CA2738025A1 - Protease inhibitors - Google Patents

Protease inhibitors Download PDF

Info

Publication number
CA2738025A1
CA2738025A1 CA2738025A CA2738025A CA2738025A1 CA 2738025 A1 CA2738025 A1 CA 2738025A1 CA 2738025 A CA2738025 A CA 2738025A CA 2738025 A CA2738025 A CA 2738025A CA 2738025 A1 CA2738025 A1 CA 2738025A1
Authority
CA
Canada
Prior art keywords
methyl
fluoro
alkyl
compound according
mmol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2738025A
Other languages
French (fr)
Inventor
Vladimir Ivanov
Bertil Samuelsson
Per-Ola Johansson
Pia Kahnberg
Horst Wahling
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Medivir AB
Original Assignee
Medivir AB
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 Medivir AB filed Critical Medivir AB
Publication of CA2738025A1 publication Critical patent/CA2738025A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

Compounds of the formula II, wherein R3 is C1-C3 alkyl or C3-C6 cycloalkyl, either of which is optionally substituted with one or two methyl and/or a fluoro, trifluoromethyl or methoxy, when R3 is C3-C6 cycloalkyl it may alternatively be gem substituted with fluoro; R4 is methyl or fluoro; m is 0, 1 or 2; E is a bond, or thiazolyl, optionally substituted with methyl or fluoro; A, is CH or N, A2 is CR6R7 or NR6, provided at least one of A1 and A2 comprises N; n is 0 or 1 such that the ring containing A1 and A2 is a saturated, nitrogen-containing ring of 5 or 6 ring atoms; R6 is H, C1-C4 alkyl, C1-C4 haloalkyl, C1-C3 alkyl-O-C1-C3 alkyl, or when A2 is C, R6 can also be C1-C4 alkoxy or F; R7 is H, C1-C4 alkyl or F; or a pharmaceutically acceptable salt, N-oxide or hydrate thereof, have utility in the treatment of disorders characterized by inappropriate expression or activation of cathepsin K, such as osteoporosis, osteoarthritis, rheumatoid arthritis or bone metastases.

Description

Protease Inhibitors Field of the invention This invention relates to inhibitors of cysteine proteases, especially those of the papain superfamily. The invention provides novel compounds useful in the prophylaxis or treatment of disorders stemming from misbalance of physiological proteases such as cathepsin K.
Description of the related art The papain superfamily of cysteine proteases is widely distributed in diverse species including mammals, invertebrates, protozoa, plants and bacteria. A number of mammalian cathepsin enzymes, including cathepsins B, F, H, K, L, 0 and S, have been ascribed to this superfamily, and inappropriate regulation of their activity has been implicated in a number of metabolic disorders including arthritis, muscular dystrophy, inflammation, glomerulonephritis and tumour invasion. Pathogenic cathepsin like enzymes include the bacterial gingipains, the malarial falcipains I, II, III et seq and cysteine proteases from Pneumocystis carinii, Trypanosoma cruzei and brucei, Crithidia fusiculata, Schistosoma spp.

The inappropriate regulation of cathepsin K has been implicated in a number of disorders including osteoporosis, gingival diseases such as gingivitis and periodontitis, Paget's disease, hypercalcaemia of malignancy and metabolic bone disease. In view of its elevated levels in chondroclasts of osteoarthritic synovium, cathepsin K is implicated in diseases characterised by excessive cartilage or matrix degradation, such as osteoarthritis and rheumatoid arthritis.

It is likely that treatment of bone and cartilage disorders such as osteoarthritis and osteoporosis will require life-long administration of a cathepsin K inhibitor, often to a patient population within or nearing the geriatric phase. This places unusually high requirements on the ease of administration of drugs intended for such disorders. For example attempts are underway to stretch the dosage regimes of the current osteoporosis drugs of the bisphosphonate class to weekly or longer administration regimes to aid compliance. However, even with improved dosing, other side effects of the bisphosphonates remain.
Bisphosphonates block bone turnover rather than attenuate it as a cathepsin K
inhibitor does.
For healthy bone it is important to maintain the remodelling process which bisphosphonates block completely. In addition, bis-phosphonates have a very long half-life in bone so if effects such as osteonecrosis of the jaw manifest themselves, it is impossible to remove the bisphosphonate from the bone. In contrast, cathepsin K inhibitors typically have a fast onset and off rate mode of action, which means that if a problem was to be identified, dosing could be halted and there would be no build up of the inhibitor in the bone matrix.
There is thus a desire for alternative osteoporosis and osteoarthritis drugs with superior pharmacokinetic and/or pharmacodynamic properties.

International patent application no W02008/007107 discloses compounds of the formula Ra Rb Rc 0 N
Rd H
O
where Rd is a substituted monocyclic ring, Rc is branched alkyl or cycloalkyl and Ra and Rb are a variety of groups including H, methyl, ethyl, ether, thioether, amine, sulphonate etc. The only compounds which are prepared have H or methoxy at this position.

There remains a need in the art for potent inhibitors of cathepsin K. Of particular benefit are inhibitors of cathepsin K which show selectivity for cathepsin K over other cathepsins (e.g.
selectivity over cathepsin S and/or cathepsin L). Potent inhibitors of cathepsin K which demonstrate properties such as high permeability and/or advantageous metabolic profiles may be expected to be of great value in a clinical setting. Cathepsin K related indications such as osteoporosis or arthritis presuppose protracted periods of administration and therefore it is desirable that the compounds have minimal toxicity or genotoxicity.

Brief description of the invention According to the present invention, there is provided a compound of formula II:
N
H
O R3 = O
A2'[ 1 H H

Al-E O O II
(R4) m wherein R3 is C1-C3 alkyl or C3-C6 cycloalkyl, either of which is optionally substituted with one or two methyl and/or a fluoro, trifluoromethyl or methoxy, when R3 is C3-C6 cycloalkyl it may alternatively be gem subsituted with fluoro;
R4 is methyl or fluoro; m is 0, 1 or 2;
E is a bond, or thiazolyl, optionally substituted with methyl or fluoro;
A, is CH or N, A2 is CR6R' or NR6, provided at least one of A, and A2 comprises N;
R6 is H, C1-C4 alkyl, C1-C4 haloalkyl, C1-C3 alkyl-O-C,-C3 alkyl, or when A2 is C, R6 can also be C1-C4 alkoxy or F;
R7 is H, C1-C4 alkyl or F
or a pharmaceutically acceptable salt, N-oxide or hydrate thereof.

It will be appreciated that the compounds of the invention can exist as hydrates, such as those of the partial formulae:

H H
O O
bN. N

and the invention extends to all such alternative forms.

Suitably R3 is C1-C3 alkyl or C3-C6 cycloalkyl, either of which is optionally substituted with one or two methyl and/or a fluoro, trifluoromethyl or methoxy.

Representative values for cycloalkyl for R3 include cyclopropyl, cyclobutyl and especially cyclopentyl or cyclohexyl, any of which being substitued with fluoro or gem fluoro. Gem-fluoro at the 2 position of a cyclopropyl, the 3 position of cyclobutyl or cyclopentyl or the 4 position of cyclopropyl is often convenient. Gem-fluoro at the 4 position of cyclohexyl is also often convenient.

In one embodiment of the invention R3 represents the side chain of leucine. In a second embodiment of the invention R3 represents the side chain of isoleucine. In a third embodiment of the invention R3 represents the side chain of cyclohexylglycine. In a fourth embodiment of the invention R3 represents the side chain of cyclopentylglycine. In a fifth embodiment of the invention, R3 represents the side chain of of O-methylthreonine. In a fifth embodiment of the invention R3 represents the side chain of 4-fluoroleucine. In a sixth embodiment of the invention R3 represents the side chain of 3-methoxyvaline.

Currently preferred values of R3 include those embodied by the partial structures:
and especially In one embodiment of the invention m represents 2. Of particular interest are compounds wherein m represents 1. Still further embodiments of the invention have m as 0, especially when the adjacent thiazolyl is substituted with Me or preferably F.

R4 suitably represents methyl or fluoro, especially fluoro. If m is 2, it is currently preferred that each R4 is the same.

When m represents 1, R4 is suitably positioned as shown by the partial structure:
O
N
H

E is conveniently a bond, that is the unsaturated nitrogen containing ring bearing Al and A2 is bonded directly to the para position of the phenyl ring. However, it is currently preferred that E
is thiazolyl, which is optionally substituted with methyl or more preferably fluoro. The preferred orientation of the thiazolyl ring is:

A2'[ J1 Al N
I
S
R4) M

where R5 is H, methyl or fluoro.

The ring containing A, and A2 is a saturated, nitrogen-containing ring of 5 or 6 ring atoms.
Thus n is 0 or 1 and suitably n is 1. Representative rings thus include pyrrolidin-l-yl, pyrrolidin-3-yl, piperazin-l-yl, piperidin-4-yl and piperidine-l-yl. The ring is conveniently substituted, for example with alkyl or haloalkyl, typically methyl or propyl or trifluromethyl.
Alternatively the ring is substituted with an ether such as methoxymethyl- or methoxyethyl-.

When A2 is carbon, the ring can alternatively be substitued with alkoxy such as methoxy, or fluoro, especially gem-fluoro.

A favoured embodiment of the invention has the formula Ila:

N

H
O R3 O J~f N `-N N
H O H O
R6-NN/ I (R4)m II a S
5 wherein R3 is branched C2-C6 alkyl or C3-C6 cycloalkyl, either of which is substituted with halo or trifluoromethyl;
R4 is methyl or fluoro; m is 0 or 1 or 2;
R5 is H, methyl or fluoro;
R6 is C1-C4 alkyl;
or a pharmaceutically acceptable salt, N-oxide or hydrate thereof (collectively referred to herein as compounds of the invention).

R5 is preferably fluoro, especially when m is 0. The remaining preferments are as defined above in relation to Formula II. References to formula II below are understood to include the corresponding embodiments of formula Ila.

Representative embodiments of formula II include:
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-carbonyl)-3-methyl-butyl]-4-[2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-yl)-1 -cyclohexyl-2-oxo-ethyl]-4-[2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-yl)-1 -cycl open tyl-2-oxo-eth yl]-4-[2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-carbonyl)-2-methyl-butyl]-4-[2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-carbonyl)-3-methyl-butyl]-4-[5-methyl-2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-yl)-1 -cyclohexyl-2-oxo-ethyl]-4-[5-methyl-2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-yl)-1 -cyclohexyl-2-oxo-ethyl]-4-[5-methyl-2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-carbonyl)-2-methyl-butyl]-4-5-methyl-2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-carbonyl)-3-methyl-butyl]-4-[5-fluoro-2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-yl)-1 -cyclohexyl-2-oxo-ethyl]-4-[5-fluoro-2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-yl)-1 -cycl open tyl-2-oxo-eth yl]-4-[5-fluoro-2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-carbonyl)-2-methyl-butyl]-4-[5-fluoro-2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-carbonyl)-3-methyl-butyl]-3-fluoro-4-[2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-yl)-1 -cyclohexyl-2-oxo-ethyl]-3-fluoro-4-[2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-yl)-1 -cycl open tyl-2-oxo-eth yl]-3-fluoro-4-[2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide;
N-[1 -(6-nitrile-3-oxo-hexahydro-furo[3,2-b]pyrrol-4-carbonyl)-2-methyl-butyl]-3-fluoro-4-[2-(4-methyl-piperazin-1 -yl)-thiazol-4-yl]-benzamide and pharmaceutically acceptable salts, N-oxides and hydrates thereof.

The C,-Cn alkyl definition of R6 or R7 is is meant to include both branched and unbranched alkyl moieties containing between one and n carbon atoms in total. Examples of such R6 groups or R7 are methyl, ethyl, propyl (n-propyl and isopropyl), butyl (n-butyl, isobutyl, tert-butyl and sec-butyl). One R6 group of particular interest is methyl. A second R6 group of particular interest is propyl (especially n-propyl). As R3 is optionally substituted with one or two methyl groups, this moiety may also define a branched alkyl chain of up to 5 C atoms.

In some embodiments of the invention A, is N.

