured that the SPF-status was preserved during the study. Trained personnel under supervision handled the mice.
Experimental Design
Identification of Animals and Allocation ofnu-nu Mice into Experimental Groups
The animals were randomly allocated to groups on arrival. The mice were numbered appropriately and weighed prior to the start of the experiment. The first mouse assigned to a cage was individually identified by tail tattooing with the lowest number for that cage; the second mouse was assigned the second (lowest) number and so on. An experimental card was placed on each cage and showed the project licence code, test group, study number, sex and individual numbers of the mice within, and identified the Home Office Licensee. In addition, these cards were colour coded to correlate with the coding for the group.
Growth and Preparation of Cell Lines for Subcutaneous Injection into nu-nu Mice
PC3 cells were cultured in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% heat inactivated foetal calf serum (FCS) supplemented with 2mM L-glutamine, penicillin (50 lU/ml) and streptomycin (50μ9/πιΙ). Cultures were incubated in a humidified incubator at 37°C, 5% C02, until sufficient cells were available to implant the mice. Cells were harvested, pooled, centrifuged, and re-suspended in cold medium. This wwe mixed with an equal volume of cold Matrigel, so that the tumour cell injection solution was a 50:50 mixture of tumour cells/medium and Matrigel. Tumour cells were injected in a volume of 100μΙ in a single flank only. PC3 cells were injected at a density of 2.5 x 106 cells per flank. The cell/Matrigel suspension was aliquoted into pre-chilled Eppendorf tubes and kept on ice prior to injection. The period between preparation of the cell/Matrigel suspension and injection of tumour cells did not exceed two hours.
Implantation of Cell Lines
Cells were harvested as detailed above and 100μΙ of cell suspension was injected (s.c.) into a single flank of each mouse.
Repeat Dosing of Compounds
Mice were administered Test Compounds 72 hours following implantation of tumour cells. Mice harbouring PC3 tumour cells were dosed with CXR6032 (Fludioxonil), or CXR6069 (Fenpiclonil) in an ethanol/PEG200/water vehicle. Refer to Table 1 for the experimental design.
Compounds were administered daily for 28 days by oral gavage in a dose volume of 10ml/kg.
Table 19: Experimental Design
Figure imgf000096_0001
Experimental Procedures Bodyweight
The bodyweight of each mouse was recorded at the time of tail tattooing, before implantation and daily prior to dosing. Clinical Observations
Prior to the start of the study, all mice were observed to ensure that they were physically normal and that they exhibited normal activity. Only normal mice were allocated to the study. Following cell inoculation, each mouse was observed twice weekly and a general assessment of condition recorded in the study diary. Animals were terminated if tumours became ulcerated or if the Home Office Project License moderate severity limit was exceeded.
Tumour Measurement
Tumour growth was measured twice weekly for the duration of the study following cell implantation once tumours became palpable. Tumour diameters were measured at four different sites - two lengths and two widths - using a digital slide gauge. This was increased to three lengths and three widths if the tumour was an irregular shape.
Tumour volumes were calculated using the formula: 4/3 π
Figure imgf000097_0001
Where d = mean diameter (n
Animals were terminated if tumour volumes exceeded 1.44cm3 or if they became ulcerated.
Terminal Procedures
On the day of termination tumour volumes were recorded and the mice were weighed and transferred to the post mortem room. The animals were killed by exposure to a rising concentration of C02.
Blood was harvested by cardiac puncture and transferred to heparinised tubes for plasma preparation. Tumours and livers were processed as follows:
Tumours
• Each tumour was removed and the weights and gross morphology recorded. • The tumour was cut in half and one half placed in 10 % neutral buffered formalin (NBF) then prepared for paraffin embedding for possible future histochemical analysis.
• The remaining half was cut in two and each piece was placed in a separate cryovial and flash frozen. Cryovial 1 was labeled microarray and cryovial 2 was labeled MS. These samples were stored at approximately - 80°C for possible future analysis.
Livers
• The liver was removed and the weight recorded.
• 2 slices of liver (approximately 2 mm strips) were taken, one from the left lobe and one from the median lobe. These were placed in 10 % neutral buffered formalin (NBF) then prepared for paraffin embedding (section 6.4) for possible future histochemical analysis.
• 2 pieces of liver were placed in separate cryovials and flash frozen. Cryovial 1 was labeled 'Microarray' and cryovial 2 was labeled 'MS'. These samples were stored at approximately - 80°C for possible future analysis.
Laboratory Procedures Plasma Preparation
Following removal into tubes suitable for plasma preparation, venous blood samples were mixed on a roller for 10 minutes. Red blood cells were removed by centrifugation at 2,000 - 3000 rpm for 10 minutes at 8 - 10 °C. The supernatant (plasma) was transferred to a second tube and stored at approximately -70 °C until required for analysis.
Clinical Chemistry
Plasma was analyzed using a Cobas Integra 400 analyzer. Kits purchased from Roche Diagnostics GmbH were used to measure ALT (cat # 20764957322), AST (cat # 20764949322) and ALP (cat # 03333752190).
Plasma Levels of Test Compounds Plasma samples were stored appropriately and were analysed only with the agreement of the study leader.
Preparation of tissues for paraffin embedding
Tumour and liver samples were placed immediately into NBF for 36-45 hours. Tissues were processed according to internal standard operating procedures. The tissues were embedded in paraffin wax for possible future histochemical analysis.
Results
Figure 4 (a) shows the bodyweight over the duration of study in PC3 xenograft mice treated with fludioxonil (CXR6032), fenpiclonil (CXR6069) and vehicle control. Figure 4 (b) and (c) shows the terminal body and liver weights respectively from the same study.
Figure 5 shows (a) the Alanine transaminase (ALT), Aspartate transaminase (AST) and Alkaline phosphatase (ALP) activity levels and (c) tumour weight from the same study. Figure 5 (b) shows the tumour volume over time.
Example 7: Metabolism and pharmacokinetic properties of fludioxonil and fenpiclonil
P450 Inhibition Study (ref CXR0979) - Determination of P450 inhibition and IC50 using human microsomes
Microsomal incubations
Microsomal incubation mixtures (500pL) were prepared in HEPES buffer (50mM HEPES, 15mM MgCI2, 0.1 mM EDTA, pH 7.6) containing a NADPH regenerating system (1 mM NADPH, 10mM glucose-6-phosphate 1 IU glucose-6-phosphate dehydrogenase), pooled human liver microsomes (0.5mg protein), and 5pL of 7 concentrations and positive control inhibitors (see Table 21).
Table 20. Microsomal incubation conditions
Figure imgf000099_0001
HEPES buffer HEPES 57mM 50mM 440 PH7.6 MgCI2 17mM 15mM
EDTA 114μΜ 100μΜ
NADPH NADPH 20mM 1mM 25 regenerating
glucose-6- 200mM 10mM
system
phosphate
glucose-6- 20 IU 1 IU
phosphate
dehydrogenase
Microsomes pooled human liver 20mg/mL 1mg/mL 25 microsomes
Substrate Phenacetin 1mM 10μΜ 5 cocktail* in 50/50
Tolbutamide 10mM 100μΜ
(v/v)
Omeprazole 1mM 10μΜ
acetonitrile/water
Bufuralol 1mM 10μΜ
Midazolam 1mM 10μΜ
Figure imgf000100_0001
* CYP1A2 activity was monitored by phenacetin O-deethylation, CYP2C9 by tolbutamide hydroxylation, CYP2C19 by omeprazole hydroxylation, CYP2D6 by bufuralol l'-hydroxylation, and CYP3A4 by midazolam hydroxylation. Table 21. Positive inhibitor controls and Test Item concentrations
Figure imgf000101_0001
Figure imgf000101_0002
Serial dilutions of each Test Item were performed in the appropriate solvents as in Table 22 below:
Figure imgf000102_0001
MeCN = Acetonitrile
After addition of the Test Items or positive control inhibitors, the samples were pre- incubated at 37°C for approximately 5 minutes. Subsequently, the NADPH regenerating system was added to the reaction mixture and incubated for 20 minutes at 37°C. Reactions were terminated by the addition of 250μΙ_ of methanol containing 5μΜ dextrorphan as an internal standard.
