WO2016189304A1 - Antibacterial compounds - Google Patents

Antibacterial compounds Download PDF

Info

Publication number
WO2016189304A1
WO2016189304A1 PCT/GB2016/051514 GB2016051514W WO2016189304A1 WO 2016189304 A1 WO2016189304 A1 WO 2016189304A1 GB 2016051514 W GB2016051514 W GB 2016051514W WO 2016189304 A1 WO2016189304 A1 WO 2016189304A1
Authority
WO
WIPO (PCT)
Prior art keywords
independently
compound
alkyl
group
mmol
Prior art date
Application number
PCT/GB2016/051514
Other languages
French (fr)
Inventor
Ian Cooper
Rolf Peter Walker
Mark Pichowicz
Andrew James Ratcliffe
Frederik Deroose
Charles John Robert Hedgecock
Peter Brandt
Johan Georg GISING
Original Assignee
Redx Pharma Plc
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 Redx Pharma Plc filed Critical Redx Pharma Plc
Publication of WO2016189304A1 publication Critical patent/WO2016189304A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents

Definitions

  • Antibacterial compounds This invention relates to antibacterial drug compounds containing a polycyclic ring system incorporating a macrocyclic ring. It also relates to pharmaceutical formulations of antibacterial drug compounds. It also relates to uses of the derivatives in treating bacterial infections and in methods of treating bacterial infections. The invention is also directed to antibacterial drug compounds which are capable of treating bacterial infections which are currently hard to treat with existing drug compounds. Such infections are frequently referred to as resistant strains. The increasing occurrence of bacterial resistance to antibiotics is viewed by many as being one of the most serious threats to the future health and happiness of centuries. Multidrug resistance has become common among some pathogens, e.g.
  • Staphylococcus aureus Streptococcus pneumoniae, Clostridium difficile and Pseudomonas aeruginosa.
  • Staphylococcus aureus a Gram positive bacterium
  • MRSA methicillin resistant Staphylococcus aureus
  • antibiotic resistant Gram negative strains such as either Escherichia coli NDM-1 (New Delhi metallo- ⁇ -lactamase) or Klebsiella pneumoniae NDM-1
  • Escherichia coli NDM-1 New Delhi metallo- ⁇ -lactamase
  • Klebsiella pneumoniae NDM-1 are also very difficult to treat.
  • antibiotics such as vancomycin and colistin are effective against these strains.
  • the fluoroquinolone antibacterial family are synthetic broad-spectrum antibiotics. They were originally introduced to treat Gram negative bacterial infections, but are also used for the treatment of Gram positive strains.
  • One problem with existing fluoroquinolones can be the negative side effects that may sometimes occur as a result of fluoroquinolone use. In general, the common side-effects are mild to moderate but, on occasion, more serious adverse effects occur.
  • CNS central nervous system
  • MRSA central nervous system
  • ⁇ -lactam antibiotics such as methicillin.
  • Bacterial resistance is also becoming a problem in the treatment of animals. Antibacterials find widespread use in industrial farming, e.g. to prevent mastitis in dairy cattle, where they are often used prophylactically. Such widespread prophylactic use has led to the build-up of resistance in certain bacterial strains which are particularly relevant to animal health.
  • a further aim of certain embodiments of this invention is to provide antibiotics in which the metabolised fragment or fragments of the drug after absorption are GRAS (Generally Regarded As Safe). Certain embodiments of the present invention satisfy some or all of the above aims.
  • the invention provides a compound of formula (I), or a pharmaceutically acceptable salt or N-oxide thereof:
  • group A is selected from a 5-12heterocycloalkyl group comprising at least one nitrogen in the ring system and a 3-6heteroalkyl group comprising at least one nitrogen in the linking chain;
  • L 3 is attached by a covalent bond to an atom selected from the nitrogen and carbon atoms which form the group A ring system or linking chain;
  • Z is independently selected from N and CR 2 ;
  • R 1 and R 2 are each independently selected from: H, C1-C4-alkyl, halogen, OR 8 , NR 8 R 9 and C 1 -C 4 -haloalkyl;
  • X 1 , X 2 , X 3 and X 4 are each independently selected from: N and CR 10 ; wherein no more than two of X 1 , X 2 , X 3 and X 4 are N; wherein a single one of X 3 and X 4 is a carbon atom attached by a covalent bond to Y 1 ;
  • Y 1 is
  • the compound of formula (I) may be a compound of formula (II): (II); wherein L 3 is attached by a covalent bond to an atom selected from N a , C a , C b and C c ; wherein if L 3 is attached to N a , R 3 is absent; and wherein the positions on C a , C b and C c to which L 3 is not attached are occupied by R 5 groups; Z is independently selected from N and CR 2 ; R 1 and R 2 are each independently selected from: H, C 1 -C 4 -alkyl, halogen, OR 8 , NR 8 R 9 and C1-C4-haloalkyl; R 3 is absent (if L 3 is joined to N a ) or is independently selected from H and C1-C4-alkyl; R 4 is independently selected from: H, C1-C4-alkyl, F, NR 8 R 9 , OR 8 , C1-C4-haloalkyl and CO
  • linker group L 3 is arranged such that the group Y 1 is attached to the end of linker group L 3 which is defined as (CR 5 R 5 ) s .
  • the atom N a , C a , C b or C c is attached to the end of linker group L 3 which is defined as (CR 5 R 5 )u.
  • C a could be described as a C(R 5 ) x group
  • C b (if present) could be described as a C(R 5 ) y group
  • C c could be described as a C(R 5 )z group
  • x and y are each an integer selected from 1 and 2
  • z is an integer selected from 0 and 1, with the values of x, y and z being selected to satisfy valency requirements.
  • the macrocyclic ring is the ring including the atoms or groups represented by L 1 , L 3 , Y 1 ,and X 4 .
  • the macrocyclic ring may also include one or more of the atoms or groups represented by N a , C a , C b , C c and X 3 .
  • the compound may be the E geometric isomer, the Z geometric isomer or a mixture thereof.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , L 1 , L 2 , L 3 , L 4 , Y 1 , X 1 , X 2 and X 4 are as defined above for formula (II).
  • r may be 2.
  • the compound of formula (I) may be a compound of formula (IV):
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , L 1 , L 2 , L 3 , L 4 , Y 1 , X 1 , X 2 and X 4 are as defined above for formula (II).
  • r may be 2.
  • the compound of formula (I) may be a compound of formula (V):
  • r may be 2.
  • the compound of formula (I) may be a compound of formula (VI):
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , L 1 , L 2 , L 3 , L 4 , Y 1 , X 1 , X 2 , and X 4 are as defined above for formula (II) and wherein R 4 is independently selected from: H, C 1 -C 4 -alkyl, C 1 -C 4 -haloalkyl and CO 2 R 8 ; or R 3 and R 4 together form a–(CR 5 R 5 ) n - group; wherein n is an integer selected from 1, 2 and 3.
  • r may be 2.
  • the compound of formula (I) may be a compound of formula (VII):
  • R 1 , R 2 , R 5 , R 6 , R 7 , L 1 , L 2 , L 3 , L 4 , Y 1 , X 1 , X 2 , X 4 and n are as defined above for formula (II) and wherein L 3 is attached by a covalent bond to an atom selected from C a , Cb and C c .
  • r may be 2.
  • the compound of formula (I) may be a compound of formula (VIII):
  • r may be 3.
  • the compound of formula (I) may be a compound of formula (IX):
  • R 1 , R 2 , R 5 , X 1 , X 2 , X 4 , Y 1 , L 1 , L 3 , L 2 , n and R 7 are as defined for formula (II) above; wherein L 3 is attached by a covalent bond to an atom selected from C d , C e and C f and wherein the positions on C d , C e and C f to which L 3 is not attached are occupied by R 5 groups.
  • the compound of formula (I) may be a compound of formula (X):
  • R 1 , R 2 , R 5 , X 1 , X 2 , X 4 , Y 1 , L 1 , L 3 , n and R 7 are as defined for formula (II) above; wherein X a is carbon or nitrogen; and wherein L 3 is attached by a covalent bond to an atom selected from C g , C h and, where X a is carbon, X a and wherein any positions on C g , C h and X a to which L 3 is not attached are occupied by R 5 groups.
  • the compound of formula (I) may be a compound of formula (XI):
  • L 3 is attached by a covalent bond to an atom selected from C a , C b and C c ; and wherein the positions on C a , C b and C c to which L 3 is not attached are occupied by R 5 groups; R 1 , R 2 , R 5 , R 6 and R 8 are each independently at each occurrence selected from: H and C1- C 4 -alkyl; X 1 , X 2 and X 4 are each independently selected from: N and CR 10 ; wherein no more than two of X 1 , X 2 and X 4 are N; L 1 is a linker group having the form -(CR 5 R 5 ) r -; wherein r is an integer selected from 2 and 3; L 3 is independently –(CR 5 R 5 ) s -Y 3 -(CR 5 R 5 ) t -Y 2 -(CR 5 R 5 ) u -; wherein s and t are each independently an integer selected from 1, 2, 3 and
  • group A may be a 5-, 6- or 7-membered heterocycloalkyl ring comprising at least one nitrogen in the ring.
  • Group A may be a piperidine, piperazine or pyrrolidine ring. It may be that the group L 1 is attached to the nitrogen or a nitrogen in the group A ring, e.g. the nitrogen or a nitrogen in the 5-, 6- or 7-membered heterocycloalkyl ring (e.g. a piperidine, piperazine or pyrrolidine ring).
  • Group A may be a 3-6 heteroalkyl group comprising at least one nitrogen in the linking chain. It may be that the group L 1 is attached to the nitrogen or a nitrogen in the group A ring.
  • L 3 is attached to C a .
  • a single R 5 group is also attached to C a , two R 5 groups are attached to C b and a single R 5 group is attached to C c , i.e. x is 1, y is 2 and z is 0.
  • L 3 is attached to C b .
  • a single R 5 group is also attached to C b , two R 5 groups are attached to C a and a single R 5 group is attached to C c , i.e. x is 2, y is 1 and z is 0.
  • L 3 is attached to C b
  • m is 1. It may be that L 3 is attached to N a .
  • R 5 groups are attached to C a
  • two R 5 groups are attached to C b and a single R 5 group is attached to C c , i.e. x is 2, y is 2 and z is 1.
  • L 3 is attached to C c .
  • two R 5 groups are attached to C a and two R 5 groups are attached to C b , i.e. x is 2, y is 2 and z is 0.
  • L 3 is attached to C a , C b or C c .
  • X 3 is a carbon atom which is attached by a covalent bond to Y 1 .
  • X 1 , X 2 and X 4 are each independently selected from: N and CR 10 ; wherein no more than two of X 1 , X 2 and X 4 are N. It may be that each of X 1 , X 2 and X 4 are CR 10 . It may be that X 1 is N.
  • X 1 is CR 10a , wherein R 10a is independently selected from: H, halo, nitro, cyano, NR 8 R 9 , OR 8 ; O-aryl, SR 8 , SOR 8 , SO3R 8 , SO2R 8 , SO2NR 8 R 8 , CO2R 8 , C(O)R 8 , CONR 8 R 8 , aryl, C 1 -C 4 -alkyl, C 2 -C 4 -alkenyl, C 2 -C 4 -alkynyl and C 1 -C 4 -haloalkyl.
  • R10a may be selected from H, halo, C1-C4-alkyl and C1-C4-haloalkyl.
  • X 1 may be CH. It may be that X 2 is N.
  • X 2 is CR 10b , wherein R 10b is independently selected from: H, halo, nitro, cyano, NR 8 R 9 , OR 8 ; O-aryl, SR 8 , SOR 8 , SO3R 8 , SO2R 8 , SO2NR 8 R 8 , CO2R 8 , C(O)R 8 , CONR 8 R 8 , aryl, C 1 -C 4 -alkyl, C 2 -C 4 -alkenyl, C 2 -C 4 -alkynyl and C 1 -C 4 -haloalkyl.
  • R10b may be selected from H, halo, C1-C4-alkyl and C1-C4-haloalkyl.
  • X 2 may be CH. It may be that X 4 is N.
  • X 4 is CR 10c , wherein R 10c is independently selected from: H, halo, nitro, cyano, NR 8 R 9 , OR 8 ; O-aryl, SR 8 , SOR 8 , SO 3 R 8 , SO 2 R 8 , SO 2 NR 8 R 8 , CO 2 R 8 , C(O)R 8 , CONR 8 R 8 , aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl.
  • R10c may be selected from H, halo, C1-C4-alkyl and C1-C4-haloalkyl.
  • X 4 may be CH. It may be that X 1 , X 2 and X 4 are each CH. It may be that X 1 , X 2 and X 4 are each CH and R 1 and R 2 are each H.
  • R 10 may be independently at each occurrence selected from H, halo, C1-C4-alkyl and C1-C4- haloalkyl. R 10 may at all occurrences be H.
  • Z is CR 2 . It may be that R 1 is H. It may be that R 2 is H, i.e. that Z is CH.
  • R 1 and R 2 are each H, i.e. that R 1 is H and Z is CH.
  • Y 1 may be selected from O and CR 5 R 5 .
  • Y 1 may be selected from O, S and NR 9 .
  • Y 1 is CR 5 R 5 .
  • Y 1 is CH 2 .
  • Y 1 is O.
  • r may be 2.
  • r may be 3.
  • each R 5 group in the linker L 1 is H.
  • L 1 may be–(CH2)2-.
  • L 1 may be–(CH2)3-.
  • m may be 0.
  • m may be 1.
  • L 5 may be absent.
  • L 5 is -L 6 -L 2 -; L 6 may be absent.
  • L 6 is–L 4 NR 6 -; L 4 may be a bond. This will typically be the case where R 3 and R 4 do not together form a– (CR 5 R 5 )n- group.
  • L 4 may be–CR 5 R 5 -. This may be the case where R 3 and R 4 together form a–(CR 5 R 5 )n- group.
  • u 1.
  • u 0.
  • s is 1.
  • t is an integer selected from 1, 2 and 3.
  • s is an integer selected from 2, 3 and 4.
  • t is an integer selected from 2, 3 and 4.
  • Y 2 is selected from O, NR 9 and S. It may be that Y 2 is O. In these embodiments, it may be that u is 1. It may be that R 5 is, at each occurrence in the group- (CR 5 R 5 )u-, H. These embodiments are particularly preferred where L 3 is attached to C a , C b or C c . It may be that the group -(CR 5 R 5 ) u - is–C(O)- and that Y 2 is selected from O and NR 9 . Thus it may be that Y 2 is NR 9 , e.g. NH. In these embodiments, it may be that Y 3 is a bond. Alternatively, it may be that Y 2 is a bond.
  • Y 2 is a bond.
  • L 3 is attached to N a and Y 3 is selected from O, NR 9 and S, it may be that u is 0 and Y 2 is a bond. It may be that Y 3 is a bond. Alternatively, it may be that Y 3 is selected from O, NR 9 and S. It may be that Y 3 is O. It may be that u is 0, Y 2 is a bond and Y 3 is selected from O, NR 9 and S (e.g. is O).
  • L 3 is attached to N a , it may be that u is 0, Y 2 is a bond and Y 3 is selected from O, NR 9 and S (e.g. Y 3 is O).
  • L 3 is attached to C a , C b or C c , it may be that u is 1, Y 2 is selected from O, NR 9 and S (e.g. is O) and Y 3 is a bond. It may be that Y 1 and Y 2 are each independently selected from O, NR 9 and S (e.g. are each O), Y 3 is a bond, u is 1, s is 1 and t is an integer selected from 1, 2 and 3. In these embodiments, it may be that L 3 is attached to C a , C b or C c . It may be that Y 1 and Y 3 are each independently selected from O, NR 9 and S (e.g.
  • Y 2 is a bond
  • u is 0,
  • s and t are each independently an integer selected from 2, 3, and 4.
  • L 3 is attached to N a .
  • Y 2 and Y 3 are each a bond.
  • Y 2 and Y 3 are each a bond and u is 0.
  • s is an integer selected from 1 and 2
  • t is an integer selected from 1, 2, 3 and 4.
  • Y 1 is O.
  • Y 1 is CR 5 R 5 .
  • Y 3 is 1,2,3-triazole.
  • the triazole may have a structure selected from:
  • Y 3 has the structure Where Y 3 is 1,2,3- triazole, it may be that s and t are each 1. Where Y 3 is 1,2,3-triazole, it may be that Y 2 is a bond. Where Y 3 is 1,2,3-triazole, it may be that Y 2 is a–O-. Where Y 3 is 1,2,3-triazole, it may be that s, t and u are each 1 and Y 2 is -O-.
  • R3 is independently selected from H and C1-C4-alkyl.
  • R 3 may be H.
  • R 3 may be C 1 -C 4 -alkyl, e.g. methyl.
  • L 4 is a bond
  • m is 0.
  • R 3 and R 4 together form a–(CR 5 R 5 )n- group; wherein n is an integer selected from 1, 2 and 3. It may be that n is 2. It may be that each R 5 group is the group formed by R 3 and R 4 is H. Thus, it may be that R 3 and R 4 together form a–CH2CH2- group. In these embodiments, it may be that m is 1. In these embodiments, it may be that L 4 is–CR 5 R 5 -. L 2 is preferably–CR 5 R 5 - and most preferably is -CH 2 -. Alternatively, L 2 may be a 3-, 4- or 5- membered cycloalkyl ring, e.g. a 4-membered cycloalkyl ring.
  • R 7 may be a monocyclic aryl group.
  • R 7 may be a phenyl group.
  • Said phenyl group may be unsubstituted or it may be substituted with from 1 to 5 substituents which are each independently at each occurrence selected from the group consisting of: halo, nitro, cyano, NR a R a , NR a S(O)2R a , NR a CONR a R a , NR a CO2R a , NR a C(O)R a , OR a ; SR a , SOR a , SO3R a , SO 2 R a , SO 2 NR a R a , CO 2 R a C(O)R a , CONR a R a , C 1 -C 4 -alkyl, C 2 -C 4 -alkenyl, C 2 -C 4 -alkynyl, C 1 -
  • R 7 may be a phenyl group which is substituted by 1 to 3 substituents independently at each occurrence selected from F, nitro, C 1 -C 4 -alkyl, C 2 -C 4 - alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl.
  • exemplary R 7 groups include 2,5-difluorophen-1-yl and 3-nitro-4-methylphen-1-yl.
  • R 7 is a bicyclic carbocyclic or heterocyclic ring system in which at least one of the two rings is aromatic or heteroaromatic.
  • R 7 may take the form:
  • V 1 , V 2 and V 3 are each independently selected from: N and CR 11 ; with the proviso that no more than two of V 1 , V 2 and V 3 are N; and wherein the ring B is a substituted or unsubstituted 5- or 6- membered saturated cycloalkyl or heterocycloalkyl ring.