Additional aspects of the invention include a pharmaceutical composition comprising a compound as defined above and a pharmaceutically acceptable carrier or diluent therefor.
A further aspect of the invention is the use of a compound as defined above in the manufacture of a medicament for the treatment of disorders mediated by cathepsin K, such as:
osteoporosis, gingival diseases (such as gingivitis and periodontitis), Paget's disease, hypercalcaemia of malignancy, metabolic bone disease, diseases characterised by excessive cartilage or matrix degradation (such as osteoarthritis and rheumatoid arthritis), bone cancers including neoplasia, pain (especially chronic pain).

Additionally provided is a method for the treatment or prevention of a disorder mediated by cathepsin K comprising the administration of a safe and effective amount of a compound of the invention for the purpose of treating or preventing said disorder which is mediated by cathepsin K.

Also provided is a compound of the invention for the treatment or prevention of a disorder mediated by cathepsin K.

Further, there is provided as an aspect of the invention novel intermediates (as described herein) which may be of use in the preparation of the compounds of the invention.

In particular there is provided a compound of the formula:
N
H O

OMe N 'H OMe H

or an N-protected derivative thereof (e.g. Boc, CBz, or Fmoc-protected). Also provided by the invention is the corresponding 1,3-dioxolane protected analogue and N-protected derivatives thereof (e.g. Boc- CBz, or Fmoc protected). A further novel intermediate of the invention is has the formula:

O
Oz: H
OMe N OMe H

or the corresponding 1,3-dioxolane protected analogue, in each case wherein the N function is optionally protected with a conventional protecting group such as Boc, CBz or Fmoc.
The compounds of the invention can form salts which form an additional aspect of the invention. Appropriate pharmaceutically acceptable salts of the compounds of Formula II
include salts of organic acids, especially carboxylic acids, including but not limited to acetate, trifluoroacetate, lactate, gluconate, citrate, tartrate, maleate, malate, pantothenate, isethionate, adipate, alginate, aspartate, benzoate, butyrate, digluconate, cyclopentanate, glucoheptanate, glycerophosphate, oxalate, heptanoate, hexanoate, fumarate, nicotinate, palmoate, pectinate, 3-phenylpropionate, picrate, pivalate, proprionate, tartrate, lactobionate, pivolate, camphorate, undecanoate and succinate, organic sulphonic acids such as methanesulphonate, ethanesulphonate, 2-hydroxyethane sulphonate, camphorsulphonate, 2-napthalenesulphonate, benzenesulphonate, p-chlorobenzenesuIphonate and p-toluenesulphonate; and inorganic acids such as hydrochloride, hydrobromide, hydroiodide, sulphate, bisulphate, hemisulphate, thiocyanate, persulphate, phosphoric and sulphonic acids.
The compounds of the invention may in some cases be isolated as the hydrate.
Hydrates are typically prepared by recrystallisation from an aqueous/organic solvent mixture using organic solvents such as dioxin, tetrahydrofuran or methanol. Hydrates can also be generated in situ by administration of the corresponding ketone to a patient.

The N-oxides of compounds of the invention can be prepared by methods known to those of ordinary skill in the art. For example, N-oxides can be prepared by treating an unoxidized form of the compound of the invention with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, meta-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 C. Alternatively, the N-oxides of the compounds of the invention can be prepared from the N-oxide of an appropriate starting material.

Examples of N-oxides of the invention include those with the partial structures:

0 ~N~S R3 \N (/ I R3 Compounds of the invention in unoxidized form can be prepared from N-oxides of the corresponding compounds of the invention by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus bichloride, tribromide, or the like) in an suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 80 C.

It should be noted that the radical positions on any molecular moiety used in the definitions may be anywhere on such moiety as long as it is chemically stable.

Radicals used in the definitions of the variables include all possible isomers unless otherwise indicated. For instance butyl includes t-butyl, i-butyl, n-butyl etc.

When any variable occurs more than one time in any constituent, each definition is independent.

Unless otherwise mentioned or indicated, the chemical designation of a compound encompasses the mixture of all possible stereochemically isomeric forms, which said compound may possess. Said mixture may contain all diastereomers and/or enantiomers of the basic molecular structure of said compound. All stereochemically isomeric forms of the compounds of the present invention both in pure form or mixed with each other are intended to be embraced within the scope of the present invention.

Pure stereoisomeric forms of the compounds and intermediates as mentioned herein are defined as isomers substantially free of other enantiomeric or diastereomeric forms of the same basic molecular structure of said compounds or intermediates. In particular, the term "stereoisomerically pure" concerns compounds or intermediates having a stereoisomeric excess of at least 80% (i.e. minimum 90% of one isomer and maximum 10% of the other possible isomers) up to a stereoisomeric excess of 100% (i.e. 100% of one isomer and none of the other), more in particular, compounds or intermediates having a stereoisomeric excess of 90% up to 100%, even more in particular having a stereoisomeric excess of 94%
up to 100%
and most in particular having a stereoisomeric excess of 97% up to 100%. The terms "enantiomerically pure" and "diastereomerically pure" should be understood in a similar way, but then having regard to the enantiomeric excess, and the diastereomeric excess, respectively, of the mixture in question.

Compounds of the invention can be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomer. While resolution of enantiomers can be carried out using covalent diasteromeric derivatives of compounds of Formula II, dissociable complexes are preferred (e.g., crystalline;
diastereoisomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities. The diastereomers can be separated by chromatography, for example HPLC or, preferably, by separation/resolution techniques based upon differences in solubility.
The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization. A more detailed description of the 5 techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981).

The compounds of formula II or any subgroup of formula II as defined herein include radioisotopes or radiomarked compounds, wherein one or more of the atoms is replaced by an 10 isotope of that atom, i.e. an atom having the same atomic number as, but an atomic mass different from, the one(s) typically found in nature. Examples of isotopes that may be incorporated into the compounds of formula I or any subgroup of formula I, include but are not limited to isotopes of hydrogen, such as 2H and 3H (also denoted D for deuterium and T for tritium respectively), carbon, such as 110 13C and 14C, nitrogen, such as 13N
and 15N, oxygen, such as 150, 170 and 180, phosphorus, such as 31P and 32P, sulphur, such as 355, fluorine, such as 18F, chlorine, such as 36CI, bromine such as 75Br, 76Br, 77Br and 82Br, and iodine, such as 1231, 1241, 1251 and 1311 The choice of isotope included in an isotope-labelled compound will depend on the specific application of that compound. For example, for drug or substrate tissue distribution assays, compounds wherein a radioactive isotope such as 3H or 14C is incorporated will generally be most useful. For radio-imaging applications, for example positron emission tomography (PET) a positron emitting isotope such as 110 18F 13N or 150 will be useful. The incorporation of a heavier isotope, such as deuterium, i.e. 2H, may provide greater metabolic stability to a compound of formula I or any subgroup of formula 1, which may result in, for example, an increased in vivo half life of the compound or reduced dosage requirements.

For synthetic convenience it will generally be preferred that the compounds of formula 11 are in the natural isotopic state.

Isotopically labelled compounds of formula I or any subgroup of formula 11 can be prepared by processes analogous to those described in the Schemes and/or Examples herein below by using the appropriate isotopically labelled reagent or starting material instead of the corresponding non-isotopically labelled reagent or starting material, or by conventional techniques known to those skilled in the art.

It will be appreciated that the invention extends to prodrugs, solvates, complexes and other forms releasing a compound of the invention in vivo.
While it is possible for the active agent to be administered alone, it is preferable to present it as part of a pharmaceutical formulation. Such a formulation will comprise the above defined active agent together with one or more acceptable carriers/excipients and optionally other therapeutic ingredients. The carrier(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient.

The formulations include those suitable for rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration, but preferably the formulation is an orally administered formulation.
The formulations may conveniently be presented in unit dosage form, e.g.
tablets and sustained release capsules, and may be prepared by any methods well known in the art of pharmacy.

Such methods include the step of bringing into association the above defined active agent with the carrier. In general, the formulations are prepared by uniformly and intimately bringing into association the active agent with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product. The invention extends to methods for preparing a pharmaceutical composition comprising bringing a compound of Formula II or its pharmaceutically acceptable salt in conjunction or association with a pharmaceutically acceptable carrier or vehicle. If the manufacture of pharmaceutical formulations involves intimate mixing of pharmaceutical excipients and the active ingredient in salt form, then it is often preferred to use excipients which are non-basic in nature, i.e. either acidic or neutral.
Formulations for oral administration in the present invention may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active agent; as a powder or granules; as a solution or a suspension of the active agent in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water in oil liquid emulsion and as a bolus etc.

With regard to compositions for oral administration (e.g. tablets and capsules), the term suitable carrier includes vehicles such as common excipients e.g. binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, polyvinylpyrrolidone (Povidone), methylcellulose, ethylcellulose, sodium carboxymethylcellulose, hydroxypropyl-methylcellulose, sucrose and starch; fillers and carriers, for example corn starch, gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid; and lubricants such as magnesium stearate, sodium stearate and other metallic stearates, glycerol stearate stearic acid, silicone fluid, talc waxes, oils and colloidal silica.
Flavouring agents such as peppermint, oil of wintergreen, cherry flavouring or the like can also be used. It may be desirable to add a colouring agent to make the dosage form readily identifiable. Tablets may also be coated by methods well known in the art.

A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active agent in a free flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent. Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may be optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active agent.

Other formulations suitable for oral administration include lozenges comprising the active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active agent in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active agent in a suitable liquid carrier.

The appropriate dosage for the compounds or formulations of the invention will depend upon the indication and the patient and is readily determined by conventional animal trials. Dosages providing intracellular (for inhibition of physiological proteases of the papain superamily) concentrations of the order 0.01-100 uM, more preferably 0.01-10 uM, such as 0.1-25 uM are typically desirable and achievable.

Compounds of the invention are prepared by a variety of solution and solid phase chemistries.
The compounds are typically prepared as building blocks reflecting the P1, P2 and P3 moieties of the end product inhibitor. Without in any way wishing to be bound by theory, or the ascription of tentative binding modes for specific variables, the notional concepts P1, P2 and P3 as used herein are provided for convenience only and have substantially their conventional Schlecter & Berger meanings and denote those portions of the inhibitor believed to fill the S1, S2, and S3 subsites respectively of the enzyme, where S1 is adjacent the cleavage site and S3 remote from the cleavage site. Compounds defined by Formula II are intended to be within the scope of the invention, regardless of binding mode.

Broadly speaking the P1 building block will have the formula:
PGN or PG'N
O \ Rb 0,Rc wherein R1 and R2 are as defined above, the two Rb groups define a ketal, such as the bis methyl ketal or together define a cyclic ketal such as 1,3-dioxolane;
and Rc is an hydroxy protecting group. Less commonly Rc is H or represents the keto function of the end-product inhibitor in cases where the P1 building block as the ketone is elongated with P2 and P3.

W005/066180 describes the preparation of intermediates towards the above P1 building blocks, including:

HO 0 .110 BnO 0 N
0-1( N //OH
(D~0~O 6 0,O 58 The first stage in the synthesis of compounds of the invention is typically the preparation in solution of a functionalized P1 building block. Scheme 1 illustrates a route to a convenient 6-aldehyde intermediate.
O H O O H O HO H

iii, iv CC/ Me Me N
H OH ) H Me bocH Me O O O 0 1c 1a 1b H O H O H O
V, vi vii viii Me Me Me N H Me N H Me N Me boc boc b H
oc 1d le if i) Dess-Martin Periodinane, DCM; ii) Trimethylorthoformate, pTs, MeOH; iii) Pd(OH)2, H2, MeOH; iv) Boc20, 10 % Na2C03, v) Dess-Martin Periodinane, DCM; vi) 1) CH3PPh3Br, KOtBu, THF;
vii) 1) 9-BBN-H, THF, 2) NaBO31 H2O, THF; viii) Dess-Martin Periodinane, DCM.
Scheme 1 The starting bicyclic alcohol (1 a) can be prepared as described in WO05/066180. Oxidation of the hydroxy function for example with Dess-Martin periodinane followed by transformation of the afforded keto function into a dimethyl ketal effected by treatment with trimethyl orthoformate in the presence of an acid like p-toluenesulphonic acid provides the ketal (1 b).
Removal of the Cbz and benzyl protecting groups effected for instance by hydrogenolysis using a catalyst like Pd(OH)2 or the like, followed by boc protection of the afforded free amine provides the alcohol (1 c). Oxidation of the afforded free alcohol using for instance Dess-Martin periodinane in a solvent like dichloromethane followed by a Wittig reaction using methyl triphenylphosphinium bromide in the presence of KOt.Bu or the like provides the olefin (1 d).
Hydroxylation of the double bond effected for example by treatment with 9-BBN-H, provides the primary alcohol (1e) which subsequently can be oxidized to the corresponding aldehyde (1f) using any suitable oxidation method such as treatment with Dess-Martin periodinane or the like.