Samples were vortexed briefly, placed on ice, and then centrifuged at approximately 3500 x g for 10 minutes to remove debris. The supernatant from each sample was then transferred to a separate vial for immediate LC-MS/MS analysis.
Mass Spectrometric analysis
Analysis to determine the concentration of the substrate metabolites was carried out using reverse phase HPLC with tandem mass spectrometric detection (LC-MS/MS). Multiple Reaction Monitoring (MRM) of the parent compound ion and specific fragment ion was monitored using a Micromass Quattro Micro Mass Spectrometer with Micromass MassLynx software version 4.0.
Data analysis
Analysis of the data was carried out using Micromass QuanLynx software Version 4.0 to determine the amount of the specific substrate metabolites produced in each sample (relative to the control samples). The results produced were plotted versus the different concentrations of the Test Items or positive inhibitor control compound, and the IC50 values calculated using Prism non-linear curve fitting software (version 4.03).
Table 23 - Test Item Results Summary
Data of potential relevance (<10μΜ) are highlighted in bold italics.
Figure imgf000103_0001
No inhibition over concentration range tested.
Conclusion
Three of the five Test Items inhibited (IC50 values below 10μΜ) one or more of the P450 enzymes tested. For this inhibition to be significant in the clinic the affected drug must have an appreciable proportion of its clearance via the P450 enzyme being inhibited i.e. fraction metabolised greater than 30%.
• Test Items Cyprodinil and Fludioxonil inhibited CYP1A2 with IC50 values of 0.621 μΜ and 1.23μΜ, respectively.
• Test Item Fluazinam inhibited CYP2C9 with an IC50 value of 0.967μΜ.
• Test Items Fludioxonil and Fenpiclonil show a similar trend in their P450 inhibition profiles reflecting their close structural relationship.
PK study (ref CXR 0966) Aim
The aim of this study was to examine the pharmacokinetics of fenpiclonil and fludioxonil administered by oral dosing to nude mice. In one treatment group, sampling took place over a 1 day period after a single oral dose of compound to obtain a 24hr post dose profile. In a second group the mice were repeat dosed, daily for 4 days, prior to PK sampling.
Method
Sufficient adult (6 - 8 weeks) female, athymic nude (nu/nu) mice were obtained from Charles River, UK. The mice were housed, up to 7 per cage, on sterile sawdust in sterile, solid-bottom, polypropylene cages. The cages were individually vented units attached to a Techniplast Slimline Air Handling Unit. This unit maintains 70-80 air changes per cage, per hour, through HEPA air filters. Bedding was changed once weekly in a laminar flow unit. The temperature was maintained within a target range of 19-23° C and relative humidity of the IVC within a range of 40-70%. Twelve-hour periods of light were cycled with twelve-hour periods of darkness.
The mice were fed with sterile RM1 diet (Special Diet Services Ltd., Stepfield, Witham, Essex, UK). Deionised water was autoclaved prior to use and changed at least once a week. Mice were allowed water and diet ad libitum and were acclimatised for at least 5 days prior to the study start.
Identification of Animals and Allocation ofnu-nu Mice into Experimental Groups
The animals were randomly allocated to groups on arrival. The mice were numbered appropriately and weighed prior to the start of the experiment. The first mouse assigned to a cage was individually identified by tail tattooing with the lowest number for that cage; the second mouse will be assigned the second (lowest) number and so on. An experimental card was placed on each cage and showed the project licence code, test group, study number, sex and individual numbers of the mice within, ans identified the Home Office Licensee. In addition, these cards were colour coded to correlate with the coding for the group.
Oral dosing
Oral dosing solutions of the Test Items were prepared at concentrations of 12mg/mL (for Fludioxonil and Fenpiclonil) in the dosing vehicle ethanol:PEG-200:water in the ratio 3:5:2 v/v. The Test Items were orally administered at a dose of 120mg/kg (for Fludioxonil and Fenpiclonil) all with a dosing volume of lOmlJkg. The bodyweight of each mouse was recorded immediately before each daily dose.
Oral pharmacokinetic samples from the 4 daily repeat dosed animals (not including control animals receiving a vehicle-only solution) were taken pre-dose (on day 4) and at 0.5, 1, 1.5, 2, 3, 4, 6 and 24 hours after administration of the final daily dose.
Oral pharmacokinetic samples from the single dosed animals were taken at 0.5, 1 , 1.5, 2, 3, 4, 6 and 24 hours after administration of the dose.
Whole blood pharmacokinetic samples
A pre-dose bleed was taken from the mice in the repeat dose groups prior to the fourth and final repeat dosing of each Test Item. Following the fourth daily oral administration of each Test Item the mice in the repeat dose groups were bled at approximately 0.5, 1 , 1.5, 2, 3, 4, 6 and 24 hours. Whole blood (0.020ml_) was taken from the tail vein of each animal and added to plastic sample vials containing milli Q water (0.020ml_) at each of the 9 timepoints. The whole blood:water samples were collected onto dry ice and stored at approximately -70°C prior to analysis.
Following a single oral administration of each Test Item, the mice were bled at approximately 0.5, 1 , 1.5, 2, 3, 4, 6 and 24 hours. Whole blood (0.020mL) was taken from the tail vein of each animal and added to plastic sample vials containing milli Q water (0.020ml_) at each of the 8 timepoints. The whole blood:water samples were collected onto dry ice and stored at approximately -70°C prior to analysis.
Terminal Procedures
At the time of termination the mice were transferred to the post mortem room or designated Laboratory. The animals were killed by exposure to a rising concentration of C02.
Blood was harvested by cardiac puncture and transferred to heparinised tubes for plasma preparation.
Plasma preparation
Following removal into tubes suitable for plasma preparation, venous blood samples were mixed on a roller for 10 minutes. Red blood cells were removed by centrifugation at 2,000 - 3000 rpm for 10 minutes at 8 - 10 °C. The supernatant (plasma) was transferred to a second tube and stored at approximately -70 °C until required for analysis.
LC-MS/MS analysis
Analysis to quantify the concentrations of each Test Item in the whole blood:water samples was carried out using reverse phase HPLC followed by mass spectrometric detection. Single analysis was performed.
Data Analysis
Blood concentration time data was analysed, where possible, using Pharsight WinNonLin software Version 5.2, for appropriate pharmacokinetic parameters such as half-life, bioavailability, Cmax, Tmax, AUC and clearance.
Result
Table 24: Plasma concentrations of fludioxonil (ng/ml) per animal for repeat dose and single dose groups
Figure imgf000106_0001
90.00 4016.30 5439.21 7147.80 5534.43 1567.92
120.00 4267.42 5344.50 8379.26 5997.06 2132.18
180.00 1612.34 1335.58 2165.80 1704.57 422.73
240.00 1426.10 2227.90 1866.42 1840.14 401.54
360.00 1621.26 1315.79 2886.68 1941.24 832.90
1440.00 43.31 45.84 45.19 44.78 1.31
Figure 6 shows graphically the changes in fludioxonil concentration over time for both single dose and for four daily repeat doses.
Table 25 - pharmacokinetic parameters following oral administration of fludioxonil
(repeat dose)
Figure imgf000107_0001
Table 26 - pharmacokinetic parameters following oral administration of fludioxonil
(single dose)
Figure imgf000107_0002
Number of point T1/2 7 6 5 6 1
Table 27: Plasma concentrations of fenpiclonil (ng/ml) per animal for repeat dose and single dose groups
Figure imgf000108_0001
Figure 7 shows graphically the changes in fenpiclonil concentration over time for both single dose and for four daily repeat doses. Table 28 - pharmacokinetic parameters following oral administration of fenpiclonil
(repeat dose)
Figure imgf000109_0001
Table 29 - pharmacokinetic parameters following oral administration of fenpiclonil (single dose)
Figure imgf000109_0002
Table 30 details the metabolism and pharmacokinetic properties of fludioxonil and fenpiclonil.