  • R 11 is independently at each occurrence selected from: H, halo, nitro, cyano, NR a R a , NR a S(O)2R a , NR a C(O)R a , NR a CONR a R a , NR a CO 2 R a , OR a ; SR a , SOR a , SO 3 R a , SO 2 R a , SO 2 NR a R a , CO 2 Ra C(O)R a , CONR a R a , CR b R b NR a R a , C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4- haloalkyl.
  • R 7 takes the form:
  • V 4 and V 5 are each independently selected from O, S, CR 12 R 12 and NR 8 ;
  • p is an integer selected from 1 and 2.
  • R 7 takes the form:
  • V 1 , V 2 and V 3 are each independently selected from: N and CH; with the proviso that no more than two of V 1 , V 2 and V 3 are N.
  • a single one of V 1 , V 2 and V 3 is N.
  • V 3 is CR 11 (e.g. CH).
  • V 1 is N and V 2 is CR 11 (e.g. CH).
  • V 2 is N and V 1 is CR 11 (e.g. CH).
  • V 2 is nitrogen.
  • V 3 is CR 11 .
  • V 1 is CR 11 .
  • R 11 is selected from H, methyl and halogen, e.g. F.
  • R 11 is selected from H and halogen, e.g. F. It may be that V 3 is CH. It may be that V 1 is CR 11 , wherein R 11 is selected from H and halogen, e.g. F. It may be that V 2 is nitrogen, V 3 is CH and V 1 is CR 11 , wherein R 11 is selected from H and halogen, e.g. F.
  • V 4 is O. Thus, it may be that both V 4 and V 5 are O.
  • Exemplary R 7 groups include:
  • R 7 may also take the form
  • V 6 is independently selected from N and CR 13 (e.g. CH); V 7 is independently selected from NR 8 , S and O; and R 13 is independently at each occurrence selected from: halo, nitro, cyano, NR a R a , NR a S(O)2R a , NR a C(O)R a , NR a CONR a R a , NR a CO 2 R a , NR a C(O)R a , OR a ; SR a , SOR a , SO 3 R a , SO 2 R a , SO 2 NR a R a , CO 2 Ra C(O)R a , CONR a R a , C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl, C1-C4-haloalkyl, and CR a R a NR a R a
  • R 13 may be independently at each occurrence selected from F, CN, OR a , nitro, C 1 -C 4 -alkyl, C 2 -C 4 -alkenyl, C 2 -C 4 -alkynyl and C 1 -C 4 -haloalkyl.
  • R 6 is selected from H or C1-C4-alkyl. Even more preferably, R 6 is H. It may be that R 8 is independently at each occurrence selected from H or C1-C4-alkyl. It may be that R 8 is at each occurrence H. It may be that R 9 is independently at each occurrence selected from H, C1-C4-alkyl, C1-C4- haloalkyl, S(O) 2 R 8 and C(O)R 8 . It may be that R 9 is independently at each occurrence selected from H, C1-C4 alkyl (e.g. methyl) and C(O)R 8 (e.g. acetate).
  • C1-C4 alkyl e.g. methyl
  • C(O)R 8 e.g. acetate
  • R 9 is at each occurrence H. It may be that L 1 , group A, r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 13 atoms. It may be that L 1 , group A, r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 14 atoms. It may be that L 1 , group A, r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 15 atoms.
  • L 1 , group A, r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 16 atoms. It may be that L 1 , group A, r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 17 atoms. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 13 atoms.
  • r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 13 atoms and R 3 and R 4 together form a–(CR 5 R 5 )n- group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 13 atoms and R 3 and R 4 do not together form a–(CR 5 R 5 ) n - group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 14 atoms.
  • r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 14 atoms and R 3 and R 4 together form a–(CR 5 R 5 ) n - group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 14 atoms and R 3 and R 4 do not together form a–(CR 5 R 5 ) n - group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 15 atoms.
  • r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 15 atoms and R 3 and R 4 together form a–(CR 5 R 5 )n- group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 15 atoms and R 3 and R 4 do not together form a–(CR 5 R 5 )n- group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 16 atoms.
  • r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 16 atoms and R 3 and R 4 together form a–(CR 5 R 5 )n- group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 16 atoms and R 3 and R 4 do not together form a–(CR 5 R 5 )n- group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 17 atoms.
  • r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 17 atoms and R 3 and R 4 together form a–(CR 5 R 5 ) n - group. It may be that r, s, t, u, Y 2 and Y 3 are selected such that the macrocyclic ring has a ring size of 17 atoms and R 3 and R 4 do not together form a–(CR 5 R 5 ) n - group.
  • the compound of formula (I) may have a structure selected from:
  • the compound of formula (I) is selected from compounds 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 and 11 in the Examples below.
  • the compound of the invention is an N-oxide
  • it will typically be a pyridine N-oxide, i.e. where the compound of the invention comprises a pyridine ring (which may form part of a bicyclic or tricyclic ring system), the nitrogen of that pyridine may be N + -O-.
  • the compound of the invention is not an N-oxide. Included within the scope of the present invention are all stereoisomers, geometric isomers and tautomeric forms of the compounds of the invention, including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof.
  • acid addition or base salts wherein the counter ion is optically active for example, d-lactate or l-lysine, or racemic, for example, dl-tartrate or dl-arginine.
  • the oxime groups present in certain compounds of the invention may be present as the E-oxime, as the Z- oxime or as a mixture of both in any proportion.
  • Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation. Where structurally isomeric forms of a compound are interconvertible via a low energy barrier, tautomeric isomerism (‘tautomerism’) can occur. This can take the form of proton tautomerism in compounds of the invention containing, for example, an imino, keto, or oxime group, or so- called valence tautomerism in compounds which contain an aromatic moiety.
  • tautomerism tautomeric isomerism
  • racemate or the racemate of a salt or derivative
  • HPLC high pressure liquid chromatography
  • the racemate or a racemic precursor
  • a suitable optically active compound for example, an alcohol, or, in the case where the compound of the invention contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid.
  • the resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted into the corresponding pure enantiomer(s) by means well known to a skilled person.
  • Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1% diethylamine.
  • Racemic mixtures may be separated by conventional techniques known to those skilled in the art - see, for example,“Stereochemistry of Organic Compounds” by E. L.
  • Cm-Cn refers to a group with m to n carbon atoms.
  • alkyl refers to a linear or branched hydrocarbon chain.
  • C 1- C 6 -alkyl may refer to methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, n-pentyl and n- hexyl.
  • the alkyl groups may be unsubstituted or substituted by one or more substituents.
  • haloalkyl refers to a hydrocarbon chain substituted with at least one halogen atom independently chosen at each occurrence from: fluorine, chlorine, bromine and iodine.
  • the halogen atom may be present at any position on the hydrocarbon chain.
  • C1-C6-haloalkyl may refer to chloromethyl, fluoromethyl, trifluoromethyl, chloroethyl e.g.1-chloroethyl and 2-chloroethyl, trichloroethyl e.g.1,2,2-trichloroethyl, 2,2,2- trichloroethyl, fluoroethyl e.g. 1-fluoroethyl and 2-fluoroethyl, trifluoroethyl e.g.
  • haloalkyl group may be a fluoroalkyl group, i.e. a hydrocarbon chain substituted with at least one fluorine atom.
  • alkenyl refers to a branched or linear hydrocarbon chain containing at least one double bond.
  • the double bond(s) may be present as the E or Z isomer (e.g. cis or trans).
  • the double bond may be at any chemically possible position of the hydrocarbon chain.
  • “C 2- C 6 -alkenyl” may refer to ethenyl, propenyl, butenyl, butadienyl, pentenyl, pentadienyl, hexenyl and hexadienyl.
  • the alkenyl groups may be unsubstituted or substituted by one or more substituents. Specific substituents for any saturated carbon in each alkenyl group independently may be fluorine, OR a or NR a R a .
  • alkynyl refers to a branched or linear hydrocarbon chain containing at least one triple bond.
  • the triple bond may be at any possible position of the hydrocarbon chain.
  • “C 2- C 6 -alkynyl” may refer to ethynyl, propynyl, butynyl, pentynyl and hexynyl.
  • the alkynyl groups may be unsubstituted or substituted by one or more substituents. Specific substituents for any saturated carbon in each alkynyl group independently may be fluorine, OR a or NR a R a .
  • carbocylic refers to a group consisting of one or more rings which are entirely formed from carbon atoms.
  • a carbocylic group can be a mono- or bicyclic cycloalkyl group, or it can comprise at least one phenyl ring.
  • heterocyclic refers to a group consisting of one or more rings wherein the ring system includes at least one heteroatom.
  • a heterocyclic group may comprise either a heteroaryl or heterocycloalkyl rings.
  • cycloalkyl refers to a saturated hydrocarbon ring system containing 3, 4, 5 or 6 carbon atoms.
  • C 3 -C 6 -cycloalkyl may refer to cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • the cycloalkyl groups may be unsubstituted or substituted by one or more substituents.
  • Specific substituents for each cycloalkyl group independently may be oxo, C1-C4-alkyl, fluorine, OR a or NHR a .
  • aromatic when applied to a substituent as a whole means a single ring or polycyclic ring system with 4n + 2 electrons in a conjugated ⁇ system within the ring or ring system where all atoms contributing to the conjugated ⁇ system are in the same plane.
  • aryl refers to an aromatic hydrocarbon ring system. The ring system has 4n +2 electrons in a conjugated ⁇ system within a ring where all atoms contributing to the conjugated ⁇ system are in the same plane.
  • the“aryl” may be phenyl and naphthyl. Equally, aryl groups may include non-aromatic carbocyclic portions.
  • the aryl group may be unsubstituted or substituted by one or more substituents.
  • Specific substituents for each aryl group independently may be C1-C4-alkyl, C1-C4-haloalkyl, cyano, halogen, OR a or NHR a .
  • heteroaryl may refer to any aromatic (i.e. a ring system containing (4n + 2) ⁇ - or n- electrons in the ⁇ -system) 5-10 membered ring system comprising from 1 to 4 heteroatoms independently selected from O, S and N (in other words from 1 to 4 of the atoms forming the ring system are selected from O, S and N).
  • any heteroaryl groups may be independently selected from: 5 membered heteroaryl groups in which the heteroaromatic ring is substituted with 1-4 heteroatoms independently selected from O, S and N; and 6-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1-3 (e.g.1-2) nitrogen atoms; 9-membered bicyclic heteroaryl groups in which the heteroaromatic system is substituted with 1-4 heteroatoms independently selected from O, S and N; 10-membered bicyclic heteroaryl groups in which the heteroaromatic system is substituted with 1-4 nitrogen atoms.
  • heteroaryl groups may be independently selected from: pyrrole, furan, thiophene, pyrazole, imidazole, oxazole, isoxazole, triazole, oxadiazole, thiadiazole, tetrazole; pyridine, pyridazine, pyrimidine, pyrazine, triazine, indole, isoindole, benzofuran, isobenzofuran, benzothiophene, indazole, benzimidazole, benzoxazole, benzthiazole, benzisoxazole, purine, quinoline, isoquinoline, cinnoline, quinazoline, quinoxaline, pteridine, phthalazine, naphthyridine.
  • Heteroaryl groups may also be 6-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1 heteroatomic group independently selected from O, S and NH and the ring also comprises a carbonyl group. Such groups include pyridones and pyranones.
  • the heteroaryl system itself may be substituted with other groups.
  • the heteroaryl group may be unsubstituted or substituted by one or more substituents. Specific substituents for each heteroaryl group independently may be C 1 -C 4 -alkyl, C 1 -C 4 -haloalkyl, cyano, halogen, OR a or NHR a .
  • m-nheterocycloalkyl may refer to a m- to n-membered monocyclic or bicyclic saturated or partially saturated group comprising 1 or 2 heteroatoms independently selected from O, S and N in the ring system (in other words 1 or 2 of the atoms forming the ring system are selected from O, S and N).
  • partially saturated it is meant that the ring may comprise one or two double bonds. This applies particularly to monocyclic rings with from 5 to 8 members. The double bond will typically be between two carbon atoms but may be between a carbon atom and a nitrogen atom.
  • heterocycloalkyl groups include; piperidine, piperazine, morpholine, thiomorpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, dihydrofuran, tetrahydropyran, dihydropyran, dioxane, azepine.
  • Bicyclic systems may be spiro-fused, i.e. where the rings are linked to each other through a single carbon atom; vicinally fused, i.e. where the rings are linked to each other through two adjacent carbon or nitrogen atoms; or they may be share a bridgehead, i.e. the rings are linked to each other through two non-adjacent carbon or nitrogen atoms.
  • heterocycloalkyl groups may be unsubstituted or substituted by one or more substituents.
  • Specific substituents for each heterocycloalkyl group may independently be oxo, C 1 -C 4 - alkyl, fluorine, OR a or NHR a .
  • 3-6 heteroalkyl refers to a 3- to 6- membered chain of atoms, wherein at least one of said atoms is a heteroatom, e.g. a nitrogen, and wherein the remainder of said atoms are carbon atoms.
  • the compound of formula (I) is an N-oxide
  • it will typically be a pyridine N-oxide, i.e. the nitrogen of the pyridine may be N + -O-.
  • the compound of the invention is not an N-oxide.
  • An aromatic ring is a phenyl ring.
  • the present invention also includes the synthesis of all pharmaceutically acceptable isotopically-labelled compounds of formulae (I) to (XI) wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number most commonly found in nature.
  • isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H and 3 H, carbon, such as 11 C, 13 C and 14 C, chlorine, such as 36 Cl, fluorine, such as 18 F, iodine, such as 123 I and 125 I, nitrogen, such as 13 N and 15 N, oxygen, such as 15 O, 17 O and 18 O, phosphorus, such as 32 P, and sulphur, such as 35 S.
  • Certain isotopically-labelled compounds for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e.
  • isotopically-labelled compounds can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described using an appropriate isotopically-labelled reagent in place of the non-labelled reagent previously employed.
  • each of the compounds of the present invention may be used as a medicament.
  • compound as defined above for the treatment of bacterial infections The compounds and formulations of the present invention may be used in the treatment of a wide range of bacterial infections.
  • the compounds can be used to treat bacterial infections caused by one or more resistant strains of bacteria.
  • the compounds can be used to treat bacterial infections caused by one or more resistant strains of Gram positive bacteria.
  • the compounds can be used to treat bacterial infections caused by one or more resistant strains of Gram negative bacteria.
  • the compounds and formulations of the invention may be used to treat infections caused by bacteria which are in the form of a biofilm.
  • resistant strains is intended to mean strains of bacteria which have shown resistance to one or more known antibacterial drug. For example, it may refer to strains which are resistant to methicillin, strains that are resistant to one or more other ⁇ -lactam antibiotics, strains that are resistant to one or more fluoroquinolones and/or strains that are resistant to one or more other antibiotics (i.e. antibiotics other than ⁇ -lactams and fluoroquinolones).
  • a resistant strain is one in which the MIC of a given compound or class of compounds for that strain has shifted to a significantly higher number than for the parent (susceptible) strain.
  • the compounds of the invention may be particularly effective at treating infections caused by Gram positive bacteria.
  • the compounds of the invention may be particularly effective at treating infections caused by Gram positive bacteria which are resistant to one or more fluoroquinolone antibiotics.
  • the compounds of the invention may be particularly effective at treating infections caused by Gram negative bacteria.
  • the compounds of the invention may be particularly effective at treating infections caused by Gram negative bacteria which are resistant to one or more fluoroquinolone antibiotics.
  • the compounds and formulations of the present invention can be used to treat or to prevent infections caused by bacterial strains associated with biowarfare. These may be strains which are category A pathogens as identified by the US government (e.g. those which cause anthrax, plague etc.) and/or they may be strains which are category B pathogens as identified by the US government (e.g. those which cause Glanders disease, mellioidosis etc).
  • the compounds and formulations of the present invention can be used to treat or to prevent infections caused by Gram positive bacterial strains associated with biowarfare (e.g. anthrax). More particularly, the compounds and formulations may be used to treat category A and/or category B pathogens as defined by the US government on 1 st Jan 2015.
  • the compounds of the invention may also be useful in treating other forms of infectious disease, e.g. fungal infections, parasitic infections and/or viral infections.
  • the compounds of the present invention can be used in the treatment of the human body. They may be used in the treatment of the animal body. In particular, the compounds of the present invention can be used to treat commercial animals such as livestock. Alternatively, the compounds of the present invention can be used to treat companion animals such as cats, dogs, etc.
  • Suitable pharmaceutically acceptable salts include, but are not limited to, salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, malic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, toluenesulphonic, benzenesulphonic, salicylic, sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.
  • pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, n
  • Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts. Hemisalts of acids and bases may also be formed, for example, hemisulfate and hemicalcium salts. Also included are acid addition or base salts wherein the counter ion is optically active, for example, d-lactate or l-lysine, or racemic, for example, dl-tartrate or dl-arginine.
  • Compounds of the invention may exist in a single crystal form or in a mixture of crystal forms or they may be amorphous.
  • compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, or spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
  • the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. For example, if the compound of the invention is administered orally, then the daily dosage of the compound of the invention may be in the range from 0.01 micrograms per kilogram body weight ( ⁇ g/kg) to 100 milligrams per kilogram body weight (mg/kg).
  • a compound of the invention, or pharmaceutically acceptable salt thereof may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the compounds of the invention, or pharmaceutically acceptable salt thereof, is in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • a pharmaceutically acceptable adjuvant diluent or carrier.
  • Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, "Pharmaceuticals - The Science of Dosage Form Designs", M. E. Aulton, Churchill Livingstone, 1988.
  • the compounds of the invention may be administered in combination with other active compounds (e.g. antifungal compounds, oncology compounds) and, in particular, with other antibacterial compounds.
  • active compounds e.g. antifungal compounds, oncology compounds
  • the compound of the invention and the other active e.g. the other antibacterial compound
  • the compound of the invention and the other active e.g. the other antibacterial compound
  • the pharmaceutical composition which is used to administer the compounds of the invention will preferably comprise from 0.05 to 99 %w (per cent by weight) compounds of the invention, more preferably from 0.05 to 80 %w compounds of the invention, still more preferably from 0.10 to 70 %w compounds of the invention, and even more preferably from 0.10 to 50 %w compounds of the invention, all percentages by weight being based on total composition.
  • the pharmaceutical compositions may be administered topically (e.g.
  • the skin in the form, e.g., of creams, gels, lotions, solutions, suspensions, or systemically, e.g. by oral administration in the form of tablets, capsules, syrups, powders, suspensions, solutions or granules; or by parenteral administration in the form of a sterile solution, suspension or emulsion for injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion); or by rectal administration in the form of suppositories; or by inhalation (i.e. in the form of an aerosol or by nebulisation).
  • oral administration in the form of tablets, capsules, syrups, powders, suspensions, solutions or granules
  • parenteral administration in the form of a sterile solution, suspension or emulsion for injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion); or by rectal administration in the form of suppositories; or by inhalation (i.e. in the form of an
  • a compound with an in vitro MIC of, for example, 16-64 ⁇ g/mL may still provide an effective treatment against certain bacterial infections.
  • the compounds of the invention may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets.