Scheme 2 illustrates a typical procedure for a 6-nitrile P1 building block commencing from the 6-aldehyde intermediate of Scheme 1.

H H 0 HO, N H H O
O

N H i\ N iH 0\ ii N H 00 if 1g 1h is NH2OH.HCl, NaOAc ii: Tf20, Et3N

Conversion of the aldehyde if (Scheme 2) to the corresponding desired nitrile 1 h (Scheme 2) proceeds via dehydration of oxime 1 g (Scheme 2). Hence the aldehyde in an ethanol/water mixture can be treated with NH2OH and sodium acetate, for example overnight at room 5 temperature. TLC can be used to monitor that the starting material had been consumed.
Conventional work-up provides the crude oxime 1g (E and Z isomers) used without further purification. The crude oxime 1g may be taken up in dichloromethane and triethylamine added at -78 C. Trifluoromethanesulfonic anhydride in an organic solvent such as dichloromethane can be added, typically while allowing the reaction to warm up to room temperature. Once the 10 reaction appears to be complete, extraction and chromatography of the residue, for example on silica gel affords a typical nitrile building block 1 h, generally in good yield. Typically only one isomer of the nitrile 1 h, eg that with C-6S stereochemistry is isolated as the reaction usually proceeds without loss of stereochemical integrity. Numerous examples in the literature show this conversion from an aldehyde to the corresponding nitrile to proceed with a similar retention 15 of stereochemistry eg. Hutt et al, Journal of Organic Chemistry, 72(26), 10130, 2007.

Typically to get to the final compound, the P1 building block such as 1h above is N-deprotected in a conventional fashion, such as treatment with acetyl chloride in methanol to remove an N-Boc protecting group. With the subsequent free amine, the P2 residue is introduced, eg via BocP2-OH using standard coupling conditions such as HATU, DIPEA in DMF. The terminal Boc protection is again removed with acetyl chloride in methanol and the P3 residue introduced via P3-OH using standard coupling conditions such as HATU, DIPEA in DMF. Finally the dimethylketal protection is removed with TFA to afford the required final compound.

Elongation is typically carried out in the presence of a suitable coupling agent e.g., benzotriazole-1-yloxytrispyrrolidinophosphonium hexafluorophosphate (PyBOP), 0-benzotriazol-I-yl-N,N,N',N'-tetramethyl-uronium hexafluorophosphate (HBTU), O-(7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyl-uronium hexafluorophosphate (HATU), 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), or 1,3-dicyclohexyl carbodiimide (DCC), optionally in the presence of I-hydroxybenzotriazole (HOBT), and a base such as N,N-diisopropylethylamine, triethylamine, N-methylmorpholine, and the like. The reaction is typically carried out at 20 to 30 C, preferably at about 25 C, and requires 2 to 24 h to complete. Suitable reaction solvents are inert organic solvents such as halogenated organic solvents (e.g., methylene chloride, chloroform, and the like), acetonitrile, N,N-dimethylformamide, ethereal solvents such as tetrahydrofuran, dioxane, and the like.
Alternatively, the above elongation coupling step can be carried out by first converting the P3/P2 building block into an active acid derivative such as succinimide ester and then reacting it with the P1 amine. The reaction typically requires 2 to 3 h to complete.
The conditions utilized in this reaction depend on the nature of the active acid derivative.
For example, if it is an acid chloride derivative, the reaction is carried out in the presence of a suitable base (e.g.
triethylamine, diisopropylethylamine, pyridine, and the like). Suitable reaction solvents are polar organic solvents such as acetonitrile, N,N-dimethylformamide, dichloromethane, or any suitable mixtures thereof.

The P2 building block is typically an N-protected amino acid such as L-leucine, L-isoleucine, O-methyl-L-threonine, L-3-hydroxyvaline, 4-fluoroleucine, L-cyclopentylglycine or L-cyclohexylglycine, and P3 typically comprises a capping group such as a benzoic acid derivative with, eg, the N-alkyl-piperazinyl-E moiety already introduced or provided with a synthon therefor in the para position.

The suitably protected individual building blocks can first be prepared and subsequently coupled together, preferably in the sequence P2+P1- P2-P1 followed by N-alkylpiperazinyl-E-benzoic acid*+P2-P1_ N-alkylpiperazinyl-E-benzoate-P2-P1, where * denotes an activated form, in order to minimise racemisation at P2.

Coupling between two amino acids, an amino acid and a peptide, or two peptide fragments can be carried out using standard coupling procedures such as the azide method, mixed carbonic-carboxylic acid anhydride (isobutyl chloroformate) method, carbodiimide (dicyclohexylcarbodiimide, diisopropylcarbodiimide, or water-soluble carbodiimide) method, active ester (p-nitrophenyl ester, N-hydroxysuccinic imido ester) method, Woodward reagent K-method, carbonyldiimidazole method, phosphorus reagents or oxidation-reduction methods.
Some of these methods (especially the carbodiimide method) can be enhanced by adding 1-hydroxybenzotriazole or 4-DMAP. These coupling reactions can be performed in either solution (liquid phase) or solid phase.
More explicitly, the coupling step involves the dehydrative coupling of a free carboxyl of one reactant with the free amino group of the other reactant in the present of a coupling agent to form a linking amide bond. Descriptions of such coupling agents are found in general textbooks on peptide chemistry, for example, M. Bodanszky, "Peptide Chemistry", 2nd rev ed., Springer-Verlag, Berlin, Germany, (1993) hereafter simply referred to as Bodanszky, the contents of which are hereby incorporated by reference. Examples of suitable coupling agents are N,N'-dicyclohexylcarbodiimide, 1-hydroxybenzotriazole in the presence of N,N'-dicyclohexylcarbodiimide or N-ethyl-N'-[ (3-dimethylamino) propyl]
carbodiimide. A practical and useful coupling agent is the commercially available (benzotriazol-1-yloxy)tris-(dimethylamino) phosphonium hexafluorophosphate, either by itself or in the present of 1-hydroxybenzotriazole or 4-DMAP. Another practical and useful coupling agent is commercially available 2-(IH-benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate. Still another practical and useful coupling agent is commercially available O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate.

The coupling reaction is conducted in an inert solvent, e. g. dichloromethane, acetonitrile or dimethylformamide. An excess of a tertiary amine, e. g. diisopropylethylamine, N-methylmorpholine, N-methylpyrrolidine or 4-DMAP is added to maintain the reaction mixture at a pH of about 8. The reaction temperature usually ranges between 0 C and 50 C and the reaction time usually ranges between 15 min and 24 h.

The functional groups of the constituent non-natural amino acids generally must be protected during the coupling reactions to avoid formation of undesired bonds. The protecting groups that can be used are listed in Greene, "Protective Groups in Organic Chemistry", John Wiley &
Sons, New York (1981) and "The Peptides: Analysis, Synthesis, Biology", Vol.
3, Academic Press, New York (1981), hereafter referred to simply as Greene, the disclosures of which are hereby incorporated by reference.

The alpha-carboxyl group of the C-terminal residue is usually protected as an ester that can be cleaved to give the carboxylic acid. Protecting groups that can be used include 1) alkyl esters such as methyl, trimethylsilyl and t-butyl, 2) aralkyl esters such as benzyl and substituted benzyl, or 3) esters that can be cleaved by mild base or mild reductive means such as trichloroethyl and phenacyl esters.

The alpha-amino group of each amino acid to be coupled is typically N-protected. Any protecting group known in the art can be used. Examples of such groups include: 1) acyl groups such as formyl, trifluoroacetyl, phthalyl, and p-toluenesulfonyl; 2) aromatic carbamate groups such as benzyloxycarbonyl (Cbz or Z) and substituted benzyloxycarbonyls, and 9-fluorenylmethyloxycarbonyl (Fmoc); 3) aliphatic carbamate groups such as tertbutyloxycarbonyl (Boc), ethoxycarbonyl, diisopropylmethoxy-carbonyl, and allyloxycarbonyl;
4) cyclic alkyl carbamate groups such as cyclopentyloxycarbonyl and adamantyloxycarbonyl;
5) alkyl groups such as triphenylmethyl and benzyl; 6) trialkylsilyl such as trimethylsilyl; and 7) thiol containing groups such as phenylthiocarbonyl and dithiasuccinoyl. The preferred alpha-amino protecting group is either Boc or Fmoc. Many amino acid derivatives suitably protected for peptide synthesis are commercially available.

The alpha-amino protecting group is typically cleaved prior to the next coupling step. When the Boc group is used, the methods of choice are trifluoroacetic acid, neat or in dichloromethane, or HCI in dioxane or in ethyl acetate. The resulting ammonium salt is then neutralized either prior to the coupling or in situ with basic solutions such as aqueous buffers, or tertiary amines in dichloromethane or acetonitrile or dimethylformamide. When the Fmoc group is used, the reagents of choice are piperidine or substituted piperidine in dimethylformamide, but any secondary amine can be used. The deprotection is carried out at a temperature between 0 C
and room temperature usually 20-22 C.

Once the inhibitor sequence is completed any remaining protecting groups are removed in whatever manner is dictated by the choice of protecting groups. These procedures are well known to those skilled in the art.

P2 building blocks in the form of N-protected L-amino acids are readily available commercially, for example L-leucine, L-isoleucine, L-cyclohexylglycine, O-methyl-L threonine and others are available commercially with a number of protecting group variants such as CBz, Boc or Fmoc.
Other variants of R3 are easily prepared from commercially available starting materials. For example compounds wherein R3 is -C(CH3)20CH3 can be prepared by reacting CBz protected (S)-(+)-2-amino-3-hydroxy-3-methylbutanoic acid with 3,3-dimethoxy-hexahydro-furo(3,2b)pyrrole to form the desired P2-P1 unit. The P2 side chain alcohol can now be methylated using methyliodide under conventional sodium hydride, imidazole, THE conditions to obtain the desired P2 without substantial racemisation of the alpha centre.
This P2-P1 moiety can now be carried through the synthesis as described herein, namely CBz removal and coupling.

W005/565299 describes the preparation of a gamma-fluoroleucine P2 building block. An alternative synthsis of Fmoc and N-Boc-gammafluoroleucine building blocks is shown in Truong et al Syn. Lett. 2005 no 8 1278-1280.
The preparation of P3 building blocks are described in W005/066180, W008/007114 or readily prepared by analogous methods. For example, Scheme E below shows the preparation of a P3 building block wherein E is a fluoro-substituted thiazolyl:

Br F
~

F F
O s s F I N ~ N \ - - / N - N / > - - N V _ / N -I V HO I /
H 3 C 0 Br IV H3CO
Y

i. HOAc, Br2, RT, 2h, 55% yield; ii. KF, 18-crown-6, CH3CN, 90 C, 16 h, 31%
yield; iii. HOAc, Br2, 45 C, 4 h, 100% yield ; iv. 4-methylpiperazine-1-carbothioamide, ethanol, 70 C, 2 h, 74%
yield, v. LiOH, THF, H2O, RT, 16 h, 79 % yield.

Scheme E
Synthesis of 4-[5-fluoro-2-(4-methyl-piperazin-1-yl)-thiazol-4-yl]-benzoic acid The starting material, methyl 4-acetylbenzoate, is commercially available.
Bromination at the a-position to the ketone is achieved with bromine in acetic acid to provide the desired 4-(2-bromo-acetyl)-benzoic acid methyl ester. Subsequent treatment of 4-(2-bromo-acetyl)-benzoic acid methyl ester with potassium fluoride in the presence of 18-crown-6 at 90 C, provides 4-(2-fluoro-acetyl)-benzoic acid methyl ester after column chromatography.
Repeated bromination at the a-position to the ketone is achieved with bromine in acetic acid to provide the desired 4-(2-bromo-2-fluoro-acetyl)-benzoic acid methyl ester. Formation of the thiazole is typically carried out by heating 4-(2-bromo-2-fluoro-acetyl)-benzoic acid methyl ester with 4-methylpiperazine-1 -carbothioamide at 70 C for 2 hours. On cooling, the desired 4-[5-fluoro-2-(4-methyl-piperazin-1-yl)-thiazol-4-yl]-benzoic acid methyl ester precipitates out. Deprotection of the methyl ester is carried out using a lithium hydroxide solution and the desired acid, 4-[5-fluoro-2-(4-methyl-piperazin-1-yl)-thiazol-4-yl]-benzoic acid is generally obtained in good yield as the dihydrochloride salt on workup with hydrochloric acid.