Table 30
Figure imgf000109_0003
Figure imgf000110_0001
mice
Example 8 - Microarray study:
A microarray study was undertaken with the 2 phenylpyrrole compounds. The aim of the study was to examine the effect of CXR6032 (Fludioxonil) and CXR6069 (Fenpiclonil) on the human tumour cell line PC3. PC3 had been chosen for this experiment because in xenograft studies, PC3 tumours showed a delayed growth response to CXR6032 and CXR6069.
Agilent Whole Genome Human Microarray 4x44K (G4122F) and Agilent 4x44K gasket slides G2534-60015 were used for the study.
Experimental Procedures Cell Culture experimental work Cell Line Culture Conditions
Cell lines were cultured according to the suppliers' specifications in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% heat inactivated foetal calf serum (FCS) supplemented with 2mM L-glutamine, penicillin (50 lU/ml) and streptomycin ^g/ml) at 37°C and 5% C02.
Plating Densities of PC3 Cells
Cells were plated into sterile, clear 6 well plates at densities of 3 x 105 cells per well, in DMEM medium containing 10% Foetal Calf Serum (FCS) and 2mM Glutamine. 4 plates, giving a total of 24 wells were plated. Dosing of Cells
Cells were incubated at 37°C and 5% C02 for 48 hours to allow the cells to attach. Once cells had reached ~75% confluency the tissue culture medium was changed as follows:
The tissue culture medium in all 6 wells on plate 1 was replaced with tissue culture medium containing 0.1% DMSO. CXR6032 and CXR6069 at the EC20 in tissue culture medium was added to 2 wells. CXR6032 and CXR6069 at the EC50 was added to the remaining 2 wells on each plate. The cells were cultured at 37°C and 5% C02 for a further 24 hours.
The EC2o and EC50 values to be used are specified in the Table 31 below:
Table 31: IC20 and IC50 concentrations for test items
Figure imgf000111_0001
Cell harvest
The medium was aspirated from all the wells and the cells washed in PBS. 700μΙ RLT buffer containing 10μΙ β-mercaptoethanol per 1ml buffer (Qiagen) was added to each well. The cells were lysed for 10 minutes on a gyro-rotator. The wells were scraped with a sterile cell scraper and lysed cells removed to labelled microtubes. RNA was isolated from cell samples according to CXR Method Sheet entitled "RNA isolation from cell lines".
Microarray experimental structure
The RNA microarray analysis involved labelled (one colour) RNA from triplicate samples of PC3 cell line treated vehicle, CXR6032, CXR6069 at EC20 and CXR6032, CXR6069 at EC5o as shown in figure 14.
Microarray experimental work Quality control ofRNA using the Agilent Bioanalyser
RNA integrity was checked using the Agilent bioanalyser and the RNA nano lab chip kit according to the CXR method sheet 'Set-up and Running of Nanochip Assay for RNA Quality Control'.
A potential problem was flagged at this point with sample 4-1 (Fenpiclonil EC20 replicate 1). The bioanalyser failed to produce a RIN for this sample. Visual examination of the electropherogram showed that the peaks were of similar appearance to all the samples that had produced a RIN of 10. The RNA concentration of this sample was approximately a third of the other samples, on the limit of the nanochip detection range. It was hypothesised that the low concentration may be why the bioanalyser program had been unable to calculate a RIN for this sample. It was decided to proceed with the labelling of all samples while remaining aware that this sample may fail to label.
Probe labelling
500ng RNA was labelled prior to microarray hybridisation using the Quick Amp Labelling Kit - Two Color (Agilent* 5190-0442), according to the CXR method sheet entitled "Transcriptional Profiling using Standard Agilent 1 Colour Protocol" v2.
Microarray Hybridisation and Scanning
Agilent whole genome human microarray 4x44K (G4122F were hybridised, washed and then scanned on an Agilent microarray scanner according to the CXR method sheet entitled, "Microarray hybridisation and scanning".
Microarray data processing to generate signature lists
Images from the scanner were processed using Agilent feature extraction software version 10.1.7. The data files were loaded into Rosetta resolver 6 software for tertiary analysis. The data were analysed to generate a "signature" gene list comprising the genes that are significantly regulated (p<0.001) according to the CXR method sheet entitled, "Microarray Data Entry and Signature List Production for One colour and Two-colour data in Resolver 6 Software". Signature lists of significantly altered genes were generated for the comparisons shown in Table 32. The lists were not filtered by fold change. The lists were named with the denominator name first followed by the test name and the number of genes in the list.
Table 32: Details signature lists generated. Denominator (D) Test (T) Number of lists
Vehicle vs. CXR6032 EC20 1
Vehicle vs. CXR6032 EC50 1
Vehicle vs. CXR6069 EC20 1
Vehicle vs. CXR6069 EC50 1
Bioinformatics
Bioinformatic analysis using Ingenuity pathways analysis™ (IPA) software was used to identify gene changes in pathways involving PIM kinase and the RhoA signalling pathway. A search was carried out in the ingenuity database under Genes and Chemicals for PIM and RhoA. Results for these searches included a list of all reported pathways that both genes are members of. Pathways were overlayed with the signature list data and tables of gene changes were produced.
Results
Treatment of cells with the test compounds at EC20 and EC50 doses caused ~500- ~4500 gene expression changes, Table 33. The top 25 up- and down-regulated genes in these 'signature' lists are shown in Appendices. Complete signature gene- list tables, available on an accompanying DVD, have not been included in this report due to their size.
Table 33: Data table showing the number of 'signature' genes changes for each treatment
Figure imgf000113_0001
Fold change data from the signature lists was overlaid on two PIM-associated pathways: granulocyte-macrophage colony stimulating factor signalling pathway (GM-CSF), Figures 8 and 9; Table 34, and acute myeloid leukemia signalling pathway (AML), Figures 10 and 11 ; Table 35. Fold change data was also overlaid on the RhoA signalling pathway, Figures 12 and 13; Table 36.
Data overlaid on the GM-CSF pathway for fludioxonil (FLU) and fenpiclonil (FEN) (EC50) show down-regulation of signal transducer and activator of transcription 1 , (STAT1). STAT1 regulates pim-1 oncogene (PIM1) at the level of transcription, (Yip- Schneider et al, 1995), although more recent data suggests that PIM1 regulation may be STAT1 independent (Ramana et al, 2002). Hence our results suggest that signalling via PIM1 may be suppressed by these compounds. It had been suggested that treatment with anti-cancer drugs can lead to the activation of survival pathways via PIM as a result of STAT3 activation, (Zemskova et al 2008). FLU (but not FEN), showed up-regulation of cyclin D1 and BCL2-like 11 apoptosis facilitator (Bcl-XL), which could indicate a compensatory effect to PIM inhibition.
Despite differences in the inhibitory effects of FLU and FEN at the level of PIM kinase activity there was no indication of large differences between the effects of the three compounds on the AML signalling pathway that features PIM (Figures 10 and 11). CCAAT/enhancer binding protein alpha, (C/EBPa), an inflammatory signalling transcription factor in the AML pathway, (Mackey et al, 2004), was significantly up- regulated by both compounds in a dose-dependent manner, (Figure 10 and 11 , Table 35).
FLU and FEN (EC50) microarray data (fold change values) overlaid on the RhoA Signalling pathway showed down-regulation of multiple genes. Both compounds led to the down-regulation of Rho associated kinase, (ROCK), spetin and actin-related protein complex (ARP2/3). Some of these genes represent interaction molecules comprising of several family members as reflected by a double-circle symbol in the IPA pathway diagrams. Not all members have been affected by the drug treatments in the same manner; hence a fold change is not shown in Table 36. Treatment with FLU also led to the down-regulation of p190 RhoGAP (RhoGAP), Rho guanine nucleotide exchange factor GEF 12 (LARG), phospholipase D1 (PLD1), citron rho- interacting, serine/threonine kinase 21 (Cit) and myosin light chain (MLC). Treatment with FEN led to down-regulation of lysophosphatidic acid receptor (LPAR), focal adhesion kinase (FAK), actin binding protein (ANLN) and ezrin (EZR). The majority of the genes down-regulated by FLU or FEN are involved in either cytokinesis or actin nucleation and polymerisation.