  • an adjuvant or a carrier for example, lactose, saccharose, sorbitol, mannitol
  • a starch for example, potato starch, corn starch or amylopectin
  • a cellulose derivative for example, gelatine or polyvinylpyrrolidone
  • a lubricant for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax
  • the cores may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide.
  • a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide.
  • the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent.
  • the compounds of the invention may be admixed with, for example, a vegetable oil or polyethylene glycol.
  • Hard gelatine capsules may contain granules of the compound using either the above-mentioned excipients for tablets.
  • liquid or semisolid formulations of the compound of the invention may be filled into hard gelatine capsules.
  • Liquid preparations for oral application may be in the form of syrups or suspensions, for example, solutions containing the compound of the invention, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol.
  • such liquid preparations may contain colouring agents, flavouring agents, sweetening agents (such as saccharine), preservative agents and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in the art.
  • the compounds of the invention may be administered as a sterile aqueous or oily solution.
  • the size of the dose for therapeutic purposes of compounds of the invention will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well known principles of medicine. Dosage levels, dose frequency, and treatment durations of compounds of the invention are expected to differ depending on the formulation and clinical indication, age, and co-morbid medical conditions of the patient.
  • the standard duration of treatment with compounds of the invention is expected to vary between one and seven days for most clinical indications. It may be necessary to extend the duration of treatment beyond seven days in instances of recurrent infections or infections associated with tissues or implanted materials to which there is poor blood supply including bones/joints, respiratory tract, endocardium, and dental tissues.
  • the present invention provides a pharmaceutical formulation comprising a compound of the invention and a pharmaceutically acceptable excipient.
  • the formulation may further comprise one or more other antibiotics, e.g. one or more fluoroquinolone antibiotics.
  • fluoroquinolone antibiotics include levofloxacin, enoxacin, fleroxacin, lomefloxacin, nadifloxacin, norfloxacin, rufloxacin, balofloxacin, grepafloxacin, pazufloxacin, sparfloxacin, temafloxacin, tosufloxacin, besifloxacin, clinafloxacin, garenoxacin, gemifloxacin, gatifloxacin, sitafloxacin, trovafloxacin, prulifloxacin, ciprofloxacin, pefloxacin, moxifloxacin, ofloxacin, delafloxacin, zabofloxacin
  • a method of treating a bacterial infection comprising treating a subject in need thereof with a therapeutically effective amount of a compound of the invention.
  • Medical uses The compounds of the present invention can be used in the treatment of the human body.
  • the compounds of the invention may be for use in treating human bacterial infections such as infections of the genitourinary system, the respiratory tract, the gastrointestinal tract, the ear, the skin, the throat, soft tissue, bone and joints (including infections caused by Staphylococcus aureus).
  • the compounds can be used to treat pneumonia, sinusitis, acute bacterial sinusitis, bronchitis, acute bacterial exacerbation of chronic bronchitis, anthrax, chronic bacterial prostatitis, acute pyelonephritis, pharyngitis, mycobacterial infections (e.g. tuberculosis or leprosy), tonsillitis, Escherichia coli, prophylaxis before dental surgery, cellulitis, acnes, cystitis, infectious diarrhoea, typhoid fever, infections caused by anaerobic bacteria, peritonitis, abdominal infection, bacteraemia, septicaemia, leprosy, sexually transmitted bacterial infection (e.g.
  • gonorrhoea Chlamydia
  • Neisseria infection e.g. gonorrhoea, meningitis
  • other fastidious Gram negative infection e.g. gonorrhoea, meningitis
  • bacterial vaginosis pelvic inflammatory disease
  • pseudomembranous colitis Helicobacter pylori
  • acute gingivitis Crohn's disease
  • rosacea fungating tumours, impetigo.
  • the compounds of the present invention may also be used in treating other conditions treatable by eliminating or reducing a bacterial infection. In this case they will act in a secondary manner alongside for example a chemotherapeutic agent used in the treatment of cancer.
  • a compound for use in the preparation of a medicament is provided.
  • the medicament may be for use in the treatment of any of the diseases, infections and indications mentioned in this specification.
  • a compound of the invention for medical use The compound may be used in the treatment of any of the diseases, infections and indications mentioned in this specification.
  • Veterinary uses They may be used in the treatment of the animal body.
  • the compounds of the present invention can be used to treat commercial animals such as livestock.
  • the livestock may be mammal (excluding humans) e.g. cows, pigs, goats, sheep, llamas, alpacas, camels and rabbits.
  • the livestock may be birds (e.g. chickens, turkeys, ducks, geese etc.).
  • the compounds of the present invention can be used to treat companion animals such as cats, dogs, etc.
  • the veterinary use may be to treat wild populations of animals in order to prevent the spread of disease to humans or to commercial animals.
  • the animals may be rats, badgers, deer, foxes, wolves, mice, kangaroos and monkeys and other apes.
  • a compound of the invention for veterinary use The compound may be used in the treatment of any of the animal diseases and infections and indications mentioned in this specification.
  • the present invention provides a veterinary formulation comprising a compound of the invention and a veterinarily acceptable excipient.
  • the methods by which the compounds may be administered for veterinary use include oral administration by capsule, bolus, tablet or drench, topical administration as an ointment, a pour-on, spot-on, dip, spray, mousse, shampoo, collar or powder formulation or, alternatively, they can be administered by injection (e.g. subcutaneously, intramuscularly or intravenously), or as an implant.
  • Such formulations may be prepared in a conventional manner in accordance with standard veterinary practice.
  • the formulations will vary with regard to the weight of active compound contained therein, depending on the species of animal to be treated, the severity and type of infection and the body weight of the animal.
  • typical dose ranges of the active ingredient are 0.01 to 100 mg per kg of body weight of the animal.
  • the range is 0.1 to 10 mg per kg.
  • the veterinary practitioner, or the skilled person will be able to determine the actual dosage which will be most suitable for an individual patient, which may vary with the species, age, weight and response of the particular patient.
  • the above dosages are exemplary of the average case; there can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • the compounds when treating animals the compounds may be administered with the animal feedstuff and for this purpose a concentrated feed additive or premix may be prepared for mixing with the normal animal feed. Certain compounds of the invention may be used in the treatment of mastitis.
  • a particularly preferred method of administration is by injection into the udder of a subject (e.g. a cow, a goat, a pig or sheep).
  • a subject e.g. a cow, a goat, a pig or sheep.
  • the words“comprise” and “contain” and variations of the words, for example“comprising” and“comprises”, means “including but not limited to”, and is not intended to (and does not) exclude other moieties, additives, components, integers or steps.
  • the singular encompasses the plural unless the context otherwise requires.
  • the indefinite article the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise.
  • Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith.
  • Sensitive functional groups may need to be protected and deprotected during synthesis of a compound of the invention. This may be achieved by conventional methods, for example as described in“Protective Groups in Organic Synthesis” by TW Greene and PGM Wuts, John Wiley & Sons Inc (1999), and references therein. Throughout this specification these abbreviations have the following meanings:
  • DMSO dimethyl sulfoxide
  • HATU 1-[Bis(dimethylamino)methylene]-1H- 1,2,3-triazolo[4,5-b]pyridinium-3-oxide hexafluorophosphate
  • HBTU N,N,N′,N′-Tetramethyl-O-(1H-benzotriazol-1-yl)uronium hexafluorophosphate
  • NMP N-methylpyrrolidinone
  • TFA trifluoroacetic acid
  • THF tetrahydrofuran
  • TMSCl trimethylsilyl chloride
  • TMSI trimethylsilyl iodide
  • TBTU N,N,N′,N′-Tetramethyl-O-(1H- benzotriazol-1-yl)uronium tetrafluoroborate
  • aldehyde (3) Reaction of pyridone (1) with commercially available 2-bromo-1,1-diethoxyethane (2), followed by hydrolysis of the acetal can generate aldehyde (3).
  • the alkylation reaction can be carried out in the presence of a base, such as Cs2CO3, in a solvent, such as dry NMP, at a temperature from 50-100 o C.
  • Hydrolysis can be effected using a strong acid, such as concentrated HCl, in a solvent, such as ACN, at room temperature.
  • Aldehyde (3) can be converted to ester (5) via reductive amination with the known amine (4), prepared as described in EP154142.
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • the ester group in (5) can be converted to the alcohol (6) using a reducing agent, such as LiAlH 4 or BHEt 3 + Li- (super hydride), in a solvent, such as THF, at a temperature from 0 o C to room temperature.
  • Allylation of alcohol (6) with allyl bromide in the presence of a base, such as NaH, in a solvent, such as THF or DMF, at a temperature from 0 o C to 60 o C, can furnish diene (7).
  • Addition of quaternary ammonium salts is optional.
  • RCM (ring controlled metathesis) of the diene (7) to macrocycle alkene (8) can be accomplished in the presence of Grubbs catalyst (first or second generation) in a suitable solvent, such as toluene, at a temperature from room temperature to 90 o C.
  • the RCM reaction may provide entirely the E isomer, entirely the Z isomer or a mixture thereof.
  • Deprotection of the cyclic ketal in (8) to release the ketone in (9) can be effected using acid catalysed conditions, such as use of a protic acid (10% HCl in a mixture of THF/H 2 O) or a Lewis acid (FeCl3 dispersed on silica).
  • Ketone (9) can be converted to amine (11) via reductive amination with amine (10).
  • the reaction can be performed using a borohydride reagent, such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80 o C. Addition of AcOH as a catalyst is optional.
  • Amine (11) can be converted to (13) (a subset of compounds of both formula (IV) and formula (VII)) under reductive amination conditions with aldehyde (12).
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • Amine (11) can be converted to (15) (another subset of compounds of both formula (IV) and formula (VII)) by coupling with (14), where LG represents a leaving group, such as a halide.
  • the reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating.
  • Scheme B The alkene double bond in ketone (9) in Scheme A can be hydrogenated to (16) by the action of H2 in the presence of a Pd/C catalyst in an alcoholic solvent, such as EtOH, at room temperature.
  • Ketone (16) can then be transformed into (17) and (18) (two further subsets of compounds of formulae (IV) and (VII)) using an analogous series of transformations to those described in Scheme A for the conversion of ketone (9) to (13) and (15).
  • Certain compounds of formula (III) and (VII) can be made following Scheme A but using the ketal (19), prepared as described in US 6645980, in place of the amine (4).
  • Macrocycles (20) and (21) (two subsets of compounds of formulae (III) and (VII)) can be prepared using an analogous series of transformations to those described in Scheme A for the conversion of ester (5) to (13) and (15).
  • Ketal (24) can be formed by Scheme C from commercially available ketone (27) under Dean Stark conditions using ethylene glycol and catalytic p-TsOH (10%) in toluene.
  • Reductive ammination of aldehyde (3) (prepared as described in Scheme A) with commercially available hydroxy amine (30) can form alcohol (31).
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional. Allylation of alcohol (31) with allyl bromide in the presence of a base, such as NaH, in a solvent, such as THF or DMF, at a temperature from 0 o C to 60 o C, can furnish diene (32).
  • a base such as NaH
  • RCM ring closing metathesis of the diene (32) to macrocycle alkene (33)
  • RCM reaction may provide entirely the E isomer, entirely the Z isomer or a mixture thereof.
  • Deprotection of the nitrogen BOC protecting group in (33) can be performed under standard conditions, such as by the action of TFA in DCM at room temperature.
  • Amine (34) can be converted to (35) (a subset of compounds of both formula (VI) and formula (VII)) by reductive amination with aldehyde (12).
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • Amine (34) can be converted to (36) (another subset of compounds of both formula (VI) and formula (VII)) by coupling with (14), where LG represents a leaving group, such as a halide.
  • the reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating.
  • Amine (42) can be formed through reaction of commercially available bromo ester (40) with commercially available cyclic acetal protected amine (41).
  • the reaction can be performed in the presence of a base, such as K2CO3 or Et3N, in a solvent, such as ACN or DCM, at a temperature from 0 o C to room temperature.
  • Aldehyde (3) (prepared as described in Scheme A) can be converted to cyclic acetal (43) via reductive amination with the amine (42).
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • the ester group in cyclic acetal (43) can be converted to the alcohol (44) using a reducing agent, such as LiAlH4 or BHEt3 + Li- (super hydride), in a solvent, such as THF, at a temperature from 0 o C to room temperature.
  • Allylation of alcohol (44) with allyl bromide in the presence of a base, such as NaH, in a solvent, such as THF or DMF, at a temperature from 0 o C to 60 o C, can furnish diene (45).
  • a base such as NaH
  • a solvent such as THF or DMF
  • Addition of quaternary ammonium salts is optional.
  • RCM (ring controlled metathesis) of the diene (45) to macrocycle alkene (46) can be accomplished in the presence of Grubbs catalyst (first or second generation) in a suitable solvent, such as toluene, at a temperature from room temperature to 90 o C.
  • the RCM reaction may provide entirely the E isomer, entirely the Z isomer or a mixture thereof.
  • Deprotection of the cyclic acetal in (46) to release the aldehyde in (47) can be effected using acid catalysed conditions, such as use of a protic acid (10% HCl in a mixture of THF/H 2 O) or a Lewis acid (FeCl 3 dispersed on silica).
  • Aldehyde (47) can be converted to amine (48) via reductive amination with amine (10).
  • the reaction can be performed using a borohydride reagent, such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of AcOH as a catalyst is optional.
  • Amine (48) can be converted to (49) (a subset of compounds of formula (V)) under reductive amination conditions with aldehyde (12).
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • Amine (48) can be converted to (50) (another subset of compounds of formula (V)) by coupling with (14), where LG represents a leaving group, such as a halide.
  • the reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K 2 CO 3 , with optional heating.
  • Certain other compounds of formula (V) can be made by Scheme G:
  • Scheme G The alkene double bond in amine (48) in Scheme G can be hydrogenated to (51) by the action of H 2 in the presence of a Pd/C catalyst in an alcoholic solvent, such as EtOH, at room temperature.
  • Amine (51) can then be transformed into (52) and (53) (two further subsets of compounds of formula (V) using an analogous series of transformations to those described in Scheme F for the conversion of amine (48) to (49) and (50).
  • macrocycles (55) and (56) (a subset of compounds of formula (V)) can be prepared using an analogous series of transformation to those described in Scheme F for the conversion of aldehyde (3) to (49) and (50).
  • Aldehyde (54) can be formed by Scheme H from reaction of (1) with commercially available 3-bromo-1,1-dimethoxypropane (57), followed by hydrolysis of the acetal.
  • the alkylation reaction can be carried out in the presence of a base, such as Cs2CO3, in a solvent, such as dry NMP, at a temperature from 50-100 o C.
  • Hydrolysis of the acetal can be effected using a strong acid, such as concentrated HCl, in a solvent, such as ACN, at room temperature.
  • Reductive amination of aldehyde (3) (prepared as described in Scheme A) with amine (60) can form diene (61).
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • RCM (ring closing metathesis) of the diene (61) to macrocycle alkene (62) can be accomplished in the presence of Grubbs catalyst (first or second generation) in a suitable solvent, such as toluene, at a temperature from room temperature to 90 o C.
  • the RCM reaction may provide entirely the E isomer, entirely the Z isomer or a mixture thereof.
  • Deprotection of the nitrogen CBZ protecting group in (62) can be performed using base, such as KOH, in a solvent mixture of H 2 O and EtOH, at a temperature from 60 o C to reflux or refluxing in neat TFA or treatment with TMSI, in a solvent, such as DCM, at a temperature from room temperature to reflux.
  • Amine (63) can be converted to (64) (a subset of compounds of both formula (III) and formula (VII)) by reduction amination with aldehyde (12).
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • Amine (63) can be converted to (65) (another subset of compounds of both formula (III) and formula (VII)) by coupling with (14), where LG represents a leaving group, such as a halide.
  • the reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating.
  • Amine (60) can be formed by Scheme J:
  • Scheme K Hydrogenation of the alkene double bond and removal of nitrogen CBZ protecting in (62) in Scheme I to give amine (68) can be effected by the action of H 2 in the presence of a Pd/C catalyst in an alcoholic solvent, such as EtOH, at room temperature. Amine (68) can then be transformed into (69) and (70) (two further subsets of compounds of both formulae (III) and (VII)) using an analogous series of transformations to those described in Scheme I for the conversion of amine (63) to (64) and (65). Certain compounds of formulae (III) and (VII) can be made by Scheme L:
  • Scheme L Reaction of pyridone (71) with 2-bromo-1,1-diethoxyethane (2), followed by hydrolysis of the acetal can generate aldehyde (72).
  • the alkylation reaction can be carried out in the presence of a base, such as Cs 2 CO 3 , in a solvent, such as dry NMP, at a temperature from 50-100 o C.
  • Hydrolysis can be effected using a strong acid, such as concentrated HCl, in a solvent, such as ACN, at room temperature.
  • Aldehyde (72) can be converted to ester (73) via reductive amination with the known amine (19), prepared as described in US 6645980.
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • the ester group in (73) can be converted to the alcohol (74) using a reducing agent, such as LiAlH4 or BHEt3 + Li- (super hydride), in a solvent, such as THF, at a temperature from 0 o C to room temperature.
  • Alcohol (74) can be converted to azide (75) by a twostep process involving treatment with paraformaldehyde in the presence of TMSCl at room temperature, followed by treatment with an azide reagent, such as NaN 3 , in a solvent, such as THF, at room temperature, in the presence of 18-Crown-6.
  • a 3+2 cycloaddition of the azide onto the alkyne in (75) to deliver macrocycle alkene (76) can be accomplished using pentamethylcyclopentadienyl ruthenium chloride [Cp*RuCl] complexes in a suitable solvent, such as toluene or dioxane, at a temperature from room temperature to 60 o C.
  • Deprotection of the cyclic acetal in (76) to release the ketone in (77) can be effected using acid catalysed conditions, such as use of a protic acid (10% HCl in a mixture of THF/H2O) or a Lewis acid (FeCl3 dispersed on silica).
  • Ketone (77) can be converted to amine (78) via reductive amination with amine (10).
  • the reaction can be performed using a borohydride reagent, such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of AcOH as a catalyst is optional.
  • Amine (78) can be converted to (79) (a subset of compounds of formulae (III) and (VII)) under reductive amination conditions with aldehyde (12).
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • Amine (78) can be converted to (80) (another subset of compounds of formulae (III) and (VII)) by coupling with (14), where LG represents a leaving group, such as a halide.
  • the reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating.
  • a base such as K2CO3, with optional heating.
  • macrocycles (81) and (82) (two further subsets of compounds of formulae (III) and (VII)) can be prepared.
  • the reaction can be carried out in a solvent, such as H 2 O or an alcohol, such as EtOH, at a temperature from room temperature to 80 o C.
  • Hydrolysis can be effected using a strong acid, such as concentrated HCl, in a solvent, such as ACN, at room temperature.
  • Aldehyde (99) can be converted to ester (100) via reductive amination with the known amine (19), prepared as described in US 6645980.