The term "N-protecting group" or "N-protected" as used herein refers to those groups intended to protect the N-terminus of an amino acid or peptide or to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, "Protective Groups in Organic Synthesis" (John Wiley &
Sons, New York, 1981), which is hereby incorporated by reference. N-protecting groups include acyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoracetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and the like; sulfonyl groups such as benzenesulfonyl, p-toluenesulfonyl, and the like, carbamate forming groups such as 5 benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-n itrobenzyloxycarbonyl, 2-n itrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4-d imethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-d imethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, 1-(p-biphenylyl)-1-methylethoxycarbonyl, a,a-dimethyl-3,5-d imethoxybenzyloxycarbonyl, 10 benzhydryloxycarbonyl, t-butoxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2-trichloroethoxycarbonyl, phenoxycarbonyl, 4-n itrophenoxycarbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, phenylthiocarbonyl, and the like; alkyl groups such as benzyl, triphenylmethyl, benzyloxymethyl and the like; and 15 silyl groups such as trimethylsilyl and the like. Favoured N-protecting groups include formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, phenylsulfonyl, benzyl (bz), t-butoxycarbonyl (BOC) and benzyloxycarbonyl (Cbz).

Hydroxy and/or carboxy protecting groups are also extensively reviewed in Greene ibid and include ethers such as methyl, substituted methyl ethers such as methoxymethyl, 20 methylthiomethyl, benzyloxymethyl, t-butoxymethyl, 2-methoxyethoxymethyl and the like, silyl ethers such as trimethylsilyl (TMS), t-butyldimethylsilyl (TBDMS) tribenzylsilyl, triphenylsilyl, t-butyldiphenylsilyl triisopropyl silyl and the like, substituted ethyl ethers such as 1-ethoxymethyl, 1-methyl-1-methoxyethyl, t-butyl, allyl, benzyl, p-methoxybenzyl, dipehenylmethyl, triphenylmethyl and the like, aralkyl groups such as trityl, and pixyl (9-hydroxy-9-phenylxanthene derivatives, especially the chloride). Ester hydroxy protecting groups include esters such as formate, benzylformate, chloroacetate, methoxyacetate, phenoxyacetate, pivaloate, adamantoate, mesitoate, benzoate and the like. Carbonate hydroxy protecting groups include methyl vinyl, allyl, cinnamyl, benzyl and the like.

Detailed Description of the Embodiments Various embodiments of the invention will now be described by way of illustration only with reference to the following Examples.

Reference Example 1 A P3 building block Step a)4-Cyanopropiophenone QN
O

As described for the preparation of 4-cyanoacetophenone (Synth. Commun 1994, 887-890), a mixture of 4-bromopropiophenone (5.65 g, 26.4 mmol), Zn(CN)2 (1.80 g, 15.3 mmol), and Pd(PPh3)4 (2.95 g, 2.6 mmol) was refluxed at 80 C in deoxygenated DMF (35 mL, stored over 4 A molecular sieves, bubbled with Ar before use) for 18 h. The mixture was partitioned between toluene (100 mL) and 2N NH4OH (100 mL). The organic phase was extracted with 2N
NH4OH (100 mL), washed with saturated aqueous NaCl (2 x 100 mL), dried, and evaporated.
A 10 mmol scale reaction was done similarly and the crude products were combined. Flash chromatography (330 g silica, 6/1 petroleum ether - EtOAc) gave white solids (5.17 g, 89%).
1 H NMR (CDCI3) 6 ppm: 1.22 (t, 3H, J = 7.2 Hz), 3.00 (q, 2H, J = 7.3 Hz), 7.75 (d, 2H, J = 8.8 Hz), 8.03 (d, 2H, J = 8.4 Hz) 13C NMR (CDCI3) 6 ppm: 7.8, 32.1, 116.1, 117.9, 128.3, 132.4, 139.7, 199.2 step b) 4-Propionylbenzoic acid \ 15 4-Cyanopropiophenone (4.67 g, 29.3 mmol) was refluxed with 2N NaOH (90 mL, 180 mmol) and dioxane (90 mL) at 95 C overnight. The mixture was diluted with water (150 mL), washed with ether (75 mL), acidified to pH 2 with concentrated HCI, and extracted with ether (3 x 75 mL). The organic phase was washed with saturated aqueous NaCl (3 x 75 mL), dried, and evaporated to give yellow solids (5.12 g, 98%).

1H NMR (CDCI3+ CD3OD) 6 ppm: 1.18 (t, 3H, J = 7.2 Hz), 2.99, (q, 2H, J = 7.1 Hz), 7.95 (d, 2H, J = 8.4 Hz), 8.08 (d, 2H, J = 8.8 Hz) 13C NMR (CDCI3) 6 ppm: 7.9, 32.1, 127.7, 130.0, 134.0, 140.0, 168.0, 200.8 Step c)Methyl 4-propionylbenzoate )__(__C00Me The benzoic acid above (890 mg, 5 mmol), NaHCO3 (1.26 g, 15 mmol) and iodomethane (935 L, 15 mmol) in DMF (10 ml-) were stirred at RT overnight. The mixture was diluted with saturated aqueous NaCl (50 ml-) and extracted with ether (3 x 50 mL). The organic phase was washed with water (50 mL), dried, and evaporated. Flash chromatography (90 g silica, 2/1 petroleum ether - EtOAc) gave white solids (744 mg, 77%).

1 H NMR (CDCI3) 6 ppm: 1.24 (t, 3H, J = 7 Hz), 3.03 (q, 2H, J = 7 Hz), 3.95 (s, 3H), 8.0 and 8.12 (ABq, 4H) Step d) Methyl 4-(2-bromopropionyl)benzoate Br a COOMe Methyl 4-propionylbenzoate (744 mg, 3.87 mmol), pyrrolidone hydrotribromide (1.98 g), and 2-pyrrolidinone (380 mg, 4.5 mmol) in THE (38 ml-) were heated at 50 C under nitrogen for 3 h.
The mixture was cooled, filtered, concentrated, and then redissolved in ether (50 mL). The ether solution was washed successively with water (20 mL), saturated aqueous Na2S2O5 (20 mL), saturated aqueous NaCl (20 mL), and water (20mL), dried and evaporated to give a yellow oil (1.025 g) that was used directly in the Hantzsch coupling. This material contained 91 % of the desired bromoketone, 5% starting ketone, and 4% 4-bromo-1-butanol, as determined by 1 H NMR.

1 H NMR (CDCI3) 6 ppm: 1.92 (d, 3H, J = 7 Hz), 3.96 (s, 3H), 5.28 (q, 1 H, J =
7 Hz), 8.07 and 8.14 (ABq, 4H) Step e)4-[2-(4-tent-Butoxycarbonylpiperazin-1-yl)-5-methylthiazol-4-yllbenzoic acid methyl ester ~--\ s BocN N4 N
COOMe All of the a-bromoketone above and 4-thionocarbonylpiperazine-1-carboxylic acid tent-butyl ester (J. Med. Chem., 1998, 5037-5054, 917 mg, 3.73 mmol) were refluxed in 36 mL THE at 70 C for 2 h, under N2. The precipitate was filtered and the filtrate evaporated to give yellow solids. Flash column chromatography (silica, 5/1 petroleum ether - EtOAc) gave 624 mg of light yellow solids. Chromatography of the precicpitate (silica, 2/1 petroleum ether - EtOAc) gave 32 mg more of compound. Total yield is 44%.

1 H NMR (CDC13) 6 ppm: 1.46 (s, 9H), 2.43 (s, 3H), 3.42, (m, 4H), 3.54 (m, 4H), 3.90 (s, 3H), 7.68 and 8.04 (ABq, 4H).

Step f) 4-[2-(4-tent-Butoxycarbonylpiperazin-1-yl)-5-methylthiazol-4-yllbenzoic acid ~-- s BocN N-/,, N
COOH
The above methyl ester (564 mg, 1.35 mmol) was heated with 1.35 mL 2N NaOH, 5 mL THF, and 3.65 mL water at 60 C for 4 h. The reaction mixture was evaporated, poured into 20 mL
saturated aqueous NaCl and 20 mL CH2C12, and then acidified to pH 3 with 5%
citric acid, in an ice bath. The layers were separated and the organic phase was extracted further with 2 x 10 mL CH2C12. The organic phases were combined, washed with water (10 mL), dried, and evaporated to give light yellow solids (537 mg, 98%).

1 H NMR (CDC13) 6 ppm: 1.48 (s, 9H), 2.47 (s, 3H), 3.47 (m, 4H), 3.57 (m, 4H), 7.74 and 8.12 (ABq, 4H).

13C NMR (CDC13) 6 ppm: 12.6, 28.3, 42.8, 48.1, 80.3, 119.1, 127.8, 128.2, 130.1, 140.5, 145.6, 154.6, 167.2, 171.4.

LC MS: (M + H)+ 404, (M - H)- 402.

Step g)4-[5-methyl-2-(4-methyl-piperazin-1-yl)-thiazol-4-yllbenzoic acid /--\ s -N N -/,,N

COOH
4-[4-(4-Carboxy-phenyl)-5-methyl-thiazol-2-yl]-piperazine-1-carboxylic acid tert-butyl ester (0.421 mmol) was dissolved in 4M HCI in 1,4-dioxane, and stirred at room temperature for 1 h. The solvent was then removed under vacuum, and the residue 4-(5-methyl-2-piperazin-1-yl-thiazol-4-yl)-benzoic acid was suspended in methanol (10 ml) and treated with AcOH/AcONa buffer (pH -5.5, 5 ml), and formaldehyde (0.547 mmol).
The reaction mixture was stirred at room temperature for 1 h, then treated with NaCNBH3 (0.547 mmol) and stirred at room temperature overnight. The solvent was then removed under vacuum, and the residue was purified by column chromatography to afford the title compound (0.403 mmol, 95%).

MS(ES) m/z 318 (100%, [M+H]+).
Reference Example 2 An alternative P3 building block 3-Fluoro- 4-[2-(4-methylpiperazin-1-yl)-thiazol-4-yllbenzoic acid HCI salt TMSCHN2 Zn, Ac20, TFA
MeOH-PhMe 0 allyl CI, CoBr2 O
Br -W OH 0 deg to rt Br \ McCN 0 ~-WoMe OH 95% OMe 36% F F F

0 -N~\N~ S
CN HBr3 v-" NH2 0 0 0 EtOH, 70 C N~ OMe OMe N_N/ I , 2-pyrrolidinone Br 72% over 2 steps THF, 60-65 C F S F

quant NN
N--~/S _Ir F

Step a)Methyl 4-bromo-3-fluorobenzoate 4-Bromo-3-fluorobenzoic acid (2.46 g, 11.2 mmol) was dissolved in MeOH (9 mL) and toluene (4 mL) and cooled in an ice bath. (Trimethylsilyl)diazomethane (11 mL, 2.0 M
in hexanes, 22 mmol) was added dropwise until the yellow color persisted. The solution was stirred at room temperature for 40 mins and then concentrated in vacuo. A second batch of carboxylic acid (2.43 g) was treated similarly. The crude product from both batches were combined and subjected to flash chromatography (silica, 5/1 pentane - EtOAc) to give the methyl ester as white solids (4.92 g, 95% yield).

1H NMR (400 MHz, CDCI3) delta ppm 7.77 (m, 1 H), 7.71 (m, 1 H), 7.64 (m, 1 H), 3.93 (s, 3H).
Step b)Methyl 4-acetoxy-3-fluorobenzoate Allyl chloride (105 pL, 1.28 mmol) and TFA (20 pL, 0.26 mmol) were added to a suspension of zinc dust (480 mg, 7.34 mmol) and anhydrous cobalt(II) bromide (96.6 mg, 0.44 mmol) in MeCN (4 mL), under inert gas. After stirring at room temperature for 10 min, the aryl bromide (1.003 g, 4.30 mmol dissolved in 5 mL MeCN) from (a) was added, followed by acetic 5 anhydride (0.45 mL, 4.79 mmol) and more MeCN (1 mL). The mixture was stirred overnight, quenched with 1 M HCI (20 mL), and then extracted with EtOAc (3 x 20 mL). The organic phase was washed successively with saturated aqueous NaHCO3 (20 mL) and saturated NaCl (2 x 20 mL), dried (Na2SO4), and concentrated. Flash chromatography (silica, 6/1 to 4/1 petroleum ether - EtOAc gave recovered bromide (161.1 mg, 16%) and the desired ketone 10 (white solids, 305.5 mg, 36%).