Conclusions
Both FLU and FEN led to a down-regulation of STAT1 , which regulates PIM1. This finding supports a previous study which suggested FLU and FEN inhibit PIM. The AML pathway, which also features PIM, was not greatly affected by either of the compounds with the exception that both compounds caused an increase in C/EBP expression that was dose-dependent.
FLU and FEN both caused the down regulation of multiple genes in the RhoA signalling pathway. All of the genes down-regulated are down-stream effectors of RhoA signalling, consistent with inhibition of Rho associated kinase.
The microarray data suggests that FLU and FEN have a similar mode of action.
Table 34: Data table showing gene changes in GM-CSF signalling pathway in CXR6032 and CXR6069 EC50 and EC20 signature lists.
Figure imgf000116_0001
Figure imgf000116_0002
Table 35: Data table showing gene changes in acute myeloid leukaemia signalling pathway in CXR6032 and CXR6069 EC50 and EC20 signature lists.
Figure imgf000117_0001
Figure imgf000117_0002
Table 36: Data table showing gene changes in RhoA signalling pathway in CXR6032 and CXR6069 EC50 and EC20 signature lists.
Figure imgf000118_0001
Figure imgf000118_0002
Table 37: Vehicle vs FLU CXR6032 EC2(>
Figure imgf000119_0001
Table 38: Vehicle vs FEN CXR6069 EC20
Figure imgf000120_0001
Table 39: Vehicle vs FLU CXR6032 EC50
Figure imgf000121_0001
Table 40: Vehicle vs FEN CXR6069 EC50
Figure imgf000122_0001
Example 9 - 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 directly to the required site. For example, Vitrasert releases ganciclovir directly into the eye to treat CMV retinitis. The direct application of this toxic agent to the site of disease 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 permeability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drug delivery.
Compounds of the invention 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 thermosensitive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into known, safe, biodegradable polymers. The active drug is delivered over time as the biopolymers dissolve.
Compounds of the invention 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 compounds to the cytoplasm and nucleus, and may be non-cell type specific and highly efficient (Derossi et al., 1998, Trends Cell Biol., 8, 84-87).
Preferably, the pharmaceutical formulation of the present invention is a unit dosage containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of the active ingredient.
The compounds of the invention can be administered by any parenteral route, in the form of a pharmaceutical formulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form. Depending upon the disorder and patient to be treated, as well as the route of administration, the compositions may be administered at varying doses.
In human therapy, the compounds of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical exipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
The compounds of the invention can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They 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.
Formulations 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 requiring 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 kind previously described.
For ophthalmic use, the compounds of the invention can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum. For application topically to the skin, the compounds of the invention can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
Generally, in humans, oral or parenteral administration of the compounds of the invention is the preferred route, being the most convenient.
For veterinary use, the compounds of the invention are administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
The formulations 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 ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. Preferred unit dosage formulations are those containing a daily dose or unit, daily sub- dose or an appropriate fraction thereof, of an active ingredient. A preferred delivery system of the invention may comprise a hydrogel impregnated with a polypeptides, polynucleotides and antibodies of the invention, which is preferably carried on a tampon which can be inserted into the cervix and withdrawn once an appropriate cervical ripening or other desirable affect on the female reproductive system†ias been produced.
It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question.
Example 10- Exemplary pharmaceutical formulations
Whilst it is possible for compounds of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, 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 pharmaceutical formulations according to the invention in which the active ingredient is a compound of the invention.
Example 10A: 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 into a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals.
Example 10B: Intramuscular injection
Active ingredient 0.20 g
Benzyl Alcohol 0.10 g
Glucofurol 75® 1.45 g
Water for Injection q.s. to 3.00 ml The active ingredient is dissolved in the glycofurol. The benzyl alcohol is then added and dissolved, and water added to 3 ml. The mixture is then filtered through a sterile micropore filter and sealed in sterile 3 ml glass vials (type 1). Example 10C: Tablet
Active ingredient 100 mg
Lactose 200 mg
Starch 50 mg
Polyvinylpyrrolidone 5 mg
Magnesium stearate 4 mg
359 mg
Tablets are prepared from the foregoing ingredients by wet granulation followed by compression.
Example 10D: Ophthalmic Solution
Active ingredient 0.5 g
Sodium chloride, analytical grade 0.9 g
Thiomersal 0.001 g
Purified water to 100 ml
pH adjusted to7.5
References:
Nishida er a/., J. Antibiot (Tokyo), Sep 18(5), 211-219, 1965. "Pyrrolnitrin, a new antifungal antibiotic. Microbiological and toxicological observations". Wente W. "Pyrrolnitrin for treatment of dermatomycoses and erythrasma in dermatologic practice", Mykosen., 20(12),463-70, 1977
Yamada M, Aoshima T, Yonezawa K. "Clinical experiences with pyrrolnitrin ointment and spray in dermatomycosis", Acta Dermatol Kyoto Engl Ed, 1968 Feb, (63(1), 75-82". No Abstract available. acotela Ruiz E, Lopez Martinez R, Lepe Santana G. "Therapeutic evaluation of pyrrolnitrin in ointment on dermatophytes and tinea versicolor", Prensa Med Mex. ,39(5- 6):294-297, 1974
Vanbreuseghem R, De Vroey C. n-vitro testing of fungistatic activity of pyrrolnitrin on pathogenic toadstools", Farmaco Prat., 32(12),617-33, 1977. Seidenari S, Di Nardo A, Motolese A, Pincelli C. "Erythema multiforme associated with contact sensitization. Description of 6 clinical cases", G Ital Dermatol Venereol., 125(1- 2), 35-40, 1990.
K Uchida, H Ymaguchi "Therapeutic efficacy of a topical antifungal solution preparation formulated with pyrrolnitrin and clotrimazole in combination (Pyroace W) in guinea pig model of tinea pedis", Jpn J Antibiot., 52(1), 68-74, 1999.
Tripathi, R.K. and Gottlieb, D. (1969) Mechanism of action of the antifungal antibiotic pyrrolnitrin. Journal of Bacteriology 100 (1): 310-328.
Arima, K., Imanaka, H., Kousaka, M., Fukuda, A and Tamura, G. (1965) Studies on pyrrolnitrin, a new antibiotic. I. Isoaltion and properties of pyrrolnitrin. J Antibiotics (Tokyo) 18(5):201-204.
Meneghini, C.L. and Angelini, G. "Contact dermatitis from pyrrolnitrin (an antimycotic agent)", Contact Dermatitis., 1(5),288-92, 1975. Lee et a/., 2004, Journal Biological Chemistry 279(3): 1885-1891
Romaguera, C. and Grimalt, F. "Five cases of contact dermatitis from pyrrolnitrine", Contact Dermatitis., (5), 352-3, 1980 Valsecchi, R., Foiadelli, L. and Cainelli, T. "Contact dermatitis from pyrrolnitrin.", Contact Dermatitis., 7(6),340, 1981
Meneghini, C.L. and Angelini, G. "Contact dermatitis from pyrrolnitrin." Contact
Dermatitis., 8(1), 55-8, 1982. Balato N, Lembo G, Cusano F, Ayala F. "Contact dermatitis from pyrrolenitrin." Contact Dermatitis., 9(3), 238, 1983.
Zhang et a/., 2007, Mol Cancer Res 5(9):909-922