  • the reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80 o C. Addition of 4 ⁇ sieves is optional.
  • the ester group in (100) can be converted to carboxylic acid (101) using base hydrolysis, such as Na2CO3 in a solvent mixture of H2O/EtOH, at a temperature from 50 o C to reflux or LiOH in a solvent mixture of H 2 O/THF, at a temperature from 50 o C to 80 o C or Et3N in H2O at a temperature from room temperature to 50 o C.
  • base hydrolysis such as Na2CO3 in a solvent mixture of H2O/EtOH
  • LiOH in a solvent mixture of H 2 O/THF
  • Et3N Et3N in H2O
  • Deprotection of the nitrogen CBZ group to give amine (102) can be achieved under standard conditions, such as use of H2 in the presence of Pd/C in an alcoholic solvent, such as EtOH, at room temperature.
  • Macrolactamisation of (102) can be effected under standard amide coupling conditions, such as HBTU, in the presence of a base, such as Et3N, in a solvent, such as DMSO, at room temperature.
  • Deprotection of the cyclic ketal in macrocycle (103) to release the ketone in (104) can be effected using acid catalysed conditions, such as use of a protic acid (10% HCl in a mixture of THF/H2O) or a Lewis acid (FeCl3 dispersed on silica).
  • Ketone (104) can be converted to amine (105) via reductive amination with amine (10).
  • the reaction can be performed using a borohydride reagent, such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C. Addition of AcOH as a catalyst is optional. Amine (105) can be converted to (106) (a subset of compounds of both formula (III) and formula (VII)) under reductive amination conditions with aldehyde (12). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80 o C.
  • a borohydride reagent such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2-dichloroethane
  • Amine (105) can be converted to (107) (another subset of compounds of both formula (III) and formula (VII)) by coupling with (14), where LG represents a leaving group, such as a halide.
  • the reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating.
  • Experimental NMR spectra were obtained on a LC Bruker AV400 using a 5 mm QNP probe (Method A) or Bruker AV1500MHz with 5mm QNP probe and Z-axis gradients (Method B).
  • MS was carried out on a Waters Alliance ZQ MS (Methods A, B, C and D) using H 2 O and ACN mobile phase with pH modification as detailed under each method. Wavelengths were 254 and 210 nm.
  • Method A (Acidic pH) Column: YMC-Triart C1850 x 2 mm, 5 ⁇ m. Flow rate: 0.8 mL/min. Injection volume: 5 ⁇ L.
  • Mobile Phase A H2O B ACN C 50% H2O / 50% ACN + 1.0% formic acid
  • Method B (Acidic pH) Column: YMC Triart-C1850 x 2 mm, 5 ⁇ m Flow rate: 0.8 mL/min. Injection volume: 5 ⁇ L
  • Example 1 ( ⁇ ) trans-6-[( ⁇ 3-oxo-2H, 3H, 4H-pyrido[3,2-b][1,4]oxazin-6-yl ⁇ methyl)amino]- 8,13-dioxa-1,4-diazatetracyclo[12.6.2.14,7.017,21]tricosa-14(22),15,17 (21),18–tetraen-20- one (a) ( ⁇ ) tert-butyl trans-3-([(benzyloxy)carbonyl]amino)-4-hydroxy-pyrrolidine-1- carboxylate 1a
  • Example 3 ( ⁇ ) cis-6- ⁇ [( ⁇ 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6- yl ⁇ methyl)amino]methyl ⁇ - 8,14-dioxa-1,4-diazatetracyclo[13.6.2.14,7.018,22]tetracosa-15 (23),16,18(22),19–tetraen-21-one
  • Example 4 7-[( ⁇ 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl ⁇ methyl)amino]-14-oxa- 1,4,10-triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22),19–tetraen-9,21- dione a) benzyl N- ⁇ 3-[(2-oxo-1,2-dihydroquinolin-7-yl)oxy]propyl ⁇ carbamate 4a
  • the aqueous was further extracted with DCM and the combined organics washed with brine and concentrated in vacuo to give a crude oil.
  • the oil was purified by flash chromatography using a gradient eluent system of 0-5% MeOH in DCM to give methyl 8- ⁇ 2-[7-(3- ⁇ [(benzyloxy)carbonyl]amino ⁇ propoxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl ⁇ -1,4-dioxa-8- azaspiro[4.5]decane-6-carboxylate 4d (418 mg, 92 % yield) as a colourless oil.
  • reaction mixture was diluted with DCM and washed with saturated aqueous NaHCO 3 , dried over Na 2 SO 4 and concentrated in vacuo.
  • the resulting residue was purified by flash column chromatography using a gradient eluent system of 0-20% MeOH in DCM/3N NH3 to give 7-[( ⁇ 3-oxo- 2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl ⁇ methyl)amino]-14-oxa-1,4,10- triazatetracyclo[13.6.2.1 4 , 8 .0 18 , 22 ]tetracosa-15(23),16,18(22),19–tetraen-9,21-dione 4 (14 mg, 70 % yield) as a yellow solid.
  • Example 8 (6S, 8S)-6- ⁇ [( ⁇ 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6- yl ⁇ methyl)amino]methyl ⁇ -15-oxa-1,4,10-triazatetracyclo[14.6.2.04,8.019,23]tetracosa- 16,18,20,23-tetraene-9,22-dione (a) 1-tert-butyl 2-methyl (2S,4S)-4-( ⁇ [(benzyloxy)carbonyl]amino ⁇ methyl)pyrrolidine-1,2- dicarboxylate 8a
  • reaction mixture was then quenched with aqueous saturated ammonium chloride solution (50 mL) and extracted with Et2O (2 x 75 mL). The combined organic extracts were dried (MgSO 4 ) and concentrated in vacuo to give a yellow gum.
  • 9d 9e A stirred mixture of tert-butyl N-[2-(2-bromoethoxy)ethyl]carbamate (1.49 g, 5.55 mmol), ( ⁇ ) methyl 9-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-1,5-dioxa-9- azaspiro[5.5]undecane-7-carboxylate 9d (1.49 g, 3.70 mmol) and Cs 2 CO 3 (3.62 g, 11.11 mmol) in DMF (20 mL) was heated at 90 o C for 1 h. The reaction was allowed to cool to room temperature and diluted with H 2 O.
  • the resulting yellow gum was purified by flash column chromatography using a gradient eluent system of 50- 100% EtOAc in Et2O to give tert-butyl N-(3- ⁇ [2-oxo-1-(2-oxoethyl)-1,2-dihydroquinolin-7- yl]oxy ⁇ propyl)carbamate 10a (1.47 g, 41 % yield) as a clear gum.
  • the resulting yellow gum was purified by flash column chromatography using a gradient eluent system of 80-100% EtOAc in heptane to give methyl (2S,4S)-4-( ⁇ [(benzyloxy)carbonyl]amino ⁇ methyl)- 1- ⁇ 2-[7-(3- ⁇ [(tert-butoxy)carbonyl]amino ⁇ propoxy)-2-oxo-1,2-dihydroquinolin-1- yl]ethyl ⁇ pyrrolidine-2-carboxylate 10b (0.7 g, 27% yield) as a yellow gum.
  • reaction mixture was then flushed through a SCX cartridge and relevant fractions concentrated in vacuo to give a yellow gum, which was dissolved in DCM and treated with 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazine-6- carbaldehyde (244 mg, 1.37 mmol).
  • the reaction mixture was stirred at room temperature for 1 h, before the addition of sodium triacetoxyborohydride (377 mg, 1.78 mmol) in one portion. After 5 min the reaction mixture was quenched with MeOH, concentrated to a low volume and purified using a SCX cartridge, eluting with MeOH/NH 3.
  • the broth dilution method involves a two-fold serial dilution of compounds in 96-well microtitre plates, giving a final concentration range of 0.03-64 ⁇ g/mL or 0.25-128 ⁇ g/mL.
  • Strains are grown in cation-adjusted Müller-Hinton broth (supplemented with 2% w/v NaCl in the case of methicillin-resistant S. aureus strains and 2% IsoVitalex in the case of Francisella tularensis) or on Müller-Hinton agar at 37°C in an ambient atmosphere.
  • the MIC is determined as the lowest concentration of compound that inhibits growth following an incubation period of 16-24 h, of 12-24 h (Bacillus anthracis), of 46-50 h (Francisella tularensis) or of 24-48 h (Yersinia pestis). Table 1 - MIC values against Gram-negative and Gram-positive bacterial strains
  • HepG2 ATCC HB-8065 Human hepatic cell line
  • HepG2 cells are seeded at 20,000 cells/well in 96-well microtitre plates in minimal essential medium (MEM) supplemented with a final concentration of 10% FBS and 1 mM sodium pyruvate.
  • MEM minimal essential medium
  • compound dilutions are prepared in Dulbecco’s minimum essential media (DMEM) supplemented with final concentrations of 0.001% FBS, 0.3% bovine albumin and 0.02% HEPES and added to cells.
  • DMEM minimum essential media
  • IC 50 (in ⁇ g/mL) of less than 1 is assigned the letter D; an IC 50 of from 1 to 10 is assigned the letter C; an IC 50 of from 10 to 100 is assigned the letter B; and an IC 50 of over 100 is assigned the letter A.

Abstract

The present invention relates to antibacterial drug compounds containing a polycyclic ring system incorporating a macrocyclic ring. The invention also relates to pharmaceutical formulations of antibacterial drug compounds. The invention also relates to uses of the derivatives in treating bacterial infections and in methods of treating bacterial infections. The invention is also directed to antibacterial drug compounds which are capable of treating bacterial infections which are currently hard to treat with existing drug compounds.

Description

Antibacterial compounds This invention relates to antibacterial drug compounds containing a polycyclic ring system incorporating a macrocyclic ring. It also relates to pharmaceutical formulations of antibacterial drug compounds. It also relates to uses of the derivatives in treating bacterial infections and in methods of treating bacterial infections. The invention is also directed to antibacterial drug compounds which are capable of treating bacterial infections which are currently hard to treat with existing drug compounds. Such infections are frequently referred to as resistant strains. The increasing occurrence of bacterial resistance to antibiotics is viewed by many as being one of the most serious threats to the future health and happiness of mankind. Multidrug resistance has become common among some pathogens, e.g. Staphylococcus aureus, Streptococcus pneumoniae, Clostridium difficile and Pseudomonas aeruginosa. Of these, Staphylococcus aureus, a Gram positive bacterium, is the most concerning due to its potency and its capacity to adapt to environmental conditions. MRSA (methicillin resistant Staphylococcus aureus) is probably the most well known group of resistant strains and has reached pandemic proportions. Of particular concern is the increasing incidence of ‘community acquired’ infections, i.e. those occurring in subjects with no prior hospital exposure. While less widespread, antibiotic resistant Gram negative strains, such as either Escherichia coli NDM-1 (New Delhi metallo-β-lactamase) or Klebsiella pneumoniae NDM-1, are also very difficult to treat. Frequently only expensive last resort antibiotics such as vancomycin and colistin are effective against these strains. The fluoroquinolone antibacterial family are synthetic broad-spectrum antibiotics. They were originally introduced to treat Gram negative bacterial infections, but are also used for the treatment of Gram positive strains. One problem with existing fluoroquinolones can be the negative side effects that may sometimes occur as a result of fluoroquinolone use. In general, the common side-effects are mild to moderate but, on occasion, more serious adverse effects occur. Some of the serious side effects that occur, and which occur more commonly with fluoroquinolones than with other antibiotic drug classes, include central nervous system (CNS) toxicity and cardiotoxicity. In cases of acute overdose there may be renal failure and seizure. In addition, an increasing number of strains of MRSA are also resistant to fluoroquinolone antibiotics, in addition to β-lactam antibiotics such as methicillin. Bacterial resistance is also becoming a problem in the treatment of animals. Antibacterials find widespread use in industrial farming, e.g. to prevent mastitis in dairy cattle, where they are often used prophylactically. Such widespread prophylactic use has led to the build-up of resistance in certain bacterial strains which are particularly relevant to animal health. In spite of the numerous different antibiotics known in the art for a variety of different infections, there continues to be a need for antibiotics that can provide an effective treatment in a reliable manner. In addition, there remains a need for antibiotic drugs which can avoid or reduce the side-effects associated with known antibiotics. It is an aim of certain embodiments of this invention to provide new antibiotics. In particular, it is an aim of certain embodiments of this invention to provide antibiotics which are active against resistant strains of Gram positive and/or Gram negative bacteria. It is an aim of certain embodiments of this invention to provide compounds which have activity which is comparable to those of existing antibiotics, and ideally which is better. It is an aim of certain embodiments of this invention to provide such activity against wild-type strains at the same time as providing activity against one or more resistant strains. It is an aim of certain embodiments of this invention to provide compounds which exhibit a smaller reduction in activity against resistant strains compared to wild-type strains than prior art compounds do. It may be that certain compounds of the invention are less active than prior art compounds but there is a benefit associated with having a more consistent activity against a range of strains. It is an aim of certain embodiments of this invention to provide antibiotics which exhibit reduced cytotoxicity relative to prior art compounds and existing therapies. It is an aim of certain embodiments of this invention to provide treatment of bacterial infections which is effective in a selective manner at a chosen site of interest. Another aim of certain embodiments of this invention is to provide antibiotics having a convenient pharmacokinetic profile and a suitable duration of action following dosing. A further aim of certain embodiments of this invention is to provide antibiotics in which the metabolised fragment or fragments of the drug after absorption are GRAS (Generally Regarded As Safe). Certain embodiments of the present invention satisfy some or all of the above aims. Compounds of the Invention
In a first aspect, the invention provides a compound of formula (I), or a pharmaceutically acceptable salt or N-oxide thereof:
Figure imgf000004_0001
group A is selected from a 5-12heterocycloalkyl group comprising at least one nitrogen in the ring system and a 3-6heteroalkyl group comprising at least one nitrogen in the linking chain; L3 is attached by a covalent bond to an atom selected from the nitrogen and carbon atoms which form the group A ring system or linking chain; Z is independently selected from N and CR2; R1 and R2 are each independently selected from: H, C1-C4-alkyl, halogen, OR8, NR8R9 and C1-C4-haloalkyl; X1, X2, X3 and X4 are each independently selected from: N and CR10; wherein no more than two of X1, X2, X3 and X4 are N; wherein a single one of X3 and X4 is a carbon atom attached by a covalent bond to Y1; Y1 is independently selected from O, CR5R5, -C(R8)=C(R8)-, NR9, S and S(O)2; R5 is independently at each occurrence selected from: H, F, C1-C4-alkyl, NR8R9, OR8, C1-C4- haloalkyl and CO2R8; or two R5 groups attached to the same carbon together form =O; L1 is a linker group having the form -(CR5R5)r-; wherein r is an integer selected from 2 and 3; L5 is absent or is -L6-L2-; L6 is absent or is–L4NR6-; L2 is independently selected from–CR5R5- and a 3-, 4- or 5- membered cycloalkyl or heterocycloalkyl ring; L3 is independently –(CR5R5)s-Y3-(CR5R5)t-Y2-(CR5R5)u-; wherein s and t are each independently an integer selected from 1, 2, 3 and 4; u is an integer independently selected from 0 and 1; Y2 and Y3 are each independently selected from a bond, O, - C(R8)=C(R8)-, 1,2,3-triazole, NR9, S and S(O)2; and wherein L1, group A, r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size from 13 to 17 atoms; L4 is independently a bond or is–CR5R5-; R6 and R8 are independently at each occurrence selected from: H, C1-C4-alkyl and C1-C4- haloalkyl; R7 is a monocyclic aromatic or heteroaromatic ring or R7 is a bicyclic carbocyclic or heterocyclic ring system in which at least one of the two rings is aromatic or heteroaromatic; R9 is independently at each occurrence selected from H, C1-C4-alkyl, C1-C4-haloalkyl, S(O)2R8, C(O)NR8R8, C(O)R8 and C(O)OR8; R10 is independently at each occurrence selected from: H, halo, nitro, cyano, NR8R9, OR8; O-aryl, SR8, SOR8, SO3R8, SO2R8, SO2NR8R8, CO2R8, C(O)R8, CONR8R8, aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; wherein each of the aforementioned alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, carbocyclic, halocycloalkyl, heterocyclic, aryl (e.g. phenyl) and heteroaryl groups and aromatic and heteroaromatic rings is optionally substituted, where chemically possible, by 1 to 5 substituents which are each independently at each occurrence selected from the group consisting of: oxo, =NRa, =NORa, halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa , CO2Ra C(O)Ra, CONRaRa, CRaRaNRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; wherein Ra is independently at each occurrence selected from H and C1-C4-alkyl. Thus, the compound of formula (I) may be a compound of formula (II):
Figure imgf000006_0001