NMR (CDC13) b ppm: 1H (400 MHz) 7.94-7.86 (m, 2H), 7.80 (dd, 1H, J = 11.2, 1.6 Hz), 3.95 (s, 3H), 2.67 (d, 3H, J = 4.4 Hz); 19F (376 MHz) -109.2 (m); 13C (100 MHz) 195.4 (d , J = 3.7 Hz), 165.1 (d, J = 2.2 Hz), 161.6 (d, J = 255 Hz), 135.8 (d, J = 8.1 Hz), 130.7 (d, J = 2.9 Hz), 129.0 d, J = 14 Hz), 125.2 (d, J = 3.6 Hz), 117.9 (d, J = 26 Hz), 52.7 (s), 31.4 (d, J = 7.3 Hz).

15 Step c)Methyl 4-(2-bromoacetoxy)-3-fluorobenzoate THE (10 mL) and 2-pyrrolidinone (91 pL, 1.20 mmol) were added to a mixture of the ketone from b) (198 mg, 1.01 mmol) and pyrrolidone hydrotribromide (532 mg, 1.07 mmol). After heating at 60-65 C for 2 h, the mixture was concentrated under vacuum and then partitioned between EtOAc (20 mL) and saturated Na2S2O3 (10 mL). The aqueous phase was extracted 20 with EtOAc (10 mL). The organic phases were combined, washed with saturated NaCl (2 x 10 mL), dried (Na2SO4), and concentrated. Flash chromatography (silica, 7/1 petroleum ether-EtOAc) gave white solids (0.2634 g) containing 84% of the desired bromide (as determined by integration of 19F NMR peaks).

NMR (CDC13) b ppm: 1H (400 MHz) 7.93 (m, 1 H), 7.88 (m, 1H), 7.79 (dd, 1H, J =
11.2, 1.6 Hz), 25 4.50 (d, 2H, J = 2.4 Hz), 3.94 (s, 3H); 19F (376 MHz) -108.4 (m).

Step d)Methyl 3-fluoro- 4-[2-(4-methylpiperazin-1-yl)-thiazol-4-yllbenzoate EtOH (5.0 mL) was added to the bromoketone above (193 mg, 0.70 mmol) and 4-methyl-piperazine-1-carbothioic acid amide (113 mg, 0.71 mmol) and the mixture was heated at 70 C
for 2h 15 min. The precipitates were filtered, washed with cold EtOH, and dried under vacuum and characterized. The procedure was repeated in a larger scale for 1.75 g bromoketone (6.36 mmol).
NMR (1/1 CDC13- CD3OD) b ppm: 1H (400 MHz) 8.20 (m, 1H), 7.86 (dd, 1H, J =
8.4, 1.6 Hz), 7.76 (dd, 1 H, J = 11.4, 1.8 Hz), 7.38 (d, 1 H, J = 2.4 Hz), 4.23 (br, 2H), 3.95, (s, 3H), 3.65 (br, 4H), 3.32 (br, 2H), 2.98 (s, 3H); 19F (376 MHz) -114.0 (m). LCMS [M+H]+ = 336.

The precipitates from both preparations were combined and suspended in saturated NaHCO3 (50 mL). The mixture was extracted with EtOAc. The organic phase was washed with water, dried (Na2SO4), and evaporated to give the title compound as cream solids (1.76 g).

Step e)3-fluoro- 4-[2-(4-methylpiperazin-1-yl)-thiazol-4-yllbenzoic acid HCI
salt The methyl ester (1.76 g, 5.25 mmol) from (d) was heated at 80 C with 6M HCI
(40 mL) for 5.5 h. More 6M HCI (10 mL) was added and the mixture was heated at 90 C for 1 h 15 min. After cooling, the mixture was then evaporated under vacuum and freeze-dried from water to give the final product as cream solids in quantitative yield.

NMR (DMSO-d6) b ppm: 1H (400 MHz) 11.60 (br, 1 H), 8.18 (t, 1 H, J = 8.0 Hz), 7.82 (dd, 1 H, J
= 8.4, 1.6 Hz), 7.72 (dd, 1 H, J = 12.0, 1.6 Hz), 7.48 (d, 1 H, J = 2.8 Hz), 4.11 (m, 2H), 3.58 (m, 2H), 3.49 (m, 2H), 3.19 (m, 2H), 2.80 (d, 3H, J = 4.4 Hz); 19F (376 MHz) -113.5 (m); 13C (100 MHz) 168.9, 166.0, 159.0 (d, J = 250 Hz), 143.4, 131.4 (d, J = 8 Hz), 129.8, 125.8 (d, J = 11 Hz), 125.6, 116.6 (d, J = 24 Hz), 111.1 (J = 15 Hz), 51.1, 45.0, 41.9. LCMS
[M+H]+ = 322.
Reference Example 3 6-aldehyde- intermediate H
O
N
O

6-Formyl-3,3-dimethoxy-hexahydro-furo[3,2-b]pyrrole-4-carboxylic acid tert-butyl ester Step a O
H
OuN ` O
O H
(3as, 6aS)-6R-benzyloxy-3-oxo-hexahydro-furo[3,2-blpvrrole-4-carboxylic acid benzyl ester Dess-Martin reagent (12.5 g, 30 mmol) was dissolved in DCM (250 mL). 6-Benzyloxy-3-hydroxy-hexahydro-furo[3,2-b]pyrrole-4-carboxylic acid benzyl ester (prepared as described in W005/066180) (7.4 g, 20 mmol) in DCM (50 mL) was added to a stirred solution of oxidant at rt under a nitrogen atmosphere over 45 min. After an additional 90 min stirring the reaction was deemed to be complete by TLC. Aqueous 10% Na2S2O3 (200 mL) was added and the mixture was stirred at rt for another 15 minutes. The two phase system was transferred into a separation funnel and extracted twice with EtOAc (200 mL and 100 mL
respectively). The combined organic phases were washed once with aqueous saturated NaHCO3 (100 mL) and brine (100 mL), dried over Na2SO4, filtered and the solvent removed in vacuo, yielding the crude product title compound as a clear oil (7.69 g,); ESI+, m/z: 368 (M+ +1).

Step b O
H
0"-"0 N 0 (3aS,6aS)-6R-benzyloxy-3,3-dimethoxy-hexahydro-furo[3,2-blpvrrole-4-carboxylic acid benzyl ester The keto derivative of step a) (7.6 g) was dissolved in dry methanol (100 mL).
Trimethyl orthoformate (30 mL) and pTsOH (0.2 g) was added at rt under a nitrogen atmosphere. The mixture was heated at 60 C for 8 hours. When the reaction was deemed to have reached completion according to TLC, it was cooled to rt and concentrated in vacuo.
The crude product was purified by column chromatography over silica gel eluting with ethyl acetate-heptane (1:4) which gave the title compound as a clear oil (5.9 g, 71 % over 2 steps); ESI+, m/z: 382 (M+ -OMe).
Step c HO H
O
N
O
H H O-(3aS,6aS)-3,3-dimethoxv-hexahvdro-furo[3,2-b1 pyrrol-6R-ol A solution of the compound of step b) (2.5 g, 6.4 mmol) in methanol (60 mL) and Pd(OH)2 (0.7 g) was stirred at rt under H2 atmosphere for 48 hours. When the reaction was deemed to have reached completion according to TLC, the mixture was filtered and concentrated in vacuo to yield the crude title compound as a brownish oil (1.15 g); ESI+, m/z: 190 (M+
+1).

Step d HO H
O
N
O
O H O-(3aS, 6aS)-6R-hydroxy-3,3-dimethoxv-hexahvdro-furo[3,2-blpyrrole-4-carboxylic acid tert-butyl ester 3,3-Dimethoxy-hexahydro-furo[3,2-b] pyrrol-6-ol from step c) (2.80 g, 14.8 mmol) was dissolved in 75 mL of a mixture of dioxan/water (2:1). A solution of 10%
Na2CO3 (25 mL) was added drop wise to pH 9-9.5. The mixture was cooled to 0 C in an ice-water bath and Boc anhydride was added in one portion. The reaction was stirred at rt overnight and the pH of the mixture was maintained at 9-9.5 by addition of more 10% solution of Na2CO3 if necessary. The reaction was monitored by TLC (50:50 ethyl acetate:isohexane). Once completed, the mixture was filtered to eliminate the salts formed and the solvent was evaporated in vacuo. The aqueous mixture was extracted with 3 x 100 mL EtOAc, the combined organic phases were washed with 100 mL of water and 100 mL brine, dried over Na2SO4, filtered and the solvent was evaporated in vacuo to afford 3.79 g of the title carbamate as a clear oil (89%), 94% pure (HPLC), ESI+, m/z: 312 (M++Na).
Step e O
N
O
O H O-O
3,3-Dimethoxy-6-oxo-hexahydro-furo[3,2-blpvrrole-4-carboxylic acid tert-butyl ester To the alcohol from step e) (3.674 g, 12.70 mmol) dissolved in DCM (80 mL) was added Dess-Martin Periodinane (7.00 g, 16.5 mmol) and the solution was stirred for 3 h at room temperature. The reaction was then quenched by the addition of 10% Na2S203 (aq) (150 mL) and the resulting slurry was stirred for 15 minutes. The mixture was transferred to a separation funnel and the phases were separated. The aqueous phase was extracted trice with DCM and the combined organic phases were subsequently washed twice with sat. NaHCO3 solution and were the dried, filtered, and concentrated. The crude material was purified by flash column chromatography (toluene/ethyl acetate 3:1) which gave the title compound (2.882 g, 79%).
Step f H
O
N
I O

3,3-Dimethoxy-6-methylene-hexahydro-furo[3,2-blpvrrole-4-carboxylic acid tert-butyl ester The keto compound from step e (1.10 g, 3.83 mmol) was dissolved in dry THE (30 mL) and the solution was cooled to 0 C. A solution of methyl triphenylphosphonium bromide (4.0 g, 11.2 mmol) and KOtBu (1.17 g, 10.5 mmol) in dry THE (40 mL) was added in 3 aliquots with 2 hours interval. After 6 hrs the solution was poured into a separatory funnel with diethyl ether (70 mL) and extracted with 10 % citric acid (aq)(2*40 mL). The organic phase was washed with saturated aqueous NaHCO3 (40 mL), dried with Na2SO4, filtered and the solvent was evaporated in vacuo. The crude product was purified by flash chromatography (heptane: ethyl acetate 4:1) which gave the title compound (524 mg, 48%) 1H NMR (CDC13, 400 MHz) b 1.48 (s, 9H), 3.27 (s, 3H), 3.40 (d, 3H, J = 16.6), 3.57- 3.64 (m, 1 H), 3.84 (d, 1 H, J = 9.5), 3.92 (d, 1 H, J = 16.3), 4.07- 4.25 (m, 1 H), 4.35- 4.49 (m, 1 H), 4.98 (bs, 1 H), 5.22 (d, 1 H, J = 16.4), 5.34 (s, 1 H).

Step q OH
H
O
N
J H O
O O-O
6-Hydroxymethyl-3,3-dimethoxy-hexahydro-furo[3,2-b]pyrrole-4-carboxylic acid tert-butyl ester 5 The olefin from step f) (524 mg, 1.84 mmol) was dissolved in dry THE (70 mL). 9-BBN-H (0.5 M in THF) (7.34 mL, 3.67 mmol) was added and the solution was stirred over night. The solvent was removed by rotary evaporation and redissolved in THE (20 mL). MeOH
(10 mL) was slowly added to the solution and when the gas evolution had ceased, H2O
(20 mL) was added to the solution followed by NaBO3. The solution was filtered after it had been stirred for 10 18 hrs and the filtrate was diluted with EtOAc (70 mL) and washed with brine (2*50 mL). The organic phase was dried with Na2SO4, filtered and the solvent was evaporated in vacuo. The crude product was purified by flash chromatography (heptane: ethyl acetate 2:1) which gave the title compound (477 mg, 86%).
1H NMR (CDC13, 400 MHz) 61.47 (s, 9H), 2.09- 2.25 (m, 2H), 3.02- 3.20 (m, 1 H), 3.29 (s, 3H), 15 3.39 (s, 3H), 3.65- 3.93 (m, 4H), 4.44 (d, 1 H, J = 5.7), 4.70- 4.84 (m, 1 H).