Claims

Claims
1. A compound for use in medicine wherein the compound is a pyrrolnitrin derivative of formula I or a salt or ester thereof.
Formula I:
Figure imgf000130_0001
A = aromatic or heteroaromatic substituted with 1-3 N in the ring
B = aromatic or a partially or fully reduced five or six membered ring. Either A or B rings can be optionally benzo fused.
Q = bond, C1-4 alky, O, -(CH2)mO-(CH2)n- (m and n = 1 , 2), S, -(CH2)mS-(CH2)n- (m and n = 1 , 2), NR (R = H or C1-4 alkyl), or -(CH2)mNR-(CH2)n- (m and n = 1, 2; R = H or C1-4 alkyl) - Y = (CX)2l CX2) NH, NR (R = H or C1-4 alkyl), S, or O.
X = one or more substituents on ring A selected from: H, Halo (from F, CI, Br, I), C1-6 alkyl (linear and branched), C1-6 alkoxy, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyi (halo is 1-3 atoms from F, CI, Br, I), C1-4 haloalkoxy (halo is 1-3 atoms from F, CI, Br, I), OH, SR (R = H or C1-4 alkyl), NR2 (R = H or C1-4 alkyl), NRS02R (R = H or C1-4 alkyl), NRCOR (R = H or C1-4 alkyl), N02, CN, C02R (R = H or C1-4 alkyl), CONR2 (R = H or C1-4 alkyl), S02R (R = H or C1-4 alkyl), SOR (R = H or C1-4 alkyl), S02NR2 (R = H or C1-4 alkyl), OCOR (R = H or C1-4 alkyl), and COR (R = C1-4 alkyl). Ring A may also be optionally substituted by 2 O atoms that when are adjacent on the ring may together represent the moiety -O-CR'2-O- where R' = H, C1-4 alkyl or F.
Z = one or more substituents on ring B selected from: H, Halo (from F, CI, Br, I), C1-6 alkyl, C1-6 alkoxy, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyi (halo is 1-3 atoms from F, CI, Br, I), C1-4 haloalkoxy (halo is 1-3 atoms from F, CI, Br, I), OH, SR (R = H or C1-4 alkyl), NR2 (R = H or C1-4 alkyl), NRS02R (R = H or C1-4 alkyl), NRCOR (R = H or C1- 4 alkyl), N02, CN, C02R (R = H or C1-4 alkyl), CONR2 (R = H or C1-4 alkyl), S02R (R = H or C1-4 alkyl), SOR (R = H or C1-4 alkyl), S02NR2 (R = H or C1-4 alkyl), OCOR (R = H or C1-4 alkyl), and COR (R = C1-4 alkyl).
2. A pharmaceutical composition comprising a pyrrolnitrin derivative of formula I or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier.
3. A method of treating cancer comprising administering to the patient an effective amount of a pyrrolnitrin derivative of formula I or a salt or an ester thereof.
4. A compound for use in the treatment of cancer wherein the compound is a pyrrolnitrin derivative of formula I or a salt or an ester thereof.
5. The compound for use described in claim 4 comprising administering to a patient an effective amount of a pyrrolnitrin derivative of formula I or a salt or an ester thereof.
6. Use of a pyrrolnitrin derivative of formula I or a salt or an ester thereof in the manufacture of a medicament for the treatment of cancer.
7. A method of treating a disease selected from vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma and osteoporosis, erectile dysfunction and other indications mediated by Rho-kinase, e.g., coronary heart disease comprising administering to the patient an effective amount of a pyrrolnitrin derivative of formula I or a salt or an ester thereof.
8. A compound for use in the treatment of a disease selected from vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma, osteoporosis, reperfusion injury, asthma, fibrosis, anti-inflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, erectile dysfunction and other indications mediated by Rho-kinase, e.g., coronary heart disease, wherein the compound is a pyrrolnitrin derivative of formula I or a salt or an ester thereof
9. The compound described in claim 8 comprising administering to a patient an effective amount of a pyrrolnitrin derivative of formula I or a salt or an ester thereof.
10. Use of a pyrrolnitrin derivative of formula I or a salt or an ester thereof in the manufacture of a medicament for the treatment of a disease selected from vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma, osteoporosis, reperfusion injury, asthma, fibrosis, anti-inflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, erectile dysfunction and other indications mediated by Rho-kinase, e.g., coronary heart disease.
11. The use described in claim 10 comprising administering to a patient an effective amount of a pyrrolnitrin derivative of formula I or a salt or an ester thereof.
12. The compound, method or use of any of claims 3-6 wherein the cancer is selected from pancreatic cancer, ovarian cancer, breast cancer, prostate cancer, liver cancer, chondrosarcoma, lung cancer, head and neck cancer, colon cancer, sarcoma, leukaemia, lymphoma, myeloma, kidney cancer, thyroid cancer and brain cancers such as glioblastoma.
13. The compound, method or use of any of claims 1-12 wherein the patient is selected from humans, cows, dogs, cats, goats, sheep, and pigs.
14. The compound, method or use of any of claims 1-13 wherein the patient is a human.
15. The compound, method or use of any of previous claim wherein the pyrrolnitrin derivative is fludioxonil or a salt or an ester thereof.
16. The compound, method or use of any previous claim wherein the pyrrolnitrin derivative is fenpiclonil or a salt or an ester thereof.
17. The compound, method, use or composition of any previous claim additionally comprising a further chemotherapeutic agent.
18. The compound, method, use or composition of claim 14 wherein the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7- hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine , Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Eriotinib, Estramustine, Etoposide (V16-213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1, Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha-2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541 , NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474,4HC,6-[4-(2-Piperidin-1- ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1 ,5-a]pyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat.
19. The compound, composition, method or use of claims 3-18 wherein the patient to be treated is not amenable to conventional chemotherapeutic agents.
20. A kit of parts comprising:
(i) pyrrolnitrin derivative of formula I or a salt or ester thereof or a pharmaceutical composition comprising a pyrrolnitrin derivative of formula I or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier;
(ii) apparatus for administering the compound or pharmaceutical composition; and (iii) instructions for use.
21. The kit of claim 20 additionally comprising a further chemotherapeutic agent.
22. The kit of claim 21 wherein the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D,
Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine , Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16- 213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1 , Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha- 2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541 , NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxei, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Tenyposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, Y 155, ZSTK474,4HC,6-[4-(2-Piperidin-1-ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1 ,5- a]pyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat.
23. A compound substantially defined herein with reference to the description for use in medicine.
24. A compound substantially defined herein with reference to the description for use in the treatment of cancer.
25. A compound substantially defined herein with reference to the description for use in the treatment of a disease selected from vasospasm, pulmonary hypertension, post operative scar formation, multiple sclerosis, hypertension (such as pulmonary hypertension), atherosclerosis, restenosis, cerebral ischemia, neuronal degeneration, nerve (such as spinal cord) injury, thrombotic disorders, asthma, glaucoma, osteoporosis, reperfusion injury, asthma, fibrosis, anti-inflammatory diseases, HIV infection, hearing loss, tinnitus, macular degeneration, erectile dysfunction and other indications mediated by Rho-kinase, e.g., coronary heart disease.
26. A method of treating cancer substantially defined herein with reference to the description.
27. A pharmaceutical composition substantially defined herein with reference to the description
28. A kit of parts substantially defined herein with reference to the description.
PCT/GB2012/050937 2011-04-28 2012-04-27 Pyrrolnitrin derivatives WO2012146936A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1107176.8A GB201107176D0 (en) 2011-04-28 2011-04-28 Pyrrolnitrin derivatives
GB1107176.8 2011-04-28