(II); wherein L3 is attached by a covalent bond to an atom selected from Na, Ca, Cb and Cc; wherein if L3 is attached to Na, R3 is absent; and wherein the positions on Ca, Cb and Cc to which L3 is not attached are occupied by R5 groups; Z is independently selected from N and CR2; R1 and R2 are each independently selected from: H, C1-C4-alkyl, halogen, OR8, NR8R9 and C1-C4-haloalkyl; R3 is absent (if L3 is joined to Na) or is independently selected from H and C1-C4-alkyl; R4 is independently selected from: H, C1-C4-alkyl, F, NR8R9, OR8, C1-C4-haloalkyl and CO2R8; or R3 and R4 together form a–(CR5R5)n- group; wherein n is an integer selected from 1, 2 and 3; or R4 and an R5 attached to the same carbon as the R4 group together form =O; R5 is independently at each occurrence selected from: H, F, C1-C4-alkyl, NR8R9, OR8, C1-C4- haloalkyl and CO2R8; or two R5 groups attached to the same carbon together form =O; X1, X2, X3 and X4 are each independently selected from: N and CR10; wherein no more than two of X1, X2, X3 and X4 are N; wherein a single one of X3 and X4 is a carbon atom attached by a covalent bond to Y1; Y1 is independently selected from O, CR5R5, -C(R8)=C(R8)-, NR9, S and S(O)2; L1 is a linker group having the form -(CR5R5)r-; wherein r is an integer selected from 2 and 3; L2 is independently selected from–CR5R5- and a 3-, 4- or 5- membered cycloalkyl or heterocycloalkyl ring; L3 is independently –(CR5R5)s-Y3-(CR5R5)t-Y2-(CR5R5)u-; wherein s and t are each independently an integer selected from 1, 2, 3 and 4; u is an integer independently selected from 0 and 1; Y2 and Y3 are each independently selected from a bond, O, - C(R8)=C(R8)-, 1,2,3-triazole, NR9, S and S(O)2; and wherein r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size from 13 to 17 atoms; L4 is independently a bond or is–CR5R5-; R6 and R8 are independently at each occurrence selected from: H, C1-C4-alkyl and C1-C4- haloalkyl; R7 is a monocyclic aromatic or heteroaromatic ring or R7 is a bicyclic carbocyclic or heterocyclic ring system in which at least one of the two rings is aromatic or heteroaromatic; R9 is independently at each occurrence selected from H, C1-C4-alkyl, C1-C4-haloalkyl, S(O)2R8, C(O)NR8R8, C(O)R8 and C(O)OR8; R10 is independently at each occurrence selected from: H, halo, nitro, cyano, NR8R9, OR8; O-aryl, SR8, SOR8, SO3R8, SO2R8, SO2NR8R8, CO2R8, C(O)R8, CONR8R8, aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; m is an integer independently selected from 0 and 1; wherein each of the aforementioned alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, carbocyclic, halocycloalkyl, heterocyclic, aryl (e.g. phenyl) and heteroaryl groups and aromatic and heteroaromatic rings is optionally substituted, where chemically possible, by 1 to 5 substituents which are each independently at each occurrence selected from the group consisting of: oxo, =NRa, =NORa, halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa , CO2Ra C(O)Ra, CONRaRa, CRbRbNRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; wherein Ra is independently at each occurrence selected from H and C1-C4-alkyl. For the absence of doubt, the linker group L3 is arranged such that the group Y1 is attached to the end of linker group L3 which is defined as (CR5R5)s. Thus, the atom Na, Ca, Cb or Cc is attached to the end of linker group L3 which is defined as (CR5R5)u. Ca could be described as a C(R5)x group, Cb (if present) could be described as a C(R5)y group and Cc could be described as a C(R5)z group, wherein x and y are each an integer selected from 1 and 2 and z is an integer selected from 0 and 1, with the values of x, y and z being selected to satisfy valency requirements. Thus if L3 is attached to Na, both x and y are 2 and Z is 1. If L3 is attached to Ca, x is 1, y is 2 and z is 1. If L3 is attached to Cb, x is 2, y is 1 and z is 1. If L3 is attached to Cc, x and y are each 2 and z is 0. The macrocyclic ring is the ring including the atoms or groups represented by L1, L3, Y1,and X4. Depending on the point of attachment of Y1 and L3, the macrocyclic ring may also include one or more of the atoms or groups represented by Na, Ca, Cb, Cc and X3. Where Y1, Y2 or Y3 is a -C(R8)=C(R8)- group, the compound may be the E geometric isomer, the Z geometric isomer or a mixture thereof. Two R5 groups attached to the same carbon may together form =O. Preferably, however, where there are two CR5R5 groups directly bonded to each other, only one of those pair of R5 groups may form =O, in other words only one of those CR5R5 groups may be C=O. I ) may be a compound of formula (III):
Figure imgf000008_0001
(III)
wherein R1, R2, R3, R4, R5, R6, R7, L1, L2, L3, L4, Y1, X1, X2 and X4 are as defined above for formula (II). In this embodiment, r may be 2. In an embodiment, the compound of formula (I) may be a compound of formula (IV):
Figure imgf000009_0001
wherein R1, R2, R3, R4, R5, R6, R7, L1, L2, L3, L4, Y1, X1, X2 and X4 are as defined above for formula (II). In this embodiment, r may be 2. In an embodiment, the compound of formula (I) may be a compound of formula (V):
Figure imgf000009_0002
wherein R1, R2, R5, R6, R7, L1, L2, L3, Y1, X1, X2 and X4 are as defined above for formula (II) and wherein R4 is independently selected from: H, C1-C4 alkyl, C1-C4 haloalkyl and CO2R8; or wherein R4 and the R5 group attached to the same carbon as R4 group together form =O. In this embodiment, r may be 2. In an embodiment, the compound of formula (I) may be a compound of formula (VI):
Figure imgf000009_0003
wherein R1, R2, R3, R5, R6, R7, L1, L2, L3, L4, Y1, X1, X2, and X4 are as defined above for formula (II) and wherein R4 is independently selected from: H, C1-C4-alkyl, C1-C4-haloalkyl and CO2R8; or R3 and R4 together form a–(CR5R5)n- group; wherein n is an integer selected from 1, 2 and 3. In this embodiment, r may be 2. In an embodiment, the compound of formula (I) may be a compound of formula (VII):
Figure imgf000010_0001
wherein R1, R2, R5, R6, R7, L1, L2, L3, L4, Y1, X1, X2, X4 and n are as defined above for formula (II) and wherein L3 is attached by a covalent bond to an atom selected from Ca, Cb and Cc. In this embodiment, r may be 2. In an embodiment, the compound of formula (I) may be a compound of formula (VIII):
Figure imgf000010_0002
wherein R1, R2, R4, R5, R6, R7, L1, L2, L3, Y1, X1, X2 and X4 are as defined above for formula (II) and wherein R4 is independently selected from: H, C1-C4-alkyl, C1-C4-haloalkyl and CO2R8; or wherein R4 and the R5 group attached to the same carbon as R4 group together form =O. In this embodiment, r may be 3. In an embodiment, the compound of formula (I) may be a compound of formula (IX):
Figure imgf000010_0003
wherein R1, R2, R5, X1, X2, X4, Y1, L1, L3, L2, n and R7 are as defined for formula (II) above; wherein L3 is attached by a covalent bond to an atom selected from Cd, Ce and Cf and wherein the positions on Cd, Ce and Cf to which L3 is not attached are occupied by R5 groups. In an embodiment, the compound of formula (I) may be a compound of formula (X):
Figure imgf000011_0001
wherein R1, R2, R5, X1, X2, X4, Y1, L1, L3, n and R7 are as defined for formula (II) above; wherein Xa is carbon or nitrogen; and wherein L3 is attached by a covalent bond to an atom selected from Cg, Ch and, where Xa is carbon, Xa and wherein any positions on Cg, Ch and Xa to which L3 is not attached are occupied by R5 groups. In an embodiment, the compound of formula (I) may be a compound of formula (XI):
Figure imgf000011_0002
L3 is attached by a covalent bond to an atom selected from Ca, Cb and Cc; and wherein the positions on Ca, Cb and Cc to which L3 is not attached are occupied by R5 groups; R1, R2, R5, R6 and R8 are each independently at each occurrence selected from: H and C1- C4-alkyl; X1, X2 and X4 are each independently selected from: N and CR10; wherein no more than two of X1, X2 and X4 are N; L1 is a linker group having the form -(CR5R5)r-; wherein r is an integer selected from 2 and 3; L3 is independently –(CR5R5)s-Y3-(CR5R5)t-Y2-(CR5R5)u-; wherein s and t are each independently an integer selected from 1, 2, 3 and 4; u is an integer independently selected from 0 and 1; Y2 and Y3 are each independently selected from a bond, O, - C(R8)=C(R8)-, 1,2,3-triazole, NR9, S and S(O)2; and wherein r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size from 13 to 17 atoms; L4 is independently a bond or is–CR5R5-; R9 is independently at each occurrence selected from H, C1-C4-alkyl, S(O)2R8, C(O)NR8R8, C(O)R8 and C(O)OR8; R10 is independently at each occurrence selected from: H, halo, nitro, cyano, NR8R9, OR8; O-aryl, SR8, SOR8, SO3R8, SO2R8, SO2NR8R8, CO2R8, C(O)R8, CONR8R8, aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; m is an integer independently selected from 0 and 1; n is an integer selected from 1, 2 or 3; V1, V2 and V3 are each independently selected from: N and CR11; with the proviso that no more than two of V1, V2 and V3 are N; the ring B is a substituted or unsubstituted 5- or 6- membered saturated cycloalkyl or heterocycloalkyl ring; R11 is independently at each occurrence selected from: H, halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa , CO2Ra C(O)Ra, CONRaRa, CRaRaNRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl. wherein each of the aforementioned alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, carbocyclic, halocycloalkyl, heterocyclic, aryl (e.g. phenyl) and heteroaryl groups and aromatic and heteroaromatic rings is optionally substituted, where chemically possible, by 1 to 5 substituents which are each independently at each occurrence selected from the group consisting of: oxo, =NRa, =NORa, halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa, CO2Ra C(O)Ra, CONRaRa, CRaRaNRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; wherein Ra is independently at each occurrence selected from H and C1-C4-alkyl. The following statements apply to compounds of any of formulae (I) to (XI). These statements are independent and interchangeable. In other words, any of the features described in any one of the following statements may (where chemically allowable) be combined with the features described in one or more other statements below. In particular, where a compound is exemplified or illustrated in this specification, any two or more of the statements below which describe a feature of that compound, expressed at any level of generality, may be combined so as to represent subject matter which is contemplated as forming part of the disclosure of this invention in this specification. Group A may be a 5-12heterocycloalkyl group comprising at least one nitrogen in the ring system. Thus, group A may be a 5-, 6- or 7-membered heterocycloalkyl ring comprising at least one nitrogen in the ring. Group A may be a piperidine, piperazine or pyrrolidine ring. It may be that the group L1 is attached to the nitrogen or a nitrogen in the group A ring, e.g. the nitrogen or a nitrogen in the 5-, 6- or 7-membered heterocycloalkyl ring (e.g. a piperidine, piperazine or pyrrolidine ring). Group A may be a 3-6heteroalkyl group comprising at least one nitrogen in the linking chain. It may be that the group L1 is attached to the nitrogen or a nitrogen in the group A ring. It may be that L3 is attached to Ca. In this case, a single R5 group is also attached to Ca, two R5 groups are attached to Cb and a single R5 group is attached to Cc, i.e. x is 1, y is 2 and z is 0. It may be that L3 is attached to Cb. In this case, a single R5 group is also attached to Cb, two R5 groups are attached to Ca and a single R5 group is attached to Cc, i.e. x is 2, y is 1 and z is 0. As would be readily apparent to the skilled person, where L3 is attached to Cb, m is 1. It may be that L3 is attached to Na. In this case, two R5 groups are attached to Ca, two R5 groups are attached to Cb and a single R5 group is attached to Cc, i.e. x is 2, y is 2 and z is 1. It may be that L3 is attached to Cc. In this case, two R5 groups are attached to Ca and two R5 groups are attached to Cb, i.e. x is 2, y is 2 and z is 0. It may be that L3 is attached to Ca, Cb or Cc. Preferably, X3 is a carbon atom which is attached by a covalent bond to Y1. In this case, X1, X2 and X4 are each independently selected from: N and CR10; wherein no more than two of X1, X2 and X4 are N. It may be that each of X1, X2 and X4 are CR10. It may be that X1 is N. Preferably, X1 is CR10a, wherein R10a is independently selected from: H, halo, nitro, cyano, NR8R9, OR8; O-aryl, SR8, SOR8, SO3R8, SO2R8, SO2NR8R8, CO2R8, C(O)R8, CONR8R8, aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl. R10a may be selected from H, halo, C1-C4-alkyl and C1-C4-haloalkyl. Thus, X1 may be CH. It may be that X2 is N. Preferably, X2 is CR10b, wherein R10b is independently selected from: H, halo, nitro, cyano, NR8R9, OR8; O-aryl, SR8, SOR8, SO3R8, SO2R8, SO2NR8R8, CO2R8, C(O)R8, CONR8R8, aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl. R10b may be selected from H, halo, C1-C4-alkyl and C1-C4-haloalkyl. Thus, X2 may be CH. It may be that X4 is N. Preferably, X4 is CR10c, wherein R10c is independently selected from: H, halo, nitro, cyano, NR8R9, OR8; O-aryl, SR8, SOR8, SO3R8, SO2R8, SO2NR8R8 , CO2R8, C(O)R8, CONR8R8, aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl. R10c may be selected from H, halo, C1-C4-alkyl and C1-C4-haloalkyl. Thus, X4 may be CH. It may be that X1, X2 and X4 are each CH. It may be that X1, X2 and X4 are each CH and R1 and R2 are each H. R10 may be independently at each occurrence selected from H, halo, C1-C4-alkyl and C1-C4- haloalkyl. R10 may at all occurrences be H. Preferably, Z is CR2. It may be that R1 is H. It may be that R2 is H, i.e. that Z is CH. Thus, it may be that R1 and R2 are each H, i.e. that R1 is H and Z is CH. Y1 may be selected from O and CR5R5. Alternatively, Y1 may be selected from O, S and NR9. It may be that Y1 is CR5R5. Thus, it may be that Y1 is C=O, In certain embodiments, Y1 is CH2. In certain other embodiments, Y1 is O. r may be 2. Alternatively, r may be 3. It may be that each R5 group in the linker L1 is H. Thus, L1 may be–(CH2)2-. Alternatively, L1 may be–(CH2)3-. m may be 0. Alternatively, m may be 1. Where m is 0, it may be that r is 3. Where m is 1, it may be that r is 2. L3 may be independently–(CR5R5)s-Y3-(CR5R5)t-Y2-(CR5R5)u-; wherein s and t are each independently an integer selected from 1, 2, 3 and 4; u is an integer independently selected from 0 and 1; Y2 and Y3 are each independently selected from a bond, O, - C(R8)=C(R8)-, NR9, S and S(O)2; and wherein r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size from 13 to 17 atoms. L5 may be absent. Preferably, L5 is -L6-L2-; L6 may be absent. Preferably, L6 is–L4NR6-; L4 may be a bond. This will typically be the case where R3 and R4 do not together form a– (CR5R5)n- group. L4 may be–CR5R5-. This may be the case where R3 and R4 together form a–(CR5R5)n- group. It may be that u is 1. It may be that u is 0. It may be that s is 1. It may be that t is an integer selected from 1, 2 and 3. It may be that s is an integer selected from 2, 3 and 4. It may be that t is an integer selected from 2, 3 and 4. It may be that Y2 is selected from O, NR9 and S. It may be that Y2 is O. In these embodiments, it may be that u is 1. It may be that R5 is, at each occurrence in the group- (CR5R5)u-, H. These embodiments are particularly preferred where L3 is attached to Ca, Cb or Cc. It may be that the group -(CR5R5)u- is–C(O)- and that Y2 is selected from O and NR9. Thus it may be that Y2 is NR9, e.g. NH. In these embodiments, it may be that Y3 is a bond. Alternatively, it may be that Y2 is a bond. When u is 0, it may be that Y2 is a bond. Where L3 is attached to Na and Y3 is selected from O, NR9 and S, it may be that u is 0 and Y2 is a bond. It may be that Y3 is a bond. Alternatively, it may be that Y3 is selected from O, NR9 and S. It may be that Y3 is O. It may be that u is 0, Y2 is a bond and Y3 is selected from O, NR9 and S (e.g. is O). Where L3 is attached to Na, it may be that u is 0, Y2 is a bond and Y3 is selected from O, NR9 and S (e.g. Y3 is O). Where L3 is attached to Ca, Cb or Cc, it may be that u is 1, Y2 is selected from O, NR9 and S (e.g. is O) and Y3 is a bond. It may be that Y1 and Y2 are each independently selected from O, NR9 and S (e.g. are each O), Y3 is a bond, u is 1, s is 1 and t is an integer selected from 1, 2 and 3. In these embodiments, it may be that L3 is attached to Ca, Cb or Cc. It may be that Y1 and Y3 are each independently selected from O, NR9 and S (e.g. are each O), Y2 is a bond, u is 0, and s and t are each independently an integer selected from 2, 3, and 4. In these embodiments, it may be that L3 is attached to Na. It may be that Y2 and Y3 are each a bond. Thus, it may be that Y2 and Y3 are each a bond and u is 0. Thus, it may be that Y2 and Y3 are each a bond, u is 0, s is an integer selected from 1 and 2 and t is an integer selected from 1, 2, 3 and 4. In these embodiments, it may be that Y1 is O. Alternatively, it may be that Y1 is CR5R5. It may be that Y3 is 1,2,3-triazole. Where Y3 is 1,2,3-triazole, the triazole may have a structure selected from:
Figure imgf000017_0001
triazole, preferably Y3 has the structure
Figure imgf000017_0002
Where Y3 is 1,2,3- triazole, it may be that s and t are each 1. Where Y3 is 1,2,3-triazole, it may be that Y2 is a bond. Where Y3 is 1,2,3-triazole, it may be that Y2 is a–O-. Where Y3 is 1,2,3-triazole, it may be that s, t and u are each 1 and Y2 is -O-.
Figure imgf000017_0003
left hand side of the L3 structure as it is depicted above is attached to Y1 and the right hand side of the L3 structure as it is depicted above is attached to group A.