Step h H
O
N
J H O
O
O
6-Formyl-3,3-dimethoxy-hexahydro-furo[3,2-blpyrrole-4-carboxylic acid tert-butyl ester To a solution of the alcohol of step g) 1 g (370 mg, 1.22 mmol) dissolved in dry DCM (10 mL) was added Dess Martin periodinane (673 mg, 1.59 mmol). The reaction was stirred for 40 minutes and then quenched by addition of 10 mL of 10% Na2S203: NaHC03(sat) 1:1. The solution was diluted with DCM (50 mL) and extracted with a 1:1 mixture of 10%
Na2S203:
NaHCO3(sat) (50 mL). The organic phase was dried with Na2SO4, filtered and evaporated. The crude product was purified by flash chromatography (heptane: ethyl acetate (2:1) which gave the title compound (290 mg, 79%).

1H NMR (CDC13, 400 MHz) 61.47 (s, 9H), 2.90- 3.06 (m, 1 H), 3.29 (s, 3H), 3.38 (s, 3H), 3.67-3.85 (m, 2H), 3.88- 4.55 (m, 3H), 4.93- 5.19 (m, 1 H), 9.64* and 9.80* (s, 1 H). * Two peaks due to rota m ers.

Reference Example 4 6-nitrile P1 building block N
H O
N
H
O~O
Step a) NOH
H
O
N
boc H O O
\
515 mg (1.71 mmol) of 6-Formyl-3,3-dimethoxy-hexahydro-furo[3,2-b]pyrrole-4-carboxylic acid tert-butyl ester from reference example 3,_130.6 mg (1.88 mmol) of hydroxylamine hydrochloride and 168 mg (2.05 mmol) of sodium acetate were stirred in ethanol-water mixtute (10/15 ml) at room temperature overnight. The mixture was evaporated and distributed between water and ethyl acetate phases. The organic phase was washed with brine, dried over sodium sulfate, evaporated and purified on silica (EtOAc-hexane 1:1) to give the mixture of cis and trans-isomers of the above depicted. Rf 0.43 and 0.48 (EtOAc-hexane 1:1). Yield 395 mg (73%) NC
H
O
N
boc H 00 The oxime of step a) (114 mg, 0.36 mmol) and triethylamine (105 L, 0.757 mmol) were dissolved in 1,5 ml of dichloromethane and cooled till -78 C.
Trifluoromethanesulfonic anhydride (61 L, 0.36 mmol) in 600 .tL of dichloromethane was added dropwise over 7 min.
The reaction mixture was allowed to warm up till room temperature and was stirred for 2 hours.
The mixture was diluted with 15 ml of DCM, washed with precooled 5% citric acid, sodium bicarbonate, brine, dried over sodium sulfate, evaporated and purified on silica (gradient EtOAc-hexane 1:3 to 1:1). Rf 0.65 (EtOAc-hexane 1:1). Yield 65 mg (61%) Reference Example 5 A typical P1/P2 deprotection and coupling /N
O H
~ON N O
H H:
O
%
The nitrile building block of reference example 4 (95 mg, 0.032 mmol) was dissolved in 5 ml of methanol cooled down to 0 C and 0.5 ml of acetyl chloride was added dropwise.
The resulting mixture was stirred at r.t. for 4h, then evaporated. The residue was dissolved in 2.5 ml of DMF, 80 mg (0.32 mmol) of Boc-Leu-OH was added, followed by addition of 0.5 ml of diisopropylethylamine. The resulting mixture was cooled down till 0 C and 160 mg (0.42 mmol) of HATU (O-(7-Azabenzotruazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophsphate) was added. The reaction mixture was allowed to warm up till room temperature and stirred for 1.5 h. Mixture was evaporated, distributed between water and ethyl acetate phases, organic phase was washed with water, brine, dried over sodium sulfate, evaporated and purified on silica (EtOAc Rf 0.35) to give 82 mg of the title compound. Yield 82 mg (62%) for 2 steps.
Reference Example 6 An alternative P3 building block o O O
0i03 F OCH3 iii.
O O

F I~ OCH3 F I~ OCH3 F OH iv. Br /

O S S
yN yN
\N) \N) i. AcOH, bromine, RT, 2 h, 55 % yield; ii. KF, acetonitrile, 18-crown-6, 90 C, 16 h; 31 % yield;
iii. AcOH, bromine, 45 C, 4 h, 100 % yield; iv. 4-Methyl-piperazine-1-carbothioic acid amide, A, 2 h, 74 % yield; LiOH, RT, 16 h, 100 % yield.

Availability of starting materials -Methyl 4-acetylbenzoate is available from Aldrich; 4-methyl-piperazine-1-carbothioic acid amide - 11 suppliers found in SciFinder (perhaps Chem Pur Products Ltd in Germany most convienient).
Step a) 4-(2-Bromo-acetyl)-benzoic acid methyl ester O
Br I OMe O
To a solution of 4-acetyl-benzoic acid methyl ester (8.4 mmol) in acetic acid (20 ml-) was added bromine (8.4 mmol). The reaction was stirred at RT for 2 h over which time the red colour disappeared and an off white precipitate formed. The product was collected by filtration and washed with cold methanol/water (200 mL 1:1) to yield a white powder (55 %). 1 H NMR
(400MHz, CDC13) 3.98 (3H, s), 4.20 (2H, s), 8.02 (2H, d, J = 8Hz), 8.18 (2H, d, J = 8Hz).

Step b) 4-(2-Fluoro-acetyl)-benzoic acid methyl ester O
F ,e We i To a suspension of potassium fluoride (3.11 mmol) in acetonitrile (1 ml-) was added 18-crown-6 (0.1 mmol) and the reaction was heated at 90 C for 30 mins. 4-(2-Bromo-acetyl)-benzoic acid (1.56 mmol) was added and the reaction heated at 90 C for 16 h. The reaction was diluted with water (10 ml-) and extracted with ethyl acetate (3 x 20 mL). The product was purified on silica eluting with 5-15 % ethyl acetate in iso-hexane to yield on concentration in vacuo of the desired fractions, the title product as a white solid (31 %). 1H
NMR (400MHz, CDC13) 3.98 (3H, s), 5.55 (2H, d, J = 50Hz), 7.95 (2H, d, J = 8Hz), 8.18 (2H, d, J = 8Hz).

Step c) 4-(2-Bromo-2-fluoro-acetyl)-benzoic acid methyl ester O
F I OMe Br O
To a suspension of 4-(2-fluoro-acetyl)-benzoic acid (1.19 mmol) in acetic acid (5 ml-) was added bromine (1.19 mmol). The reaction was heated at 45 C for 4 h over which time a green solution formed. The reaction was concentrated in vacuo and azeotroped twice with toluene to yield the title compound as a green solid (100 %). The product was used crude in the next step. 1 H NMR (400MHz, CDC13) 3.98 (3H, s), 7.04 (1 H, s), 8.05 - 8.10 (4H, m).

Step d)4-[5-Fluoro-2-(4-methyl-piperazin-1-yl)-thiazol-4-yl]-benzoic acid methyl ester F I OMe s )=N
N) NJ
/
4-(2-Bromo-2-fluoro-acetyl)-benzoic acid methyl ester (1.18 mmol) and 4-methyl-piperazine-1-carbothioic acid amide (1.18 mmol) were dissolved in ethanol (10 mL). The reaction was heated at reflux for 2 h. The reaction was cooled to RT causing the product to precipitate. The product was collected by filtration and washed with cold ethanol. The product was given an aqueous sodium bicarbonate work up to yield the title compound as a colourless oil (74 %).
MS (ES+) 337 (M+H, 100%).

Step f) 4-[5-Fluoro-2-(4-methyl-piperazin-1-yl)-thiazol-4-yl]-benzoic acid di-hydrochloride O
F I OH

~, N
0 N>
NJ
/
To a solution of 4-[5-fluoro-2-(4-methyl-piperazin-1-yl)-thiazol-4-yl]-benzoic acid methyl ester (0.43 mmol) in tetrahydrofuran/water (2.5 mL, 4:1) was added lithium hydroxide (0.5 mmol).
The reaction was stirred at RT for 16 h. The reaction was concentrated in vacuo and hydrochloric acid (2N, 3 ml-) was added causing the product to precipitate as a white solid.
The product was collected by filtration to yield the title product as a white solid (79 %). MS
(ES+) 322 (M+H, 100%).

Example 1 N
O H
\ N N O
N N / 0 H,.
NI o S
N41-(6-cyano-3-oxo-hexahyd ro-fu rof3,2-blpyrrole-4-carbonyl)-3-methyl-butyll-4-f2-(4-methyl-piperazin-1-yl)thiazol-4-yll-benzamide Step a) /N
/
O H
\ N N O
N/~ N I / H O H,, N I i0 0 S

The P1/P2 building block of reference example 5 (60 mg, 0.147 mmol) was dissolved in 3 ml of methanol, cooled down to 0 C and 0.4 mL of acetyl chloride was added dropwise. The 10 resulting mixture was stirred at r.t. for 4h, then evaporated. The residue was dissolved in 2.5 mL of DMF, 52 mg (0.147 mmol) of the P3 building block (prepared as in W00566180 as the acid was added, followed by addition of 0.5 mL of diisopropylethylamine. The resulting mixture was cooled down till 0 C and 70 mg (0.184 mmol) of HATU (O-(7-azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophsphate) was added. The reaction mixture was 15 allowed to warm up till room temperature and stirred for 1.5 h. The mixture was evaporated, distributed between water and ethyl acetate phases, and the organic phase washed with water, brine, dried over sodium sulfate, evaporated and purified on silica (5%
MeOH in EtOAc).
The desired fractions were combined and concentrated in vacuo to afford the desired material in a yield of 56 mg (64 %), LC/MS 597 (M+1) Step b) N
O H
\ N N O
N I / H 0 H,, S

The ketal of step a) (56 mg, 0.094 mmol) was treated with 3 mL of TFA-water mixture (2.5%
water in TFA) for 4 h. The reaction was monitored by LC/MS. The reaction mixture was evaporated, dissolved in acetonitrile (5 mL), stirred with solid sodium carbonate for 1 h, then solids were filtered off, the mother liquor was concentrated in vacuo, and purified by preparative HPLC (NH4OAc buffer, 30-80 system (MeCN-water) to give 25 mg of desired product (yield 45 %). LC/MS M+1 551, M+19 569 (hydrate form) Example 2 NC