Publications (1)

Publication Number Publication Date
WO2012146936A1 true WO2012146936A1 (en) 2012-11-01

Family

ID=44202940

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2012/050937 WO2012146936A1 (en) 2011-04-28 2012-04-27 Pyrrolnitrin derivatives

Country Status (2)

Country Link
GB (1) GB201107176D0 (en)
WO (1) WO2012146936A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104803914A (en) * 2015-03-05 2015-07-29 成都理工大学 Hexahydro-azepine oxy-benzamide compounds serving as Rho kinase inhibitor
US9200004B2 (en) 2013-01-15 2015-12-01 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9278950B2 (en) 2013-01-14 2016-03-08 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
USRE46117E1 (en) 1999-12-22 2016-08-23 Teva Pharmaceuticals International Gmbh Modulators of dopamine neurotransmission
US9458128B2 (en) 2012-05-24 2016-10-04 Orion Corporation Catechol O-methyltransferase activity inhibiting compounds
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9556197B2 (en) 2013-08-23 2017-01-31 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US9580418B2 (en) 2014-07-14 2017-02-28 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
CN108137595A (en) * 2015-06-18 2018-06-08 左坚 Prevent and treat the method and composition of hearing loss
CN110327465A (en) * 2019-07-06 2019-10-15 五邑大学 A kind of compound antiprostate cancer
WO2020049208A1 (en) * 2018-09-09 2020-03-12 Fundacio Privada Institut De Recerca De La Sida - Caixa Aurora kinase as target for treating, preventing or curing hiv infection or aids
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
CN110974828A (en) * 2019-12-24 2020-04-10 苏州大学 Application of compound Axitinib in preparation of medicine for treating cerebrovascular diseases and pharmaceutical composition of compound Axitinib
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
CN115611927A (en) * 2020-01-06 2023-01-17 刘力 New compound of vinca antitumor drug
US11613548B2 (en) 2021-02-19 2023-03-28 Sudo Biosciences Limited Substituted pyridines, pyridazines, pyrimidines, and 1,2,4-triazines as TYK2 inhibitors
WO2023056295A1 (en) * 2021-09-30 2023-04-06 Regeneron Pharmaceuticals, Inc. Treatment of glaucoma with rho guanine nucleotide exchange factor 12 (arhgef12) inhibitors
US11857551B1 (en) 2020-07-10 2024-01-02 Ting Therapeutics Llc Methods for the prevention and treatment of hearing loss
EP4126014A4 (en) * 2020-03-23 2024-04-17 Univ Texas Combination therapeutics using tumor treating fields (ttfields)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0018737A1 (en) 1979-04-11 1980-11-12 Mitsui Petrochemical Industries, Ltd. Process for producing spherical carrier particles for olefin polymerisation catalyst, catalyst comprising such carrier particles, and use of such catalyst in olefin polymerisation
JPH07147016A (en) 1993-11-24 1995-06-06 Matsushita Electric Ind Co Ltd In-line sputtering device
WO2000057913A1 (en) 1999-03-25 2000-10-05 Welfide Corporation Preventives/remedies for interstitial pneumonia and pulmonary fibrosis
EP1057485A1 (en) * 1998-01-14 2000-12-06 Nippon Shinyaku Co., Ltd. Potassium channel activators
WO2000078351A1 (en) 1999-06-18 2000-12-28 Mitsubishi Pharma Corporation Osteogenesis promoters
WO2001032156A2 (en) * 1999-11-02 2001-05-10 Dalhousie University Methods for treating fibroproliferative diseases
US20020165594A1 (en) * 2001-01-19 2002-11-07 Merrill A. Biel Apparatus and method of photodynamic eradication of organisms utilizing pyrrolnitrin
US20080220103A1 (en) * 2005-10-24 2008-09-11 Jay Birnbaum Method for treating/controlling/killing fungi and bacteria on living animals
WO2009062746A2 (en) * 2007-11-17 2009-05-22 Bayer Consumer Care Ag Topical drugs for use in antifungal therapy
WO2011146031A1 (en) * 2010-05-18 2011-11-24 Bilgic Mahmut Pharmaceutical composition comprising n- acetylcysteine and a xanthine

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0018737A1 (en) 1979-04-11 1980-11-12 Mitsui Petrochemical Industries, Ltd. Process for producing spherical carrier particles for olefin polymerisation catalyst, catalyst comprising such carrier particles, and use of such catalyst in olefin polymerisation
JPH07147016A (en) 1993-11-24 1995-06-06 Matsushita Electric Ind Co Ltd In-line sputtering device
EP1057485A1 (en) * 1998-01-14 2000-12-06 Nippon Shinyaku Co., Ltd. Potassium channel activators
WO2000057913A1 (en) 1999-03-25 2000-10-05 Welfide Corporation Preventives/remedies for interstitial pneumonia and pulmonary fibrosis
WO2000078351A1 (en) 1999-06-18 2000-12-28 Mitsubishi Pharma Corporation Osteogenesis promoters
WO2001032156A2 (en) * 1999-11-02 2001-05-10 Dalhousie University Methods for treating fibroproliferative diseases
US20020165594A1 (en) * 2001-01-19 2002-11-07 Merrill A. Biel Apparatus and method of photodynamic eradication of organisms utilizing pyrrolnitrin
US20080220103A1 (en) * 2005-10-24 2008-09-11 Jay Birnbaum Method for treating/controlling/killing fungi and bacteria on living animals
WO2009062746A2 (en) * 2007-11-17 2009-05-22 Bayer Consumer Care Ag Topical drugs for use in antifungal therapy
WO2011146031A1 (en) * 2010-05-18 2011-11-24 Bilgic Mahmut Pharmaceutical composition comprising n- acetylcysteine and a xanthine

Non-Patent Citations (66)