It may be that any R5 group that is attached to a carbon which is also attached to a nitrogen or oxygen atom is independently at each occurrence selected from: H, C1-C4-alkyl, C1-C4- haloalkyl and CO2R8; or two R5 groups attached to the same carbon together form =O. It may be that R5 is independently at each occurrence selected from: H, C1-C4-alkyl, C1-C4- haloalkyl and CO2R8; or two R5 groups attached to the same carbon together form =O. It may be that each R5 group in the linker L3 is H. It may be that R5 is at all occurrences H. It may be that R3 is independently selected from H and C1-C4-alkyl. Thus, R3 may be H. R3 may be C1-C4-alkyl, e.g. methyl. It may be that, where L4 is a bond, R4 is independently at each occurrence selected from: H, C1-C4-alkyl, C1-C4-haloalkyl and CO2R8; or R3 and R4 together form a–(CR5R5)n- group; wherein n is an integer selected from 1, 2 and 3; or R4 and an R5 attached to the same carbon as the R4 group together form =O. It may be that R4 is independently selected from: H, C1-C4-alkyl, C1-C4-haloalkyl and CO2R8; or that R4 and an R5 attached to the same carbon as R4 group together form =O. Thus, it may be that R3 is independently selected from H and C1-C4-alkyl and R4 is independently selected from: H, C1-C4-alkyl, C1-C4-haloalkyl and CO2R8; or that R4 and an R5 attached to the same carbon as R4 together form =O. In these embodiments, it may be that m is 0. It may be that R3 and R4 together form a–(CR5R5)n- group; wherein n is an integer selected from 1, 2 and 3. It may be that n is 2. It may be that each R5 group is the group formed by R3 and R4 is H. Thus, it may be that R3 and R4 together form a–CH2CH2- group. In these embodiments, it may be that m is 1. In these embodiments, it may be that L4 is–CR5R5-. L2 is preferably–CR5R5- and most preferably is -CH2-. Alternatively, L2 may be a 3-, 4- or 5- membered cycloalkyl ring, e.g. a 4-membered cycloalkyl ring. R7 may be a monocyclic aryl group. Thus, R7 may be a phenyl group. Said phenyl group may be unsubstituted or it may be substituted with from 1 to 5 substituents which are each independently at each occurrence selected from the group consisting of: halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaCONRaRa, NRaCO2Ra, NRaC(O)Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa , CO2Ra C(O)Ra, CONRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl, C1- C4-haloalkyl, and CRbRbNRaRa. R7 may be a phenyl group which is substituted by 1 to 3 substituents independently at each occurrence selected from F, nitro, C1-C4-alkyl, C2-C4- alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl. Exemplary R7 groups include 2,5-difluorophen-1-yl and 3-nitro-4-methylphen-1-yl. Preferably, R7 is a bicyclic carbocyclic or heterocyclic ring system in which at least one of the two rings is aromatic or heteroaromatic. Thus, R7 may take the form:
Figure imgf000019_0001
wherein V1, V2 and V3 are each independently selected from: N and CR11; with the proviso that no more than two of V1, V2 and V3 are N; and wherein the ring B is a substituted or unsubstituted 5- or 6- membered saturated cycloalkyl or heterocycloalkyl ring. R11 is independently at each occurrence selected from: H, halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa , CO2Ra C(O)Ra, CONRaRa, CRbRbNRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4- haloalkyl. Preferabl , R7 takes the form:
Figure imgf000019_0002
wherein V4 and V5 are each independently selected from O, S, CR12R12 and NR8 ; R12 is independently at each occurrence selected from: H, fluoro, cyano, CO2Ra, C(O)Ra, CONRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; or any two R12 groups which are attached to the same carbon together form a group selected from: =O, =NRa, and =NORa ; and p is an integer selected from 1 and 2. Preferabl , R7 takes the form:
Figure imgf000019_0003
It may be that V1, V2 and V3 are each independently selected from: N and CH; with the proviso that no more than two of V1, V2 and V3 are N. Preferably, a single one of V1, V2 and V3 is N. Preferably, V3 is CR11 (e.g. CH). Thus, it may be that V1 is N and V2 is CR11 (e.g. CH). Alternatively, it may be that V2 is N and V1 is CR11 (e.g. CH). It may be that V2 is nitrogen. It may be that V3 is CR11. It may be that V1 is CR11. It may be that R11 is selected from H, methyl and halogen, e.g. F. It may be that R11 is selected from H and halogen, e.g. F. It may be that V3 is CH. It may be that V1 is CR11, wherein R11 is selected from H and halogen, e.g. F. It may be that V2 is nitrogen, V3 is CH and V1 is CR11, wherein R11 is selected from H and halogen, e.g. F. Preferably R12 is at each occurrence H, F, C1-C4-alkyl or C1-C4-haloalkyl; or any two R12 groups which are attached to the same carbon together form a =O group. In a preferred embodiment, V4 is O. Thus, it may be that both V4 and V5 are O. It may be that V4 is O and V5 is S. It may be that V4 is O and V5 is NR8 (e.g. NH). V4 can also be S. Thus, it may be that V4 is S and V5 is NR8 (e.g. NH). It may be that V5 is NR8 (e.g. NH). In this case it is preferable that the–CR12R12- group attached to said V5 is C=O. p may be 1. Preferably, p is 2. In a specific embodiment, V4 is O, V5 is O, p is 2 and R12 is in all instances H. In another specific embodiment, V4 is O, V5 is S, p is 2 and R12 is in all instances H. In yet another specific embodiment, V4 is O, V5 is NH, p is 2, the–CR12R12- group attached to V5 is C=O and the–CR12R12- group attached to V4 is CH2. Exemplary R7 groups include:
F O N HN
O , ,
Figure imgf000021_0001
R7 may also take the form
Figure imgf000021_0002
, wherein V6 is independently selected from N and CR13 (e.g. CH); V7 is independently selected from NR8, S and O; and R13 is independently at each occurrence selected from: halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, NRaC(O)Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa , CO2Ra C(O)Ra, CONRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl, C1-C4-haloalkyl, and CRaRaNRaRa. R13 may be independently at each occurrence selected from F, CN, ORa, nitro, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl.
Figure imgf000021_0003
may a e e orm .
An exemplary R7 group is
Figure imgf000021_0004
. Preferably, R6 is selected from H or C1-C4-alkyl. Even more preferably, R6 is H. It may be that R8 is independently at each occurrence selected from H or C1-C4-alkyl. It may be that R8 is at each occurrence H. It may be that R9 is independently at each occurrence selected from H, C1-C4-alkyl, C1-C4- haloalkyl, S(O)2R8 and C(O)R8. It may be that R9 is independently at each occurrence selected from H, C1-C4 alkyl (e.g. methyl) and C(O)R8 (e.g. acetate). It may be that R9 is at each occurrence H. It may be that L1, group A, r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 13 atoms. It may be that L1, group A, r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 14 atoms. It may be that L1, group A, r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 15 atoms.
It may be that L1, group A, r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 16 atoms. It may be that L1, group A, r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 17 atoms. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 13 atoms. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 13 atoms and R3 and R4 together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 13 atoms and R3 and R4 do not together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 14 atoms. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 14 atoms and R3 and R4 together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 14 atoms and R3 and R4 do not together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 15 atoms. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 15 atoms and R3 and R4 together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 15 atoms and R3 and R4 do not together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 16 atoms. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 16 atoms and R3 and R4 together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 16 atoms and R3 and R4 do not together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 17 atoms. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 17 atoms and R3 and R4 together form a–(CR5R5)n- group. It may be that r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size of 17 atoms and R3 and R4 do not together form a–(CR5R5)n- group. The compound of formula (I) may have a structure selected from:
Figure imgf000023_0001
Figure imgf000023_0002
Figure imgf000024_0001
Figure imgf000025_0001
It may be that the compound of formula (I) is selected from compounds 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 and 11 in the Examples below. Where the compound of the invention is an N-oxide, it will typically be a pyridine N-oxide, i.e. where the compound of the invention comprises a pyridine ring (which may form part of a bicyclic or tricyclic ring system), the nitrogen of that pyridine may be N+-O-. Alternatively, it may be that the compound of the invention is not an N-oxide. Included within the scope of the present invention are all stereoisomers, geometric isomers and tautomeric forms of the compounds of the invention, including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Also included are acid addition or base salts wherein the counter ion is optically active, for example, d-lactate or l-lysine, or racemic, for example, dl-tartrate or dl-arginine. Compounds of the invention containing one or more asymmetric carbon atoms can exist as two or more stereoisomers. Where a compound of the invention contains a double bond such as a C=C or C=N group, geometric cis/trans (or Z/E) isomers are possible. Specifically, the oxime groups present in certain compounds of the invention may be present as the E-oxime, as the Z- oxime or as a mixture of both in any proportion. Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation. Where structurally isomeric forms of a compound are interconvertible via a low energy barrier, tautomeric isomerism (‘tautomerism’) can occur. This can take the form of proton tautomerism in compounds of the invention containing, for example, an imino, keto, or oxime group, or so- called valence tautomerism in compounds which contain an aromatic moiety. Conventional techniques for the preparation/isolation of individual enantiomers when necessary include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of the invention contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted into the corresponding pure enantiomer(s) by means well known to a skilled person. Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1% diethylamine. Concentration of the eluate affords the enriched mixture. When any racemate crystallises, crystals of two different types are possible. The first type is the racemic compound (true racemate) referred to above wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts. The second type is the racemic mixture or conglomerate wherein two forms of crystal are produced in equimolar amounts each comprising a single enantiomer. While both of the crystal forms present in a racemic mixture have identical physical properties, they may have different physical properties compared to the true racemate. Racemic mixtures may be separated by conventional techniques known to those skilled in the art - see, for example,“Stereochemistry of Organic Compounds” by E. L. Eliel and S. H. Wilen (Wiley, 1994). It follows that a single compound may exhibit more than one type of isomerism. The term Cm-Cn refers to a group with m to n carbon atoms. The term“alkyl” refers to a linear or branched hydrocarbon chain. For example, C1-C6-alkyl may refer to methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, n-pentyl and n- hexyl. The alkyl groups may be unsubstituted or substituted by one or more substituents. Specific substituents for each alkyl group independently may be fluorine, ORa or NRaRa. The term“haloalkyl” refers to a hydrocarbon chain substituted with at least one halogen atom independently chosen at each occurrence from: fluorine, chlorine, bromine and iodine. The halogen atom may be present at any position on the hydrocarbon chain. For example, C1-C6-haloalkyl may refer to chloromethyl, fluoromethyl, trifluoromethyl, chloroethyl e.g.1-chloroethyl and 2-chloroethyl, trichloroethyl e.g.1,2,2-trichloroethyl, 2,2,2- trichloroethyl, fluoroethyl e.g. 1-fluoroethyl and 2-fluoroethyl, trifluoroethyl e.g. 1,2,2- trifluoroethyl and 2,2,2-trifluoroethyl, chloropropyl, trichloropropyl, fluoropropyl, trifluoropropyl. A haloalkyl group may be a fluoroalkyl group, i.e. a hydrocarbon chain substituted with at least one fluorine atom.
The term“alkenyl” refers to a branched or linear hydrocarbon chain containing at least one double bond. The double bond(s) may be present as the E or Z isomer (e.g. cis or trans). The double bond may be at any chemically possible position of the hydrocarbon chain. For example,“C2-C6-alkenyl” may refer to ethenyl, propenyl, butenyl, butadienyl, pentenyl, pentadienyl, hexenyl and hexadienyl. The alkenyl groups may be unsubstituted or substituted by one or more substituents. Specific substituents for any saturated carbon in each alkenyl group independently may be fluorine, ORa or NRaRa.
The term“alkynyl” refers to a branched or linear hydrocarbon chain containing at least one triple bond. The triple bond may be at any possible position of the hydrocarbon chain. For example,“C2-C6-alkynyl” may refer to ethynyl, propynyl, butynyl, pentynyl and hexynyl. The alkynyl groups may be unsubstituted or substituted by one or more substituents. Specific substituents for any saturated carbon in each alkynyl group independently may be fluorine, ORa or NRaRa.
The term“carbocyclic” refers to a group consisting of one or more rings which are entirely formed from carbon atoms. A carbocylic group can be a mono- or bicyclic cycloalkyl group, or it can comprise at least one phenyl ring. The term“heterocyclic” refers to a group consisting of one or more rings wherein the ring system includes at least one heteroatom. A heterocyclic group may comprise either a heteroaryl or heterocycloalkyl rings. The term“cycloalkyl” refers to a saturated hydrocarbon ring system containing 3, 4, 5 or 6 carbon atoms. For example, “C3-C6-cycloalkyl” may refer to cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The cycloalkyl groups may be unsubstituted or substituted by one or more substituents. Specific substituents for each cycloalkyl group independently may be oxo, C1-C4-alkyl, fluorine, ORa or NHRa.
The term“aromatic” when applied to a substituent as a whole means a single ring or polycyclic ring system with 4n + 2 electrons in a conjugated π system within the ring or ring system where all atoms contributing to the conjugated π system are in the same plane. The term“aryl” refers to an aromatic hydrocarbon ring system. The ring system has 4n +2 electrons in a conjugated π system within a ring where all atoms contributing to the conjugated π system are in the same plane. For example, the“aryl” may be phenyl and naphthyl. Equally, aryl groups may include non-aromatic carbocyclic portions. The aryl group may be unsubstituted or substituted by one or more substituents. Specific substituents for each aryl group independently may be C1-C4-alkyl, C1-C4-haloalkyl, cyano, halogen, ORa or NHRa.
The term“heteroaryl” may refer to any aromatic (i.e. a ring system containing (4n + 2) π- or n- electrons in the π-system) 5-10 membered ring system comprising from 1 to 4 heteroatoms independently selected from O, S and N (in other words from 1 to 4 of the atoms forming the ring system are selected from O, S and N). Thus, any heteroaryl groups may be independently selected from: 5 membered heteroaryl groups in which the heteroaromatic ring is substituted with 1-4 heteroatoms independently selected from O, S and N; and 6-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1-3 (e.g.1-2) nitrogen atoms; 9-membered bicyclic heteroaryl groups in which the heteroaromatic system is substituted with 1-4 heteroatoms independently selected from O, S and N; 10-membered bicyclic heteroaryl groups in which the heteroaromatic system is substituted with 1-4 nitrogen atoms. Specifically, heteroaryl groups may be independently selected from: pyrrole, furan, thiophene, pyrazole, imidazole, oxazole, isoxazole, triazole, oxadiazole, thiadiazole, tetrazole; pyridine, pyridazine, pyrimidine, pyrazine, triazine, indole, isoindole, benzofuran, isobenzofuran, benzothiophene, indazole, benzimidazole, benzoxazole, benzthiazole, benzisoxazole, purine, quinoline, isoquinoline, cinnoline, quinazoline, quinoxaline, pteridine, phthalazine, naphthyridine. Heteroaryl groups may also be 6-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1 heteroatomic group independently selected from O, S and NH and the ring also comprises a carbonyl group. Such groups include pyridones and pyranones. The heteroaryl system itself may be substituted with other groups. The heteroaryl group may be unsubstituted or substituted by one or more substituents. Specific substituents for each heteroaryl group independently may be C1-C4-alkyl, C1-C4-haloalkyl, cyano, halogen, ORa or NHRa.
The term“m-nheterocycloalkyl” may refer to a m- to n-membered monocyclic or bicyclic saturated or partially saturated group comprising 1 or 2 heteroatoms independently selected from O, S and N in the ring system (in other words 1 or 2 of the atoms forming the ring system are selected from O, S and N). By partially saturated it is meant that the ring may comprise one or two double bonds. This applies particularly to monocyclic rings with from 5 to 8 members. The double bond will typically be between two carbon atoms but may be between a carbon atom and a nitrogen atom. Examples of heterocycloalkyl groups include; piperidine, piperazine, morpholine, thiomorpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, dihydrofuran, tetrahydropyran, dihydropyran, dioxane, azepine. Bicyclic systems may be spiro-fused, i.e. where the rings are linked to each other through a single carbon atom; vicinally fused, i.e. where the rings are linked to each other through two adjacent carbon or nitrogen atoms; or they may be share a bridgehead, i.e. the rings are linked to each other through two non-adjacent carbon or nitrogen atoms. The heterocycloalkyl groups may be unsubstituted or substituted by one or more substituents. Specific substituents for each heterocycloalkyl group may independently be oxo, C1-C4- alkyl, fluorine, ORa or NHRa.
The term 3-6heteroalkyl refers to a 3- to 6- membered chain of atoms, wherein at least one of said atoms is a heteroatom, e.g. a nitrogen, and wherein the remainder of said atoms are carbon atoms.
Where the compound of formula (I) is an N-oxide, it will typically be a pyridine N-oxide, i.e. the nitrogen of the pyridine may be N+-O-. Alternatively, it may be that the compound of the invention is not an N-oxide.
An aromatic ring is a phenyl ring. The present invention also includes the synthesis of all pharmaceutically acceptable isotopically-labelled compounds of formulae (I) to (XI) wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number most commonly found in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2H and 3H, carbon, such as 11C, 13C and 14C, chlorine, such as 36Cl, fluorine, such as 18F, iodine, such as 123I and 125I, nitrogen, such as 13N and 15N, oxygen, such as 15O, 17O and 18O, phosphorus, such as 32P, and sulphur, such as 35S. Certain isotopically-labelled compounds, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, i.e.2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labelled compounds can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described using an appropriate isotopically-labelled reagent in place of the non-labelled reagent previously employed. Uses, methods of treatment and pharmaceutical formulations Each of the compounds of the present invention may be used as a medicament. Thus, in another aspect of the invention, there is provided compound as defined above for the treatment of bacterial infections. The compounds and formulations of the present invention may be used in the treatment of a wide range of bacterial infections. In some embodiments, the compounds can be used to treat bacterial infections caused by one or more resistant strains of bacteria. In a further embodiment, the compounds can be used to treat bacterial infections caused by one or more resistant strains of Gram positive bacteria. In a further embodiment, the compounds can be used to treat bacterial infections caused by one or more resistant strains of Gram negative bacteria. The compounds and formulations of the invention may be used to treat infections caused by bacteria which are in the form of a biofilm. The term‘resistant strains’ is intended to mean strains of bacteria which have shown resistance to one or more known antibacterial drug. For example, it may refer to strains which are resistant to methicillin, strains that are resistant to one or more other β-lactam antibiotics, strains that are resistant to one or more fluoroquinolones and/or strains that are resistant to one or more other antibiotics (i.e. antibiotics other than β-lactams and fluoroquinolones). A resistant strain is one in which the MIC of a given compound or class of compounds for that strain has shifted to a significantly higher number than for the parent (susceptible) strain. The compounds of the invention may be particularly effective at treating infections caused by Gram positive bacteria. The compounds of the invention may be particularly effective at treating infections caused by Gram positive bacteria which are resistant to one or more fluoroquinolone antibiotics. The compounds of the invention may be particularly effective at treating infections caused by Gram negative bacteria. The compounds of the invention may be particularly effective at treating infections caused by Gram negative bacteria which are resistant to one or more fluoroquinolone antibiotics. The compounds and formulations of the present invention can be used to treat or to prevent infections caused by bacterial strains associated with biowarfare. These may be strains which are category A pathogens as identified by the US government (e.g. those which cause anthrax, plague etc.) and/or they may be strains which are category B pathogens as identified by the US government (e.g. those which cause Glanders disease, mellioidosis etc). In a specific embodiment, the compounds and formulations of the present invention can be used to treat or to prevent infections caused by Gram positive bacterial strains associated with biowarfare (e.g. anthrax). More particularly, the compounds and formulations may be used to treat category A and/or category B pathogens as defined by the US government on 1st Jan 2015. The compounds of the invention may also be useful in treating other forms of infectious disease, e.g. fungal infections, parasitic infections and/or viral infections. The compounds of the present invention can be used in the treatment of the human body. They may be used in the treatment of the animal body. In particular, the compounds of the present invention can be used to treat commercial animals such as livestock. Alternatively, the compounds of the present invention can be used to treat companion animals such as cats, dogs, etc. The compounds of the invention may be obtained, stored and/or administered in the form of a pharmaceutically acceptable salt. Suitable pharmaceutically acceptable salts include, but are not limited to, salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, malic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, toluenesulphonic, benzenesulphonic, salicylic, sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids. Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts. Hemisalts of acids and bases may also be formed, for example, hemisulfate and hemicalcium salts. Also included are acid addition or base salts wherein the counter ion is optically active, for example, d-lactate or l-lysine, or racemic, for example, dl-tartrate or dl-arginine. Compounds of the invention may exist in a single crystal form or in a mixture of crystal forms or they may be amorphous. Thus, compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, or spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose. For the above-mentioned compounds of the invention the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. For example, if the compound of the invention is administered orally, then the daily dosage of the compound of the invention may be in the range from 0.01 micrograms per kilogram body weight (μg/kg) to 100 milligrams per kilogram body weight (mg/kg).
A compound of the invention, or pharmaceutically acceptable salt thereof, may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the compounds of the invention, or pharmaceutically acceptable salt thereof, is in association with a pharmaceutically acceptable adjuvant, diluent or carrier. Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, "Pharmaceuticals - The Science of Dosage Form Designs", M. E. Aulton, Churchill Livingstone, 1988.
The compounds of the invention may be administered in combination with other active compounds (e.g. antifungal compounds, oncology compounds) and, in particular, with other antibacterial compounds. The compound of the invention and the other active (e.g. the other antibacterial compound) may be administered in different pharmaceutical formulations either simultaneously or sequentially with the other active. Alternatively, the compound of the invention and the other active (e.g. the other antibacterial compound) may form part of the same pharmaceutical formulation.