NC H NC ~/ N~T YNC N~-HO /\) N O Ac CI N HATU-coupling O N O 0 TFA/ H2O 0 H O McOH H O H N 0 HN O /
H N O O H

boc HCI I ,'N F
N \ N F
(2) ~N/ S r-' N
~S
(3) (4) The protected P1-P2 building block of reference example 5 (1) (33 mg, 0.08mmol) was dissolved in 3 ml of methanol cooled down to 0 C and 0.4 ml of acetyl chloride was added dropwise acid was added. The resulting mixture was stirred at r.t. for 4h, then evaporated. The residue was dissolved in 2.5 ml of DMF, 29 mg (0.08 mmol) of the P3 acid of reference example 2 (as a HCI salt) was added, followed by addition of 0.5 ml of diisopropylethylamine.
The resulting mixture was cooled down till 0 C and 39 mg (0.101 mmol) of HATU
(O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate) was added. The reaction mixture was allowed to warmed up till room temperature and stirred for 1.5 h. Mixture was evaporated, distributed between water and ethyl acetate phases, organic phase was washed with water, brine, dried over sodium sulfate, evaporated and purified on silica (5%
MeOH in EtOAc). Yield of (3) 32mg (67%), LC/MS 615 (M+1) Ketal (3) (32 mg, 0.054mmol) was treated with 3 ml of TFA-water mixture (2,5%
water in TFA) for 4 h. The reaction was monitored by LC/MS. Reaction mixture was evaporated, dissolved in acetonitrile (5 ml), stirred with solid sodium carbonate for 1 h, then solids were filtered off, mother liquor was concentrated i vacuo, and purified on prep. LC/MS purified by prep. HPLC
(NH4OAc buffer, 30_80 system (MeCN-water) to give 13 mg of product (yield 42%). LC/MS
M+1 569, M+19 587 (hydrate form) Example 3 NC
NC H NC H O NCN~=
NN N N O AcCI HATU-coupling N O H O TFA HzOO H O McOH O H N O HN O / H ,N O H

boc N
HCI r'N~S
F N
(1) (2) -N J 'N /' S F
(5) (6) The protected P1 P2 bilding block of reference example 5 (1) (33 mg, 0.08mmol) was dissolved in 3 ml of methanol cooled down to 0 C and 0.4 ml of acetyl chloride was added dropwise acid was added. The resulting mixture was stirred at r.t. for 4h, then evaporated.
The residue was dissolved in 2.5 ml of DMF, 29 mg (0.08 mmol) of the P3 acid of reference example 6, as a HCI salt) was added, followed by addition of 0.5 ml of diisopropylethylamine.
The resulting mixture was cooled down till 0 C and 39 mg (0.101 mmol) of HATU (O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophsphate) was added. The reaction mixture was allowed to warm up till room temperature and stirred for 1.5 h. Mixture was evaporated, distributed between water and ethyl acetate phases, organic phase was washed with water, brine, dried over sodium sulfate, evaporated and purified on silica (5% MeOH
in EtOAc). Yield of (5) 30mg (63%), LC/MS 615 (M+1) Ketal (5) (30 mg, 0.051 mmol) was treated with 3 ml of TFA-water mixture (2,5%
water in TFA) for 4 h. The reaction was monitored by LC/MS. Reaction mixture was evaporated, dissolved in acetonitrile (5 ml), stirred with solid sodium carbonate for 1 h, then solids were filtered off, mother liquor was concentrated i vacuo, and purified on prep. LC/MS purified by prep. HPLC
(NH4OAc buffer, 30_80 system (MeCN-water) to give 11 mg of product (yield 38%). LC/MS
M+1 569, M+19 587 (hydrate form) Biological Examples Determination of cathepsin K of cathepsin K proteolic catalytic activitycatalic activity Convenient assays for cathepsin K are carried out using human recombinant enzyme, such as that described in PDB.

ID BC016058 standard; mRNA; HUM; 1699 BP.

DE Homo sapiens cathepsin K (pycnodysostosis), mRNA (cDNA clone MGC:23107 RX MEDLINE;. RX PUBMED; 12477932.

DR RZPD; IRALp962G1234.

DR SWISS-PROT; P43235;

The recombinant cathepsin K can be expressed in a variety of commercially available expression systems including E coli, Pichia and Baculovirus systems. The purified enzyme is activated by removal of the prosequence by conventional methods.

Standard assay conditions for the determination of kinetic constants used a fluorogenic peptide substrate, typically H-D-Ala-Leu-Lys-AMC, and were determined in either 100 mM
Mes/Tris, pH 7.0 containing 1 mM EDTA and 10 mM 2-mercaptoethanol or100mMNa phosphate, imM
EDTA, 0.1 %PEG4000 pH 6.5 or 100 mM Na acetate, pH 5.5 containing 5 mM EDTA
and 20 mM cysteine, in each case optionally with 1 M DTT as stabiliser. The enzyme concentration used was 5 nM. The stock substrate solution was prepared at 10 mM in DMSO.
Screens were carried out at a fixed substrate concentration of 60 pM and detailed kinetic studies with doubling dilutions of substrate from 250 pM. The total DMSO concentration in the assay was kept below 3%. All assays were conducted at ambient temperature. Product fluorescence (excitation at 390 nm, emission at 460 nm) was monitored with a Labsystems Fluoroskan Ascent fluorescent plate reader. Product progress curves were generated over 15 minutes following generation of AMC product.

Cathepsin S Ki determination The assay uses baculovirus-expressed human cathepsin S and the boc-Val-Leu-Lys-AMC
fluorescent substrate available from Bachem in a 384 well plate format, in which 7 test compounds can be tested in parallel with a positive control comprising a known cathepsin S
inhibitor comparator.

Substrate dilutions 280p1/well of 12.5% DMSO are added to rows B - H of two columns of a 96 deep well polypropylene plate. 70p1/well of substrate is added to row A. 2 x 250p1/well of assay buffer (100mM Na phosphate, 100mM NaCl, pH 6.5) is added to row A, mixed, and double diluted down the plate to row H.

Inhibitor dilutions 100 p1/well of assay buffer is added to columns 2-5 and 7-12 of 4 rows of a 96 well V bottom polypropylene plate. 200p1/well of assay buffer is added to columns 1 and 6.

The first test compound prepared in DMSO is added to column 1 of the top row, typically at a volume to provide between 10 and 30 times the initially determined rough K.
The rough Ki is calculated from a preliminary run in which 10 pl/well of 1mM boc-VLK-AMC (1/10 dilution of 10 mM stock in DMSO diluted into assay buffer) is dispensed to rows B to H and 20 pl/well to row A of a 96 well Microfluor TM plate. 2 pl of each 10mM test compound is added to a separate well on row A, columns 1-10. Add 90 pl assay buffer containing 1 mM DTT and 2 nM cathepsin S to each well of rows B-H and 180 pl to row A.Mix row A using a multichannel pipette and double dilute to row G. Mix row H and read in the fluorescent spectrophotometer. The readings are Prism data fitted to the competitive inhibition equation, setting S = 100 pM and KM
= 100 pM to obtain an estimate of the K;, up to a maximum of 100 pM.

The second test compound is added to column 6 of the top row, the third to column 1 of the second row etc. Add 1 pl of comparator to column 6 of the bottom row. Mix column 1 and double dilute to column 5. Mix column 6 and double dilute to column 10.

Using an 8-channel multistepping pipette set to 5 x 10 pl, distribute 10 pl/well of substrate to the 384 well assay plate. Distribute the first column of the substrate dilution plate to all columns of the assay plate starting at row A. The tip spacing of the multichannel pipette will correctly skip alternate rows. Distribute the second column to all columns starting at row B.
Using a 12-channel multistepping pipette set to 4 x 1 Opl, distribute 1 Opl/well of inhibitor to the 384 well assay plate. Distribute the first row of the inhibitor dilution plate to alternate rows of the assay plate starting at Al. The tip spacing of the multichannel pipette will correctly skip alternate columns. Similarly, distribute the second, third and fourth rows to alternate rows and columns starting at A2, B1 and B2 respectively.

Mix 20 ml assay buffer and 20 pl 1 M DTT. Add sufficient cathepsin S to give 2 nM final concentration.

Using the a distributor such as a Multidrop 384, add 30 pl/well to all wells of the assay plate and read in fluorescent spectrophotomoter such as an Ascent.

Fluorescent readings, (excitation and emission wavelengths 390nm and 460nm respectively, set using bandpass filters) reflecting the extent of enzyme cleavage of the fluorescent substrate, notwithstanding the inhibitor, are linear rate fitted for each well.

Fitted rates for all wells for each inhibitor are fitted to the competitive inhibition equation using 5 SigmaPlot 2000 to determine V, Km and Ki values.

Cathepsin L Ki The procedure above with the following amendments is used for the determination of Ki for cathepsin L.

The enzyme is commercially available human cathepsin L (for example Calbiochem). The 10 substrate is H-D-Val-Leu-Lys-AMC available from Bahcem. The assay buffer is 100mM sodium acetate 1 mM EDTA, pH5.5) The DMSO stock (10mM in 100%DMSO) is diluted to 10%
in assay buffer. Enzyme is prepared at 5nM concentration in assay buffer plus 1 mM dithiothreitol just before use. 2u1 of 10mM inhibitor made up in 100% DMSO is dispensed into row A. 1 Opl of pM substrate (=1/200 dilution of 10 mM stock in DMSO,diluted in assay buffer) 15 Inhibition Studies Potential inhibitors are screened using the above assay with variable concentrations of test compound. Reactions were initiated by addition of enzyme to buffered solutions of substrate and inhibitor. K; values were calculated according to equation 1.

VS
vo =
KM I+ I +S (1) Ki where vo is the velocity of the reaction, V is the maximal velocity, S is the concentration of 20 substrate with Michaelis constant of KM, and I is the concentration of inhibitor.

Results are presented as:
A under 50 nanomolar B 50-500 nanomolar C 501-1000 nanomolar 25 D 1001 - 5000 nanomolar E 5001 - 10 000 nanomolar F in excess of 10 000 nanomolar Example Test Number Ki cathepsin K Ki cathepsin S Ki cathepsin L
1 Test 1 A D B
1 Test2 3.0nM 3300nM 530nM
The compounds of formula II are thus potent inhibitors of cathepsin K and yet selective over the closely related cathepsin S and L.

Metabolic Stability Compounds of the invention and the indicated comparative examples were tested for metabolic stability in a cytosol assay in which the compounds were incubated with commercially available human hepatic cytosol fractions and the disappearance of the compound monitored by HPLC or LC/MS. Pooled human liver cytosol fractions are less likely to represent outlier individuals than blood from a single individual and can be stored frozen, unlike whole blood. The cytosol assay thus provides a consistent assay testbed as a guide to the stability of a compound in the in vivo environment, such as when exposed to whole blood.

In short, test compounds (2 pM) are incubated in pooled human liver cytosol (Xenotech LLC
Lenexa US, 1 mg/mL protein in 0.1 M phosphate buffer, pH 7.4) at 37 centigrade over a one hour period. The incubations are initiated by the addition of 1 mM NADPH co-factor. Timed sub-samples were taken at 0, 20, 40 and 60 minutes and "crash precipitated" by the addition of 3 volumes of ice-cold acetonitrile. The samples were centrifuged at reduced temperature and the supernatants were separated and analyzed by LC-MS-MS.

Alternatively, an analogous stability assay is carried out in human or monkey whole blood and/or commerically available liver microsomes.

Example Structure CLint CLint whole blood HLM
uI/min/mg uI/min/mg comparative 9 49 H
example O O
N
r ~N \ N I/ H O H
S

Example 1 INI NA NA
H
O O

/~ N
_-N \ N I/ O H H O
H
/N-_~
s The Comparative Example represents a compound bearing a carbon-carbon bond at the 6 position within the scope of W02008/007107 cited above. It was prepared in a facile manner from compound 1 d (scheme 1). Hence with the exocyclic alkene 1 d in hand, stereoselective hydrogenation of the alkene with Adams' catalyst (platinum dioxide) in ethyl acetate under a hydrogen atmosphere, proceeded with syn addition of hydrogen. This hydrogenation afforded essentially one product, namely the C-6 methyl isomer (LCMS [M+H] = 288 found) with R-stereochemistry in good yield. The facial selectivity seen here for the hydrogenation step, is similar to that reported previously in the literature for a closely related bicyclic structure (Srinivas et al, Synlett, 1999, 555-556). The thus prepared building block was deprotected, elongated and oxidised to the active keto form as for the compopunds of the invention exemplified above.

Improved stability in vivo allows for a better distribution of the compound in the body throughout the day, notwithstanding QD or BID dosing. This is particularly important for indications such as osteoporosis where diurnal variation is significant.

Permeability This experiment measures transport of inhibitors through the cells of the human gastroenteric canal. The assay uses the well known Caco-2 cells with a passage number between 40 and 60.

Apical to basolateral transport Generally every compound will be tested in 2-4 wells. The basolateral and the apical wells will contain 1.5 mL and 0.4 mL transport buffer (TB), respectively, and the standard concentration of the tested substances is 10 M. Furthermore all test solutions and buffers will contain 1 %
DMSO. Prior to the experiment the transport plates are pre-coated with culture medium containing 10% serum for 30 minutes to avoid nonspecific binding to plastic material. After 21 to 28 days in culture on filter supports the cells are ready for permeability experiments.
Transport plate no 1 comprises 3 rows of 4 wells each. Row 1 is denoted Wash, row 2 "30 minutes" and row 3 "60 minutes". Transport plate no 2 comprises 3 rows of 4 wells, one denoted row 4 "90 minutes", row 5 "120 minutes and the remaining row unassigned.