* Cited by examiner, † Cited by third party
Title
A DRAPER ET AL: "Occupational asthma from fungicides fluazinam and chlorothalonil", OCCUP ENVIRON MED, 1 January 2003 (2003-01-01), pages 76 - 77, XP055035015, Retrieved from the Internet <URL:http://oem.bmj.com/content/60/1/76.full.pdf#page=1&view=FitH> [retrieved on 20120808] *
AMANO ET AL., EXP CELL RES, vol. 261, 2000, pages 44
ARIMA, K.; IMANAKA, H.; KOUSAKA, M.; FUKUDA, A; TAMURA, G.: "Studies on pyrrolnitrin, a new antibiotic. I. Isoaltion and properties of pyrrolnitrin", J ANTIBIOTICS (TOKYO, vol. 18, no. 5, 1965, pages 201 - 204
ASPENSTROM CURR OPIN CELL BIOL, vol. 11, 1999, pages 95
BALATO N; LEMBO G; CUSANO F; AYALA F.: "Contact dermatitis from pyrrolenitrin.", CONTACT DERMATITIS., vol. 9, no. 3, 1983, pages 238
BUHL ET AL., J BIOL CHEM, vol. 270, 1995, pages 24631
CHELLAIAH ET AL., JBIOL CHEM, vol. 275, 2000, pages 11993
CHITALEY ET AL., NAT MED, vol. 7, 2001, pages 119
DEROSSI ET AL., TRENDS CELL BIOL., vol. 8, 1998, pages 84 - 87
ETO ET AL., AM JPHYSIOL HEART CIRC PHYSIOL, vol. 278, 2000, pages H 1744
FRESHNEY: "Culture of Animal Cells: A Manual of Basic Technique", 1987, WILY-LISS
FUKATA ET AL., J CELL BIOL, vol. 141, 1998, pages 409
FUKATA ET AL., TRENDS PHARCOL SCI, vol. 22, 2001, pages 32
GINGRAS ET AL., BIOCHEM J, vol. 348, 2000, pages 273
GIOVANELLA ET AL., J. NATL. CAN. INST, vol. 52, 1974, pages 921 - 30
GOETZL ET AL., CANCER RES, vol. 59, 1999, pages 4732
HARA ET AL., J NEUROSURG, vol. 93, 2000, pages 94
HITOMI ET AL., LIFE SCI, vol. 67, 2000, pages 1929
HODGE; STERNER, J IND HYG TOXICOL., vol. 31, no. 2, March 1949 (1949-03-01), pages 79 - 92
HONJO ET AL., INVEST OPHTHALMOL VIS SCI, vol. 42, 2001, pages 137
HORI Y ET AL: "WB2838 Ä3-CHLORO-4-(2-AMINO-3-CHLOROPHENYL)-PYRROLEÜ: NON-STEROIDAL ANDROGEN-RECEPTOR ANTAGONIST PRODUCED BY A PSEUDOMONAS", JOURNAL OF ANTIBIOTICS, JAPAN ANTIBIOTICS RESEARCH ASSOCIATION, TOKYO, JP, vol. 46, no. 9, 1 January 1993 (1993-01-01), pages 1327 - 1333, XP008052470, ISSN: 0021-8820 *
ITOH ET AL., NAT MED, vol. 5, 1999, pages 221
K UCHIDA; H YMAGUCHI: "Therapeutic efficacy of a topical antifungal solution preparation formulated with pyrrolnitrin and clotrimazole in combination (Pyroace W) in guinea pig model of tinea pedis", JPN J ANTIBIOT., vol. 52, no. 1, 1999, pages 68 - 74
KAWAGUCHI ET AL., EUR JPHARMACOL, vol. 403, 2000, pages 203
KIM ET AL., NEUROSURGERY, vol. 46, 2000, pages 440
KLAGES ET AL., I CELL BIOL, vol. 144, 1999, pages 745
LEE ET AL., JOURNAL BIOLOGICAL CHEMISTRY, vol. 279, no. 3, 2004, pages 1885 - 1891
LI ET AL., CLIN. EXP. METASTASIS, vol. 17, 1999, pages 423 - 9
LIAO ET AL., J CARDIOVASCULAR PHARMACOL 2007, vol. 50, no. 1, 2010, pages 17 - 24
MACKAY ET AL., JBIOL CHEM, vol. 273, 1998, pages 20685
MACKAY; HALL, J BIOL CHEM, vol. 273, 1998, pages 20685
MACOTELA RUIZ E; LOPEZ MARTINEZ R; LEPE SANTANA G.: "Therapeutic evaluation of pyrrolnitrin in ointment on dermatophytes and tinea versicolor", PRENSA MED MEX., vol. 39, no. 5-6, 1974, pages 294 - 297
MATSUI ET AL., EMBO J, vol. 15, 1996, pages 2208
MENEGHINI, C.L.; ANGELINI, G.: "Contact dermatitis from pyrrolnitrin (an antimycotic agent", CONTACT DERMATITIS., vol. 1, no. 5, 1975, pages 288 - 92
MENEGHINI, C.L.; ANGELINI, G.: "Contact dermatitis from pyrrolnitrin.", CONTACT DERMATITIS., vol. 8, no. 1, 1982, pages 55 - 8
NAKAHARA ET AL., EUR J PHARMACOL, vol. 389, 2000, pages 103
NEGORO ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 262, 1999, pages 211
NISHIDA ET AL., J. ANTIBIOT (TOKYO, vol. 18, no. 5, September 1965 (1965-09-01), pages 211 - 219
PILKINGTON ET AL., ANTICANCER RES., vol. 17, 1997, pages 4107 - 9
RETZER ET AL., CELL SIGNAL, vol. 12, 2000, pages 645
RETZER ET AL., FEBS LETT, vol. 466, 2000, pages 70
RIBATTA ET AL., INTI. J. DEV. BIOL., vol. 40, 1999, pages 1189 - 97
ROMAGUERA, C.; GRIMALT, F.: "Five cases of contact dermatitis from pyrrolnitrine", CONTACT DERMATITIS., no. 5, 1980, pages 352 - 3
SAHAI ET AL., CURR BIOL, vol. 9, 1999, pages 136
SANCHEZ-MADRID ET AL., EMBO J, vol. 18, 1999, pages 501
SATO ET AL., CIRC RES, vol. 87, 2000, pages 195
SEASHOLTZ ET AL., CIRC RES, vol. 84, 1999, pages 1186
SEIDENARI S; DI NARDO A; MOTOLESE A; PINCELLI C.: "Erythema multiforme associated with contact sensitization. Description of 6 clinical cases", G ITAL DERMATOL VENEREOL., vol. 125, no. 1-2, 1990, pages 35 - 40
SETOGUCHI ET AL., BR J PHARMACOL, vol. 132, 2001, pages 111
SOMLYO ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 269, 2000, pages 652
SOMLYO ET AL., J PHYSIOL (LOND, vol. 522, 2000, pages 177
SUMI ET AL., J BIO CHEM, vol. 276, 2001, pages 670
TOSHIMA ET AL., STROKE, vol. 31, 2000, pages 2245
TREVAN: "The error of determination of toxicity", PROC ROY SOC, vol. 101B, 1927, pages 483 - 514
TRIPATHI, R.K.; GOTTLIEB, D.: "Mechanism of action of the antifungal antibiotic pyrrolnitrin", JOURNAL OF BACTERIOLOGY, vol. 100, no. 1, 1969, pages 310 - 328
UCHIDA ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 269, 2000, pages 633
UEHATA ET AL., NATURE, vol. 389, 1997, pages 990
VALSECCHI, R.; FOIADELLI, L.; CAINELLI, T.: "Contact dermatitis from pyrrolnitrin.", CONTACT DERMATITIS., vol. 7, no. 6, 1981, pages 340
VAN AELST ET AL., GENES DEV, vol. 11, 1997, pages 2295
VANBREUSEGHEM R; DE VROEY C: "In-vitro testing of fungistatic activity of pyrrolnitrin on pathogenic toadstools", FARMACO PRAT., vol. 32, no. 12, 1977, pages 617 - 33
WENTE W.: "Pyrrolnitrin for treatment of dermatomycoses and erythrasma in dermatologic practice", MYKOSEN, vol. 20, no. 12, 1977, pages 463 - 70, XP008154434
WENTE W: "[Pyrrolnitrin for treatment of dermatomycoses and erythrasma in dermatologic practice].", MYKOSEN DEC 1977 LNKD- PUBMED:600302, vol. 20, no. 12, December 1977 (1977-12-01), pages 463 - 470, XP008154434, ISSN: 0027-5557 *
YAMADA M; AOSHIMA T; YONEZAWA K: "Clinical experiences with pyrrolnitrin ointment and spray in dermatomycosis", ACTA DERMATOL KYOTO ENGL ED, vol. 63, no. 1, February 1968 (1968-02-01), pages 75 - 82
YAMAMOTO ET AL., J CARDIOVASC PHARMACOL, vol. 35, 2000, pages 203
ZHANG ET AL., J CELL SCI, vol. 108, 1995, pages 2285
ZHANG ET AL., MOL CANCER RES, vol. 5, no. 9, 2007, pages 909 - 922