Examples of other bacterial compounds which could be administered with the compounds of the invention are penems, carbapenems, fluoroquinolones, β-lactams, vancomycin, erythromycin or any other known antibiotic drug molecule. Depending on the mode of administration of the compounds of the invention, the pharmaceutical composition which is used to administer the compounds of the invention will preferably comprise from 0.05 to 99 %w (per cent by weight) compounds of the invention, more preferably from 0.05 to 80 %w compounds of the invention, still more preferably from 0.10 to 70 %w compounds of the invention, and even more preferably from 0.10 to 50 %w compounds of the invention, all percentages by weight being based on total composition. The pharmaceutical compositions may be administered topically (e.g. to the skin) in the form, e.g., of creams, gels, lotions, solutions, suspensions, or systemically, e.g. by oral administration in the form of tablets, capsules, syrups, powders, suspensions, solutions or granules; or by parenteral administration in the form of a sterile solution, suspension or emulsion for injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion); or by rectal administration in the form of suppositories; or by inhalation (i.e. in the form of an aerosol or by nebulisation).
If administered topically, high-dosages of the compounds of the invention can be administered. Thus, a compound with an in vitro MIC of, for example, 16-64 μg/mL may still provide an effective treatment against certain bacterial infections.
For oral administration the compounds of the invention may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets. If coated tablets are required, the cores, prepared as described above, may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide. Alternatively, the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent.
For the preparation of soft gelatine capsules, the compounds of the invention may be admixed with, for example, a vegetable oil or polyethylene glycol. Hard gelatine capsules may contain granules of the compound using either the above-mentioned excipients for tablets. Also liquid or semisolid formulations of the compound of the invention may be filled into hard gelatine capsules. Liquid preparations for oral application may be in the form of syrups or suspensions, for example, solutions containing the compound of the invention, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol. Optionally such liquid preparations may contain colouring agents, flavouring agents, sweetening agents (such as saccharine), preservative agents and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in the art.
For intravenous (parenteral) administration the compounds of the invention may be administered as a sterile aqueous or oily solution.
The size of the dose for therapeutic purposes of compounds of the invention will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well known principles of medicine. Dosage levels, dose frequency, and treatment durations of compounds of the invention are expected to differ depending on the formulation and clinical indication, age, and co-morbid medical conditions of the patient. The standard duration of treatment with compounds of the invention is expected to vary between one and seven days for most clinical indications. It may be necessary to extend the duration of treatment beyond seven days in instances of recurrent infections or infections associated with tissues or implanted materials to which there is poor blood supply including bones/joints, respiratory tract, endocardium, and dental tissues.
In another aspect the present invention provides a pharmaceutical formulation comprising a compound of the invention and a pharmaceutically acceptable excipient. The formulation may further comprise one or more other antibiotics, e.g. one or more fluoroquinolone antibiotics. Illustrative fluoroquinolone antibiotics include levofloxacin, enoxacin, fleroxacin, lomefloxacin, nadifloxacin, norfloxacin, rufloxacin, balofloxacin, grepafloxacin, pazufloxacin, sparfloxacin, temafloxacin, tosufloxacin, besifloxacin, clinafloxacin, garenoxacin, gemifloxacin, gatifloxacin, sitafloxacin, trovafloxacin, prulifloxacin, ciprofloxacin, pefloxacin, moxifloxacin, ofloxacin, delafloxacin, zabofloxacin, avarofloxacin, finafloxacin. In another aspect of the invention is provided a method of treating a bacterial infection, the method comprising treating a subject in need thereof with a therapeutically effective amount of a compound of the invention. Medical uses The compounds of the present invention can be used in the treatment of the human body. The compounds of the invention may be for use in treating human bacterial infections such as infections of the genitourinary system, the respiratory tract, the gastrointestinal tract, the ear, the skin, the throat, soft tissue, bone and joints (including infections caused by Staphylococcus aureus). The compounds can be used to treat pneumonia, sinusitis, acute bacterial sinusitis, bronchitis, acute bacterial exacerbation of chronic bronchitis, anthrax, chronic bacterial prostatitis, acute pyelonephritis, pharyngitis, mycobacterial infections (e.g. tuberculosis or leprosy), tonsillitis, Escherichia coli, prophylaxis before dental surgery, cellulitis, acnes, cystitis, infectious diarrhoea, typhoid fever, infections caused by anaerobic bacteria, peritonitis, abdominal infection, bacteraemia, septicaemia, leprosy, sexually transmitted bacterial infection (e.g. gonorrhoea, Chlamydia), Neisseria infection (e.g. gonorrhoea, meningitis) or other fastidious Gram negative infection, bacterial vaginosis, pelvic inflammatory disease, pseudomembranous colitis, Helicobacter pylori, acute gingivitis, Crohn's disease, rosacea, fungating tumours, impetigo. The compounds of the present invention may also be used in treating other conditions treatable by eliminating or reducing a bacterial infection. In this case they will act in a secondary manner alongside for example a chemotherapeutic agent used in the treatment of cancer. In yet another aspect of the invention is provided a compound for use in the preparation of a medicament. The medicament may be for use in the treatment of any of the diseases, infections and indications mentioned in this specification. In an aspect of the invention is provided a compound of the invention for medical use. The compound may be used in the treatment of any of the diseases, infections and indications mentioned in this specification. Veterinary uses They may be used in the treatment of the animal body. In particular, the compounds of the present invention can be used to treat commercial animals such as livestock. The livestock may be mammal (excluding humans) e.g. cows, pigs, goats, sheep, llamas, alpacas, camels and rabbits. The livestock may be birds (e.g. chickens, turkeys, ducks, geese etc.). Alternatively, the compounds of the present invention can be used to treat companion animals such as cats, dogs, etc. The veterinary use may be to treat wild populations of animals in order to prevent the spread of disease to humans or to commercial animals. In this case, the animals may be rats, badgers, deer, foxes, wolves, mice, kangaroos and monkeys and other apes. In an aspect of the invention is provided a compound of the invention for veterinary use. The compound may be used in the treatment of any of the animal diseases and infections and indications mentioned in this specification. In another aspect the present invention provides a veterinary formulation comprising a compound of the invention and a veterinarily acceptable excipient. The methods by which the compounds may be administered for veterinary use include oral administration by capsule, bolus, tablet or drench, topical administration as an ointment, a pour-on, spot-on, dip, spray, mousse, shampoo, collar or powder formulation or, alternatively, they can be administered by injection (e.g. subcutaneously, intramuscularly or intravenously), or as an implant. Such formulations may be prepared in a conventional manner in accordance with standard veterinary practice. The formulations will vary with regard to the weight of active compound contained therein, depending on the species of animal to be treated, the severity and type of infection and the body weight of the animal. For parenteral, topical and oral administration, typical dose ranges of the active ingredient are 0.01 to 100 mg per kg of body weight of the animal. Preferably the range is 0.1 to 10 mg per kg. In any event, the veterinary practitioner, or the skilled person, will be able to determine the actual dosage which will be most suitable for an individual patient, which may vary with the species, age, weight and response of the particular patient. The above dosages are exemplary of the average case; there can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention. As an alternative, when treating animals the compounds may be administered with the animal feedstuff and for this purpose a concentrated feed additive or premix may be prepared for mixing with the normal animal feed. Certain compounds of the invention may be used in the treatment of mastitis. In this regard, a particularly preferred method of administration is by injection into the udder of a subject (e.g. a cow, a goat, a pig or sheep). Throughout the description and claims of this specification, the words“comprise” and “contain” and variations of the words, for example“comprising” and“comprises”, means “including but not limited to”, and is not intended to (and does not) exclude other moieties, additives, components, integers or steps. Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise. Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith.
Synthesis The skilled man will appreciate that adaptation of methods known in the art could be applied in the manufacture of the compounds of the present invention. For example, the skilled person will be immediately familiar with standard textbooks such as "Comprehensive Organic Transformations - A Guide to Functional Group Transformations", RC Larock, Wiley-VCH (1999 or later editions), "March's Advanced Organic Chemistry - Reactions, Mechanisms and Structure”, MB Smith, J. March, Wiley, (5th edition or later) “Advanced Organic Chemistry, Part B, Reactions and Synthesis”, FA Carey, RJ Sundberg, Kluwer Academic/Plenum Publications, (2001 or later editions), "Organic Synthesis - The Disconnection Approach", S Warren (Wiley), (1982 or later editions), "Designing Organic Syntheses" S Warren (Wiley) (1983 or later editions), "Guidebook To Organic Synthesis" RK Mackie and DM Smith (Longman) (1982 or later editions), etc., and the references therein as a guide. The skilled chemist will exercise his judgement and skill as to the most efficient sequence of reactions for synthesis of a given target compound and will employ protecting groups as necessary. This will depend inter alia on factors such as the nature of other functional groups present in a particular substrate. Clearly, the type of chemistry involved will influence the choice of reagent that is used in the said synthetic steps, the need, and type, of protecting groups that are employed, and the sequence for accomplishing the protection / deprotection steps. These and other reaction parameters will be evident to the skilled person by reference to standard textbooks and to the examples provided herein. Sensitive functional groups may need to be protected and deprotected during synthesis of a compound of the invention. This may be achieved by conventional methods, for example as described in“Protective Groups in Organic Synthesis” by TW Greene and PGM Wuts, John Wiley & Sons Inc (1999), and references therein. Throughout this specification these abbreviations have the following meanings:
Ac = acetyl ACN = acetonitrile
aq. = aqueous Bn = benzyl
BOC = tert-butyloxycarbonyl CBZ = carboxybenzyl
DCM = Dichloromethane DIPEA = diisopropylethylamine
DMF = N, N-dimethylformamide
DMSO = dimethyl sulfoxide HATU = 1-[Bis(dimethylamino)methylene]-1H- 1,2,3-triazolo[4,5-b]pyridinium-3-oxide hexafluorophosphate
HBTU = N,N,N′,N′-Tetramethyl-O-(1H-benzotriazol-1-yl)uronium hexafluorophosphate NMP = N-methylpyrrolidinone TFA = trifluoroacetic acid
THF = tetrahydrofuran TMSCl = trimethylsilyl chloride
TMSI = trimethylsilyl iodide TBTU = N,N,N′,N′-Tetramethyl-O-(1H- benzotriazol-1-yl)uronium tetrafluoroborate Certain compounds of the invention can be made according to the following general synthetic schemes. Certain compounds of the invention can be made according to or analogously to the methods described in the Examples. General Synthetic Schemes
Certain compounds of formula (IV) and (VII) can be made by Scheme A:
Figure imgf000040_0001
Scheme A
Reaction of pyridone (1) with commercially available 2-bromo-1,1-diethoxyethane (2), followed by hydrolysis of the acetal can generate aldehyde (3). The alkylation reaction can be carried out in the presence of a base, such as Cs2CO3, in a solvent, such as dry NMP, at a temperature from 50-100oC. Hydrolysis can be effected using a strong acid, such as concentrated HCl, in a solvent, such as ACN, at room temperature. Aldehyde (3) can be converted to ester (5) via reductive amination with the known amine (4), prepared as described in EP154142. The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. The ester group in (5) can be converted to the alcohol (6) using a reducing agent, such as LiAlH4 or BHEt3 +Li- (super hydride), in a solvent, such as THF, at a temperature from 0oC to room temperature. Allylation of alcohol (6) with allyl bromide in the presence of a base, such as NaH, in a solvent, such as THF or DMF, at a temperature from 0oC to 60oC, can furnish diene (7). Addition of quaternary ammonium salts is optional. RCM (ring controlled metathesis) of the diene (7) to macrocycle alkene (8) can be accomplished in the presence of Grubbs catalyst (first or second generation) in a suitable solvent, such as toluene, at a temperature from room temperature to 90oC. The RCM reaction may provide entirely the E isomer, entirely the Z isomer or a mixture thereof. Deprotection of the cyclic ketal in (8) to release the ketone in (9) can be effected using acid catalysed conditions, such as use of a protic acid (10% HCl in a mixture of THF/H2O) or a Lewis acid (FeCl3 dispersed on silica). Ketone (9) can be converted to amine (11) via reductive amination with amine (10). The reaction can be performed using a borohydride reagent, such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80oC. Addition of AcOH as a catalyst is optional. Amine (11) can be converted to (13) (a subset of compounds of both formula (IV) and formula (VII)) under reductive amination conditions with aldehyde (12). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. Amine (11) can be converted to (15) (another subset of compounds of both formula (IV) and formula (VII)) by coupling with (14), where LG represents a leaving group, such as a halide. The reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating. Certain other compounds of both formula (IV) and formula (VI) can be made by Scheme B:
Figure imgf000041_0001
Scheme B The alkene double bond in ketone (9) in Scheme A can be hydrogenated to (16) by the action of H2 in the presence of a Pd/C catalyst in an alcoholic solvent, such as EtOH, at room temperature. Ketone (16) can then be transformed into (17) and (18) (two further subsets of compounds of formulae (IV) and (VII)) using an analogous series of transformations to those described in Scheme A for the conversion of ketone (9) to (13) and (15). Certain compounds of formula (III) and (VII) can be made following Scheme A but using the ketal (19), prepared as described in US 6645980, in place of the amine (4). Macrocycles (20) and (21) (two subsets of compounds of formulae (III) and (VII)) can be prepared using an analogous series of transformations to those described in Scheme A for the conversion of ester (5) to (13) and (15).
Figure imgf000042_0001
Following Scheme A but using the ketal (24) in place of amine (4), macrocycles (25) and (26) (two further subsets of compounds of formulae (IV) and (VII)) can be prepared using an analogous series of transformations to those described in Scheme A for the conversion of ester (5) to (13) and (15).
Figure imgf000043_0001
Ketal (24) can be formed by Scheme C from commercially available ketone (27) under Dean Stark conditions using ethylene glycol and catalytic p-TsOH (10%) in toluene.
Figure imgf000043_0002
Scheme C Following Scheme B macrocycles (28) and (29) (two further subsets of compounds of formulae (IV) and (VII)) can be prepared.
Figure imgf000043_0003
Certain compounds of formula (VI) and (VII) can be made by Scheme D:
Figure imgf000044_0001
(36)
Scheme D
Reductive ammination of aldehyde (3) (prepared as described in Scheme A) with commercially available hydroxy amine (30) can form alcohol (31). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. Allylation of alcohol (31) with allyl bromide in the presence of a base, such as NaH, in a solvent, such as THF or DMF, at a temperature from 0oC to 60oC, can furnish diene (32). RCM (ring closing metathesis) of the diene (32) to macrocycle alkene (33) can be accomplished in the presence of Grubbs catalyst (first or second generation) in a suitable solvent, such as toluene, at a temperature from room temperature to 90oC. The RCM reaction may provide entirely the E isomer, entirely the Z isomer or a mixture thereof. Deprotection of the nitrogen BOC protecting group in (33) can be performed under standard conditions, such as by the action of TFA in DCM at room temperature. Amine (34) can be converted to (35) (a subset of compounds of both formula (VI) and formula (VII)) by reductive amination with aldehyde (12). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. Amine (34) can be converted to (36) (another subset of compounds of both formula (VI) and formula (VII)) by coupling with (14), where LG represents a leaving group, such as a halide. The reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating. Certain other compounds of both formula (VI) and formula (VII) can be made by Scheme E
Figure imgf000045_0001
Scheme E The alkene double bond in amine (34) in Scheme D can be hydrogenated to (37) by the action of H2 in the presence of a Pd/C catalyst in an alcoholic solvent, such as EtOH, at room temperature. Amine (37) can then be transformed into (38) and (39) (two further subsets of compounds of formulae (VI) and (VII)) using an analogous series of transformations to those described in Scheme D for the conversion of amine (34) to (35) and (36).
Certain compounds of formula (V) can be made by Scheme F: H
Figure imgf000046_0001
5
Scheme F Amine (42) can be formed through reaction of commercially available bromo ester (40) with commercially available cyclic acetal protected amine (41). The reaction can be performed in the presence of a base, such as K2CO3 or Et3N, in a solvent, such as ACN or DCM, at a temperature from 0oC to room temperature. Aldehyde (3) (prepared as described in Scheme A) can be converted to cyclic acetal (43) via reductive amination with the amine (42). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. The ester group in cyclic acetal (43) can be converted to the alcohol (44) using a reducing agent, such as LiAlH4 or BHEt3+Li- (super hydride), in a solvent, such as THF, at a temperature from 0oC to room temperature. Allylation of alcohol (44) with allyl bromide in the presence of a base, such as NaH, in a solvent, such as THF or DMF, at a temperature from 0oC to 60oC, can furnish diene (45). Addition of quaternary ammonium salts is optional. RCM (ring controlled metathesis) of the diene (45) to macrocycle alkene (46) can be accomplished in the presence of Grubbs catalyst (first or second generation) in a suitable solvent, such as toluene, at a temperature from room temperature to 90oC. The RCM reaction may provide entirely the E isomer, entirely the Z isomer or a mixture thereof. Deprotection of the cyclic acetal in (46) to release the aldehyde in (47) can be effected using acid catalysed conditions, such as use of a protic acid (10% HCl in a mixture of THF/H2O) or a Lewis acid (FeCl3 dispersed on silica). Aldehyde (47) can be converted to amine (48) via reductive amination with amine (10). The reaction can be performed using a borohydride reagent, such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of AcOH as a catalyst is optional. Amine (48) can be converted to (49) (a subset of compounds of formula (V)) under reductive amination conditions with aldehyde (12). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. Amine (48) can be converted to (50) (another subset of compounds of formula (V)) by coupling with (14), where LG represents a leaving group, such as a halide. The reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating. Certain other compounds of formula (V) can be made by Scheme G:
Figure imgf000047_0001
Scheme G The alkene double bond in amine (48) in Scheme G can be hydrogenated to (51) by the action of H2 in the presence of a Pd/C catalyst in an alcoholic solvent, such as EtOH, at room temperature. Amine (51) can then be transformed into (52) and (53) (two further subsets of compounds of formula (V) using an analogous series of transformations to those described in Scheme F for the conversion of amine (48) to (49) and (50). Following Scheme F, but using aldehyde (54) in place of (3), macrocycles (55) and (56) (a subset of compounds of formula (V)) can be prepared using an analogous series of transformation to those described in Scheme F for the conversion of aldehyde (3) to (49) and (50).
Figure imgf000048_0001
Aldehyde (54) can be formed by Scheme H from reaction of (1) with commercially available 3-bromo-1,1-dimethoxypropane (57), followed by hydrolysis of the acetal. The alkylation reaction can be carried out in the presence of a base, such as Cs2CO3, in a solvent, such as dry NMP, at a temperature from 50-100oC. Hydrolysis of the acetal can be effected using a strong acid, such as concentrated HCl, in a solvent, such as ACN, at room temperature.