The culture medium from the apical wells is removed and the inserts are transferred to a wash row (No. 1) in a transport plate (plate no.1) out of 2 plates without inserts, which have already been prepared with 1.5 mL transport buffer (HBSS, 25 mM HEPES, pH 7.4) in rows 1 to 5. In A-B screening the TB in basolateral well also contains 1 % Bovine Serum Albumin.

0.5 mL transport buffer (HBSS, 25 mM MES, pH 6.5) is added to the inserts and the cell monolayers equilibrated in the transport buffer system for 30 minutes at 37 C
in a polymix shaker. After being equilibrated to the buffer system the Transepithelial electrical resistance value (TEER) is measured in each well by an EVOM chop stick instrument. The TEER values are usually between 400 to 1000 S2 per well (depends on passage number used).

The transport buffer (TB, pH 6.5) is removed from the apical side and the insert is transferred to the 30 minutes row (No. 2) and fresh 425 .tL TB (pH 6.5), including the test substance is added to the apical (donor) well. The plates are incubated in a polymix shaker at 37 C with a low shaking velocity of approximately 150 to 300 rpm.

After 30 minutes incubation in row 2 the inserts will be moved to new pre-warmed basolateral (receiver) wells every 30 minutes; row 3 (60 minutes), 4 (90 minutes) and 5 (120 minutes).
25 .tL samples will be taken from the apical solution after -2 minutes and at the end of the experiment. These samples represent donor samples from the start and the end of the experiment.

300 .tL will be taken from the basolateral (receiver) wells at each scheduled time point and the post value of TEER is measured at the end the experiment. To all collected samples acetonitrile will be added to a final concentration of 50% in the samples. The collected samples will be stored at -20 C until analysis by HPLC or LC-MS.

Basolateral to apical transport Generally every compound will be tested in 2-4 wells. The basolateral and the apical wells will contain 1.55 mL and 0.4 mL TB, respectively, and the standard concentration of the tested substances is 10 .tM. Furthermore all test solutions and buffers will contain 1 % DMSO. Prior to the experiment the transport plates are precoated with culture medium containing 10% serum for 30 minutes to avoid nonspecific binding to plastic material.

After 21 to 28 days in culture on filter supports the cells are ready for permeability experiments.
The culture medium from the apical wells are removed and the inserts are transferred to a wash row (No.1) in a new plate without inserts (Transport plate).

The transport plate comprises 3 rows of 4 wells. Row 1 is denoted "wash" and row 3 is the "experimental row". The transport plate has previously been prepared with 1.5 mL TB (pH 7.4) in wash row No. 1 and with 1.55 mL TB (pH 7.4), including the test substance, in experimental row No. 3 (donor side).

0.5 mL transport buffer (HBSS, 25 mM MES, pH 6.5) is added to the inserts in row No. 1 and the cell monolayers are equilibrated in the transport buffer system for 30 minutes, 37 C in a polymix shaker. After being equilibrated to the buffer system the TEER value is measured in each well by an EVOM chop stick instrument.

The transport buffer (TB, pH 6.5) is removed from the apical side and the insert is transferred to row 3 and 400 .tL fresh TB, pH 6.5 is added to the inserts. After 30 minutes 250 .tL is withdrawn from the apical (receiver) well and replaced by fresh transport buffer. Thereafter 250 L samples will be withdrawn and replaced by fresh transport buffer every 30 minutes until the end of the experiment at 120 minutes, and finally a post value of TEER is measured at the end of the experiment. A 25 .tL samples will be taken from the basolateral (donor) compartment after -2 minutes and at the end of the experiment. These samples represent donor samples from the start and the end of the experiment.

To all collected samples acetonitrile will be added to a final concentration of 50% in the samples. The collected samples will be stored at -20 C until analysis by HPLC
or LC-MS.
Calculation Determination of the cumulative fraction absorbed, FAcum, versus time. FAcum is calculated from:

Y CRI
FACUM
CDI
Where CRi is the receiver concentration at the end of the interval i and CDi is the donor concentration at the beginning of interval i. A linear relationship should be obtained.
The determination of permeability coefficients (Papp, cm/s) are calculated from:

(k - VR) 5 Papp = (A . 60) where k is the transport rate (min-1) defined as the slope obtained by linear regression of cumulative fraction absorbed (FAcum ) as a function of time (min), VR is the volume in the receiver chamber (mL), and A is the area of the filter (cm2).

Reference compounds Category of absorption in Markers % absorption in man man PASSIVE TRANSPORT
Low (0-20%) Mannitol 16 Methotrexate 20 Moderate (21-75%) Acyclovir 30 High (76-100%) Propranolol 90 Caffeine 100 ACTIVE TRANSPORT
Amino acid transporter L-Phenylalanine 100 ACTIVE EFFLUX
PGP-MDR1 Digoxin 30 Greater permeability through the gastrointestinal tissue is advantageous in that it allows for the use of a smaller dose to achieve similar levels of exposure to a less permeable compound administered in a higher dose. A low dose is advantageous in that minimises the cost of goods for a daily dose, which is a crucial parameter in a drug which is taken for protracted time periods.

Mutagenicity The mutagenic potential of compounds is conveniently tested in the Ames Test, typically carried out in a variety of bacterial strains such as Salmonella typhimurium TA100, TA102, TA
1535, TA 1537 with and without liver S9 fraction activation, for example at 30, 300 and 3000 ug/plate concentrations.

Ames testing is readily available at a number of CROs around the world.
Abbreviations DMF dimethylformamide DCM dichloromethane TBDMS tert-butyldimethylsilyl RT room temperature THE tetrahydrofuran Ac acetyl TLC thin layer chromatography DMAP dimethylaminopyridine EtOAc ethyl acetate um micromolar All references referred to in this application, including patents and patent applications, are incorporated herein by reference to the fullest extent possible.

Throughout the specification and the claims which follow, unless the context requires otherwise, the word `comprise', and variations such as `comprises' and `comprising', will be understood to imply the inclusion of a stated integer, step, group of integers or group of steps but not to the exclusion of any other integer, step, group of integers or group of steps.

Claims (15)

  1. Claims A compound of formula II:

    wherein R3 is C1-C3 alkyl or C3-C6 cycloalkyl, either of which is optionally substituted with one or two methyl and/or a fluoro, trifluoromethyl or methoxy, when R3 is C3-C6 cycloalkyl it may alternatively be gem subsituted with fluoro;
    R4 is methyl or fluoro; m is 0, 1 or 2;
    E is a bond, or thiazolyl, optionally substituted with methyl or fluoro;
    A1 is CH or N, A2 is CR6R7 or NR6, provided at least one of A1 and A2 comprises N;
    n is 0 or 1 such that the ring containing A1 and A2 is a saturated, nitrogen-containing ring of 5 or 6 ring atoms;
    R6 is H, C1-C4 alkyl, C1-C4 haloalkyl, C1-C3 alkyl-O-C1-C3 alkyl, or when A2 is C, R6 can also be C1-C4 alkoxy or F;
    R7 is H, C1-C4 alkyl or F;
    or a pharmaceutically acceptable salt, N-oxide or hydrate thereof.
  2. 2. A compound according to claim 1, with the formula IIa:

    wherein R3 is branched C2-C6 alkyl or C3-C6 cycloalkyl, either of which is optionally substituted with one or two fluoro or with a trifluoromethyl;
    R4 is methyl or fluoro; m is 0, 1 or 2;
    R5 is H, methyl or fluoro;
    R6 is C1-C6 alkyl;
    or a pharmaceutically acceptable salt, N-oxide or hydrate thereof.
  3. 3. A compound according to any one of the preceding claims, wherein R3 is the side chain of leucine.
  4. 4. A compound according to any one of the preceding claims, wherein m represents 0 and R5 represents F.
  5. 5. A compound according to any one of claims 1 to 4, wherein n represents 1, R4 is F and R5 is H.
  6. 6. A compound according to claim 5, wherein R4 is positioned as shown by the partial structure:

  7. 7. A compound according to any of claims 2-6, wherein R6 is CH3.
  8. 8. A compound according to claim 1 which is selected from:

    or a pharmaceutically acceptable salt, hydrate or N-oxide thereof.
  9. 9. A pharmaceutical composition comprising a compound as defined in any preceding claim and a pharmaceutically acceptable carrier or diluent therefor.
  10. 10. Use of a compound as defined in any of claims 1 to 8 in the manufacture of a medicament for the treatment or prevention of disorder selected from:
    osteoporosis, gingival diseases (such as gingivitis and periodontitis), Paget's disease, hypercalcaemia of malignancy, metabolic bone disease, diseases characterised by excessive cartilage or matrix degradation (such as osteoarthritis and rheumatoid arthritis), bone cancers including neoplasia, pain (especially chronic pain).
  11. 11. A compound according to any one of claims 1 to 8 for use in the treatment or prevention of a disorder selected from:
    osteoporosis, gingival diseases (such as gingivitis and periodontitis), Paget's disease, hypercalcaemia of malignancy, metabolic bone disease, diseases characterised by excessive cartilage or matrix degradation (such as osteoarthritis and rheumatoid arthritis), bone cancers including neoplasia, pain (especially chronic pain).
  12. 12. A method for the treatment of a disorder mediated by cathepsin K
    comprising administering a safe and effective amount of a compound according to any one of claims 1 to 8 to a subject in need thereof.
  13. 13. The method of claim 12, wherein the disorder is selected from:
    osteoporosis, gingival diseases (such as gingivitis and periodontitis), Paget's disease, hypercalcaemia of malignancy, metabolic bone disease, diseases characterised by excessive cartilage or matrix degradation (such as osteoarthritis and rheumatoid arthritis), bone cancers including neoplasia, pain (especially chronic pain).
  14. 14. A compound of the formula:

    wherein the Rb groups define a ketal, such as the bis methyl ketal or together define a cyclic ketal such as 1,3-dioxolane, or an N-protected derivative thereof.
  15. 15. A compound according to claim 14 which is:

CA2738025A 2008-09-24 2009-09-24 Protease inhibitors Abandoned CA2738025A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB0817425.2A GB0817425D0 (en) 2008-09-24 2008-09-24 Protease inhibitors
GB0817425.2 2008-09-24
PCT/EP2009/062407 WO2010034789A1 (en) 2008-09-24 2009-09-24 Protease inhibitors

Publications (1)

Publication Number Publication Date
CA2738025A1 true CA2738025A1 (en) 2010-04-01

Family

ID=39952075

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2738025A Abandoned CA2738025A1 (en) 2008-09-24 2009-09-24 Protease inhibitors

Country Status (10)

Country Link
EP (1) EP2350089A1 (en)
JP (1) JP2012503626A (en)
KR (1) KR20110059657A (en)
CN (1) CN102224155A (en)
AU (1) AU2009295899A1 (en)
BR (1) BRPI0919073A2 (en)
CA (1) CA2738025A1 (en)
EA (1) EA201170480A1 (en)
GB (1) GB0817425D0 (en)
WO (1) WO2010034789A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201314503D0 (en) * 2013-08-13 2013-09-25 Medivir Ab Cysteine protease inhibitor salt
US20220193048A1 (en) 2019-04-05 2022-06-23 Universite De Bretagne Occidentale Protease-activated receptor-2 inhibitors for the treatment of sensory neuropathy induced by a marine neurotoxic poisoning

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0003111D0 (en) * 2000-02-10 2000-03-29 Novartis Ag Organic compounds
SE0400022D0 (en) * 2004-01-08 2004-01-08 Medivir Ab New compounds
GB0614052D0 (en) * 2006-07-14 2006-08-23 Amura Therapeutics Ltd Compounds

Also Published As

Publication number Publication date
JP2012503626A (en) 2012-02-09
AU2009295899A1 (en) 2010-04-01
CN102224155A (en) 2011-10-19
EA201170480A1 (en) 2011-12-30
KR20110059657A (en) 2011-06-02
GB0817425D0 (en) 2008-10-29
WO2010034789A1 (en) 2010-04-01
BRPI0919073A2 (en) 2016-03-15
EP2350089A1 (en) 2011-08-03

Similar Documents

Publication Publication Date Title
US10723709B2 (en) Protease inhibitors
CA2690344C (en) Cysteine protease inhibitors
CA2738025A1 (en) Protease inhibitors
WO2008114054A1 (en) Protease inhibitors

Legal Events

Date Code Title Description
FZDE Discontinued

Effective date: 20130924