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE46117E1 (en) 1999-12-22 2016-08-23 Teva Pharmaceuticals International Gmbh Modulators of dopamine neurotransmission
US9458128B2 (en) 2012-05-24 2016-10-04 Orion Corporation Catechol O-methyltransferase activity inhibiting compounds
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9278950B2 (en) 2013-01-14 2016-03-08 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9200004B2 (en) 2013-01-15 2015-12-01 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10517858B2 (en) 2013-01-15 2019-12-31 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as PIM kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9849120B2 (en) 2013-01-15 2017-12-26 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10265307B2 (en) 2013-01-15 2019-04-23 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10828290B2 (en) 2013-01-15 2020-11-10 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as pim kinase inhibitors
US11229631B2 (en) 2013-01-15 2022-01-25 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9556197B2 (en) 2013-08-23 2017-01-31 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US9580418B2 (en) 2014-07-14 2017-02-28 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
CN104803914B (en) * 2015-03-05 2017-11-14 成都理工大学 Hexahydro azatropylidene oxybenzamide class compound as Rho kinase inhibitors
CN104803914A (en) * 2015-03-05 2015-07-29 成都理工大学 Hexahydro-azepine oxy-benzamide compounds serving as Rho kinase inhibitor
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9802918B2 (en) 2015-05-29 2017-10-31 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
CN108137595A (en) * 2015-06-18 2018-06-08 左坚 Prevent and treat the method and composition of hearing loss
EP3310783B1 (en) * 2015-06-18 2020-11-04 Ting Therapeutics LLC Compositions for the prevention of hearing loss
US10336728B2 (en) 2015-09-09 2019-07-02 Incyte Corporation Salts of a Pim kinase inhibitor
US11505540B2 (en) 2015-09-09 2022-11-22 Incyte Corporation Salts of a Pim kinase inhibitor
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US11066387B2 (en) 2015-09-09 2021-07-20 Incyte Corporation Salts of a Pim kinase inhibitor
US11053215B2 (en) 2015-10-02 2021-07-06 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US10450296B2 (en) 2015-10-02 2019-10-22 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
WO2020049208A1 (en) * 2018-09-09 2020-03-12 Fundacio Privada Institut De Recerca De La Sida - Caixa Aurora kinase as target for treating, preventing or curing hiv infection or aids
CN110327465B (en) * 2019-07-06 2021-12-14 五邑大学 Compound anti-prostatic cancer medicine
CN110327465A (en) * 2019-07-06 2019-10-15 五邑大学 A kind of compound antiprostate cancer
CN110974828A (en) * 2019-12-24 2020-04-10 苏州大学 Application of compound Axitinib in preparation of medicine for treating cerebrovascular diseases and pharmaceutical composition of compound Axitinib
CN110974828B (en) * 2019-12-24 2023-03-31 苏州大学 Application of compound Axitinib in preparation of medicine for treating cerebrovascular diseases and pharmaceutical composition of compound Axitinib
CN115611927A (en) * 2020-01-06 2023-01-17 刘力 New compound of vinca antitumor drug
EP4126014A4 (en) * 2020-03-23 2024-04-17 Univ Texas Combination therapeutics using tumor treating fields (ttfields)
US11857551B1 (en) 2020-07-10 2024-01-02 Ting Therapeutics Llc Methods for the prevention and treatment of hearing loss
US11613548B2 (en) 2021-02-19 2023-03-28 Sudo Biosciences Limited Substituted pyridines, pyridazines, pyrimidines, and 1,2,4-triazines as TYK2 inhibitors
WO2023056295A1 (en) * 2021-09-30 2023-04-06 Regeneron Pharmaceuticals, Inc. Treatment of glaucoma with rho guanine nucleotide exchange factor 12 (arhgef12) inhibitors

Also Published As

Publication number Publication date
GB201107176D0 (en) 2011-06-15

Similar Documents

Publication Publication Date Title
WO2012146936A1 (en) Pyrrolnitrin derivatives
WO2012146933A1 (en) Cyprodinil for use in medicine
US10233157B2 (en) SIK inhibitor for use in a method of treating an inflammatory and/or immune disorder
US20230346786A1 (en) Chiral diaryl macrocycles and uses thereof
Zebisch et al. Signaling through RAS-RAF-MEK-ERK: from basics to bedside
Vargas et al. Inhibitors of BTK and ITK: state of the new drugs for cancer, autoimmunity and inflammatory diseases
Garcia-Echeverria et al. Drug discovery approaches targeting the PI3K/Akt pathway in cancer
Das et al. Pro-survival autophagy: An emerging candidate of tumor progression through maintaining hallmarks of cancer
Drygin et al. Potential Use of Selective and Nonselective Pim Kinase Inhibitors for Cancer Therapy: Miniperspective
JP6479812B2 (en) Combination of an ALK inhibitor and a CDK inhibitor to treat a cell proliferative disorder
Warkentin et al. Overcoming myelosuppression due to synthetic lethal toxicity for FLT3-targeted acute myeloid leukemia therapy
Howe et al. Focal adhesion kinase inhibitors in combination with erlotinib demonstrate enhanced anti-tumor activity in non-small cell lung cancer
US20160058741A1 (en) Thiadiazolidinone Derivatives
AU2014288857A1 (en) A pharmaceutical combination for the treatment of melanoma
KR20160020502A (en) Pharmaceutical combinations
Wu et al. CDK9 inhibitors reactivate p53 by downregulating iASPP
Stauffer et al. Blocking the PI3K/PKB pathway in tumor cells
Zhelev et al. From Roscovitine to CYC202 to Seliciclib–from bench to bedside: discovery and development
US9629851B2 (en) ROCK in combination with MAPK pathway
WO2017117386A1 (en) Methods of treating cancer using network brakes
Wu et al. Discovery of Selective and Potent Macrocyclic CDK9 Inhibitors for the Treatment of Osimertinib-Resistant Non-Small-Cell Lung Cancer
WO2020227202A1 (en) Compositions and methods for treating cancer
WO2018204987A1 (en) Carbonic anhydrase inhibitors
US20230158034A1 (en) Co-treatment with cdk4/6 and cdk2 inhibitors to suppress tumor adaptation to cdk2 inhibitors
Raja GSK3B regulates epithelial-mesenchymal transition and cancer stem cell properties and is a novel drug target for triple-negative breast cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12725869

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12725869

Country of ref document: EP

Kind code of ref document: A1