Figure imgf000048_0002
Scheme H Following Scheme G macrocycles (58) and (59) (two further subsets of compounds of formulae (V)) can be prepared
Figure imgf000049_0001
Scheme I Reductive amination of aldehyde (3) (prepared as described in Scheme A) with amine (60) can form diene (61). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. RCM (ring closing metathesis) of the diene (61) to macrocycle alkene (62) can be accomplished in the presence of Grubbs catalyst (first or second generation) in a suitable solvent, such as toluene, at a temperature from room temperature to 90oC. The RCM reaction may provide entirely the E isomer, entirely the Z isomer or a mixture thereof. Deprotection of the nitrogen CBZ protecting group in (62) can be performed using base, such as KOH, in a solvent mixture of H2O and EtOH, at a temperature from 60oC to reflux or refluxing in neat TFA or treatment with TMSI, in a solvent, such as DCM, at a temperature from room temperature to reflux. Amine (63) can be converted to (64) (a subset of compounds of both formula (III) and formula (VII)) by reduction amination with aldehyde (12). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. Amine (63) can be converted to (65) (another subset of compounds of both formula (III) and formula (VII)) by coupling with (14), where LG represents a leaving group, such as a halide. The reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating. Amine (60) can be formed by Scheme J:
Figure imgf000050_0001
Scheme J Allylation of hydroxyl (66), prepared as described in WO2000/47207, can form alkene (67). The reaction can be performed with allyl bromide in the presence of a base, such as NaH, in a solvent, such as THF or DMF, at a temperature from 0oC to 60oC. Deprotection of the nitrogen BOC protecting group in (67) to give amine (60) can be performed under standard conditions, such as by the action of TFA in DCM at room temperature. Certain other compounds of formula (III) and (VII) can be made by Scheme K:
Figure imgf000051_0001
Scheme K Hydrogenation of the alkene double bond and removal of nitrogen CBZ protecting in (62) in Scheme I to give amine (68) can be effected by the action of H2 in the presence of a Pd/C catalyst in an alcoholic solvent, such as EtOH, at room temperature. Amine (68) can then be transformed into (69) and (70) (two further subsets of compounds of both formulae (III) and (VII)) using an analogous series of transformations to those described in Scheme I for the conversion of amine (63) to (64) and (65). Certain compounds of formulae (III) and (VII) can be made by Scheme L:
Figure imgf000052_0001
Figure imgf000052_0002
Scheme L Reaction of pyridone (71) with 2-bromo-1,1-diethoxyethane (2), followed by hydrolysis of the acetal can generate aldehyde (72). The alkylation reaction can be carried out in the presence of a base, such as Cs2CO3, in a solvent, such as dry NMP, at a temperature from 50-100oC. Hydrolysis can be effected using a strong acid, such as concentrated HCl, in a solvent, such as ACN, at room temperature. Aldehyde (72) can be converted to ester (73) via reductive amination with the known amine (19), prepared as described in US 6645980. The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. The ester group in (73) can be converted to the alcohol (74) using a reducing agent, such as LiAlH4 or BHEt3+Li- (super hydride), in a solvent, such as THF, at a temperature from 0oC to room temperature. Alcohol (74) can be converted to azide (75) by a twostep process involving treatment with paraformaldehyde in the presence of TMSCl at room temperature, followed by treatment with an azide reagent, such as NaN3, in a solvent, such as THF, at room temperature, in the presence of 18-Crown-6. A 3+2 cycloaddition of the azide onto the alkyne in (75) to deliver macrocycle alkene (76) can be accomplished using pentamethylcyclopentadienyl ruthenium chloride [Cp*RuCl] complexes in a suitable solvent, such as toluene or dioxane, at a temperature from room temperature to 60oC. Deprotection of the cyclic acetal in (76) to release the ketone in (77) can be effected using acid catalysed conditions, such as use of a protic acid (10% HCl in a mixture of THF/H2O) or a Lewis acid (FeCl3 dispersed on silica). Ketone (77) can be converted to amine (78) via reductive amination with amine (10). The reaction can be performed using a borohydride reagent, such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of AcOH as a catalyst is optional. Amine (78) can be converted to (79) (a subset of compounds of formulae (III) and (VII)) under reductive amination conditions with aldehyde (12). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. Amine (78) can be converted to (80) (another subset of compounds of formulae (III) and (VII)) by coupling with (14), where LG represents a leaving group, such as a halide. The reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating. Following Scheme L, but using Cu(I) catalytic systems in the 3+2 cycloaddition reaction of the azide onto the alkyne in (75), macrocycles (81) and (82) (two further subsets of compounds of formulae (III) and (VII)) can be prepared. The reaction can be carried out in a solvent, such as H2O or an alcohol, such as EtOH, at a temperature from room temperature to 80oC.
Figure imgf000053_0001
Certain compounds of formulae (III) and (VII) can be made following Scheme I but using trans amine (83) in place of amine (60). Macrocycles (84) and (85) (two subsets of compounds of formulae (III) and (VII)) can be prepared using an analogous series of transformations to those described in Scheme I for the conversion of (61) to (64) and (65).
Figure imgf000054_0001
Trans amine (83) can be formed by Scheme M:
Figure imgf000054_0002
Scheme M Allylation of trans hydroxyl (86), prepared as described in WO2006/002047, can form alkene (87). The reaction can be performed with allyl bromide in the presence of a base, such as NaH, in a solvent, such as THF or DMF, at a temperature from 0oC to 60oC. Deprotection of the nitrogen BOC protecting group in (87) to amine (83) can be performed under standard conditions, such as by the action of TFA in DCM at room temperature. Following Scheme K macrocycles (88) and (89) (two further subsets of compounds of formulae (III) and (VII)) can be prepared.
Figure imgf000055_0001
Following Scheme M but using cis hydroxyl (90), prepared as described in WO2006/002047, cis amine (91) can be prepared.
Figure imgf000055_0002
Certain compounds of formulae (III) and (VII) can be made following Scheme I but using cis amine (91) in place of amine (60). Macrocycles (92) and (93) (two subsets of compounds of formulae (III) and (VII)) can be prepared using an analogous series of transformations to those described in Scheme I for the conversion of (61) to (64) and (65).
Figure imgf000055_0003
Following Scheme K macrocycles (94) and (95) (two further subsets of compounds of formulae (III) and (VII)) can be prepared.
Figure imgf000056_0001
Certain compounds of formula (III) and (VII) can be made following Scheme N:
Figure imgf000057_0001
Scheme N Alkylation of phenol (96) with commercially available benzyl N-(3-bromopropyl)carbamate (97) can form carbamate (98). The reaction can be performed in the presence of a base, such as K2CO3, in a solvent such as acetone or EtOH, at a temperature from room temperature to 70oC. Reaction of carbamate (98) with commercially available 2-bromo-1,1- diethoxyethane (2), followed by hydrolysis of the acetal can generate aldehyde (99). The alkylation reaction can be carried out in the presence of a base, such as Cs2CO3, in a solvent, such as dry NMP, at a temperature from 50-100oC. Hydrolysis can be effected using a strong acid, such as concentrated HCl, in a solvent, such as ACN, at room temperature. Aldehyde (99) can be converted to ester (100) via reductive amination with the known amine (19), prepared as described in US 6645980. The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2- dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. The ester group in (100) can be converted to carboxylic acid (101) using base hydrolysis, such as Na2CO3 in a solvent mixture of H2O/EtOH, at a temperature from 50oC to reflux or LiOH in a solvent mixture of H2O/THF, at a temperature from 50oC to 80oC or Et3N in H2O at a temperature from room temperature to 50oC. Deprotection of the nitrogen CBZ group to give amine (102) can be achieved under standard conditions, such as use of H2 in the presence of Pd/C in an alcoholic solvent, such as EtOH, at room temperature. Macrolactamisation of (102) can be effected under standard amide coupling conditions, such as HBTU, in the presence of a base, such as Et3N, in a solvent, such as DMSO, at room temperature. Deprotection of the cyclic ketal in macrocycle (103) to release the ketone in (104) can be effected using acid catalysed conditions, such as use of a protic acid (10% HCl in a mixture of THF/H2O) or a Lewis acid (FeCl3 dispersed on silica). Ketone (104) can be converted to amine (105) via reductive amination with amine (10). The reaction can be performed using a borohydride reagent, such as sodium triacetoxyborohydride in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of AcOH as a catalyst is optional. Amine (105) can be converted to (106) (a subset of compounds of both formula (III) and formula (VII)) under reductive amination conditions with aldehyde (12). The reaction can be performed using a borohydride reagent, such as tetramethylammonium triacetoxyborohydride or sodium triacetoxyborohydride, in a solvent, such as THF or 1,2-dichloroethane, at a temperature from room temperature to 80oC. Addition of 4Å sieves is optional. Amine (105) can be converted to (107) (another subset of compounds of both formula (III) and formula (VII)) by coupling with (14), where LG represents a leaving group, such as a halide. The reaction can be performed in a solvent, such as DMF or THF, in the presence of a base, such as K2CO3, with optional heating. Experimental NMR spectra were obtained on a LC Bruker AV400 using a 5 mm QNP probe (Method A) or Bruker AV1500MHz with 5mm QNP probe and Z-axis gradients (Method B). MS was carried out on a Waters Alliance ZQ MS (Methods A, B, C and D) using H2O and ACN mobile phase with pH modification as detailed under each method. Wavelengths were 254 and 210 nm. Method A (Acidic pH) Column: YMC-Triart C1850 x 2 mm, 5 ^m. Flow rate: 0.8 mL/min. Injection volume: 5 μL. Mobile Phase A H2O B ACN C 50% H2O / 50% ACN + 1.0% formic acid
Figure imgf000059_0001
Method B (Acidic pH) Column: YMC Triart-C1850 x 2 mm, 5 ^m Flow rate: 0.8 mL/min. Injection volume: 5 μL
Figure imgf000059_0002
Figure imgf000060_0002
Method C (Basic pH)
Column: YMC-Triart C1850 x 2 mm, 5 ^m. Flow rate: 0.8 mL/min. Injection volume: 5 μL. Mobile Phase A H2O
B ACN
C 50% H2O / 50% ACN + 1.0% ammonia
Figure imgf000060_0001
Method D (Basic pH)
Column YMC Triart-C1850 x 2 mm, 5 µm Flow rate: 0.8 mL/min. Injection volume: 10 μL Mobile Phase A H2O
B ACN
C 50% H2O / 50% ACN + 1.0% NH3
Figure imgf000060_0003
Figure imgf000061_0002
Preparative HPLC was performed using column: XBridgeTM prep C18 5 μM OBD 19 x 100 mm. Flow rate: 20 mL/min. Method A
Waters 3100 Mass detector using H2O and CH3CN + formic acid (0.05%-0.1%)
Method B
Waters 2767 Sample Manager (0.05% NH3) Intermediate A: 2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]acetaldehyde (a) 7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-2-one
Figure imgf000061_0001
A mixture of 7-hydroxy-1,2-dihydroquinolin-2-one (4.0 g, 24.82 mmol), K2CO3 (5.15 g, 37.23 mmol) and allyl bromide (2.6 mL, 29.78 mmol) in acetone (40 mL) was heated at 60oC for 16 h. After cooling to room temperature the mixture was filtered and the solid collected was washed with acetone (2 x 15 mL). The filtrates were combined and the solvent removed in vacuo. The resulting residual solid was triturated with MeOH to furnish 7-(prop- 2-en-1-yloxy)-1,2-dihydroquinolin-2-one (3.7 g, 74 % yield) as a white solid. LC-MS (Method C) 202.4 [M+H]+; RT 2.14 min (b) 2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]acetaldehyde A
Figure imgf000062_0001
To a mixture of 7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-2-one (28.5 g, 141.64 mmol) and Cs2CO3 (55.4 g, 167.97 mmol) in NMP (230 mL) was added 2-bromo-1,1-diethoxyethane (23.44 mL, 155.8 mmol) and the suspension heated at 100oC for 16 h. On cooling H2O (500 mL) was added and the mixture was extracted with EtOAc (3 x 350 mL). The combined organics were washed with H2O and brine, dried (MgSO4) and concentrated in vacuo. The residue was taken up in THF (150 mL) and 2M HCl (150 mL) and heated to 55oC for 2 h. On cooling the reaction mixture was extracted with EtOAc (200 mL x 3) and the combined organics were washed with H2O and brine, dried (MgSO4) and concentrated in vacuo. The resulting residue was triturated with petroleum ether then Et2O and dried to give 2-[2-oxo-7- (prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]acetaldehyde (10.4 g, 30 % yield).
Intermediate B: 2-[7-(but-3-en-1-yloxy)-2-oxo-1,2-dihydroquinolin-1-yl]acetaldehyde (a) 7-(but-3-en-1-yloxy)-1,2-dihydroquinolin-2-one
Figure imgf000062_0002
A mixture of 7-hydroxy-1,2-dihydroquinolin-2-one (25.0 g, 155.13 mmol), 4-bromobut-1-ene (16.53 mL, 162.88 mmol) and Cs2CO3 (75.81 g, 232.69 mmol) was heated at 150oC for 16 h in NMP (60 mL). On cooling the reaction mixture was poured into H2O (1 L). The resulting solids were filtered, washed with H2O and dried to give 7-(but-3-en-1-yloxy)-1,2- dihydroquinolin-2-one (11.0 g), which was used without further purification. LC-MS (Method C ) 216.4 [M+H]+; RT 2.35 min (b) 2-[7-(but-3-en-1-yloxy)-2-oxo-1,2-dihydroquinolin-1-yl]acetaldehyde B
Figure imgf000063_0001
To a mixture of 7-(but-3-en-1-yloxy)-1,2-dihydroquinolin-2-one (8.31 g, 38.61 mmol) and Cs2CO3 (15.1 g, 46.34 mmol) in NMP (80 mL) was added 2-bromo-1,1-diethoxyethane (6.39 mL, 42.48 mmol) and the suspension was heated at 100oC for 32 h. On cooling H2O (250 mL) was added and the mixture was extracted with EtOAc (3 x 180 mL). The combined organics were concentrated in vacuo then dissolved in THF (70 mL) and 2M HCl (70 mL) and the mixture was heated to 55oC for 2 h. On cooling the mixture was extracted with EtOAc (3 x 70 mL) and the combined organics washed with H2O and brine, dried (MgSO4) and concentrated in vacuo. The resulting residue was triturated with petroleum ether (40-60), which resulted in a brown solid being formed. The solid was collected and purified by flash column chromatography eluting with 25-100% EtOAc in petroleum ether (40-60) to give 2-[7-(but-3-en-1-yloxy)-2-oxo-1,2-dihydroquinolin-1-yl]acetaldehyde (2.5 g, 25 % yield) as a yellow solid. Example 1: (±) trans-6-[({3-oxo-2H, 3H, 4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]- 8,13-dioxa-1,4-diazatetracyclo[12.6.2.14,7.017,21]tricosa-14(22),15,17 (21),18–tetraen-20- one (a) (±) tert-butyl trans-3-([(benzyloxy)carbonyl]amino)-4-hydroxy-pyrrolidine-1- carboxylate 1a
Figure imgf000063_0002
1a To a vigorously stirred solution of (±) tert-butyl trans-3-amino-4-hydroxy-pyrrolidine-1- carboxylate (6.57 g, 32.49 mmol) and Na2CO3 (4.13 g, 38.98 mmol) in a mixture of H2O (75 mL) and 1,4-dioxane (40 mL) at 0oC was added dropwise a solution of benzyl chloroformate (5.6 mL, 39.0 mmol) in 1,4-dioxane. The reaction was allowed to warm to room temperature and stirred overnight. The mixture was extracted with EtOAc (3 x 100 mL) and the combined organics were washed with brine, dried (MgSO4) and concentrated in vacuo to give (±) tert-butyl trans-3-([(benzyloxy)carbonyl]amino)-4-hydroxy-pyrrolidine-1- carboxylate 1a (11.7 g) as a crude brown oil, which was used without further purification. (b) (±) tert-butyl trans-3-([(benzyloxy)carbonyl]amino)-4-(prop-2-en-1-yloxy)-pyrrolidine- 1-carboxylate 1b
Figure imgf000064_0001
A solution of (±) tert-butyl trans-3-([(benzyloxy)carbonyl]amino)-4-hydroxy-pyrrolidine-1- carboxylate 1a (1.50 g, 4.46 mmol) in THF (40 mL) at 0oC was treated with NaH (60% dispersion in mineral oil) (196 mg, 4.91 mmol). The reaction mixture was allowed to warm to room temperature, stirred for a further 20 min and treated dropwise with a solution of allyl bromide (0.46 mL, 5.35 mmol) in THF (5 mL). After stirring at room temperature overnight the reaction mixture was partitioned between Et2O and H2O. The aqueous phase was further extracted with Et2O. The combined organics were dried (MgSO4) and concentrated in vacuo to give a colourless gum, which was purified by flash column chromatography using a gradient eluent system of 30-100% Et2O in petroleum ether (40-60) to afford (±) tert-butyl trans-3-([(benzyloxy)carbonyl]amino)-4-(prop-2-en-1-yloxy)-pyrrolidine-1- carboxylate 1b (800 mg, 48 % yield). (c) (±) benzyl N-[trans-4-(prop-2-en-1-yloxy)pyrrolidin-3-yl]carbamate 1c
Figure imgf000065_0001
A solution of (±) tert-butyl trans-3-([(benzyloxy)carbonyl]amino)-4-(prop-2-en-1-yloxy)- pyrrolidine-1-carboxylate 1b (2.54 g, 6.75 mmol) in DCM (100 mL) was treated with TFA (20.7 mL, 269.9 mmol). After stirring at room temperature for 2 h the reaction mixture was concentrated in vacuo and the resulting residue was partitioned between EtOAc and saturated aqueous K2CO3 solution. The organics were separated, dried (MgSO4) and concentrated in vacuo to give (±) benzyl N-[trans-4-(prop-2-en-1-yloxy)pyrrolidin-3- yl]carbamate 1c (1.48 g, 79 % yield) as a colourless gum. (d) (±) benzyl N-(trans-1-(2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]ethyl)- 4-(prop-2-en-1-yloxy)pyrrolidin-3-yl)carbamate 1d
Figure imgf000065_0002
A solution of 2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]acetaldehyde Intermediate A (233 mg, 0.96 mmol) and (±) benzyl N-[trans-4-(prop-2-en-1- yloxy)pyrrolidin-3-yl]carbamate 1c (240 mg, 0.87 mmol) in DCM (10 mL) was stirred over pre-activated 3A molecular sieves. After 5 min sodium triacetoxyborohydride (263 mg, 1.24 mmol) was added and the reaction further stirred at room temperature for 2 h. Saturated aqueous NaHCO3 solution was added and the DCM layer was separated. The aqueous was further extracted with DCM. The combined organics were dried (MgSO4) and concentrated in vacuo to give a crude oil. The oil was purified by flash column chromatography using a gradient eluent system of 0-100% EtOAc in petroleum ether (40- 60) to give (±)-benzyl N-(trans-1-(2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1- yl]ethyl)-4-(prop-2-en-1-yloxy)pyrrolidin-3-yl)carbamate 1d (165 mg, 38% yield) as a colourless oil. LC-MS (Method C) 504.5 [M+H]+; RT 3.32 min (e) (±) benzyl N-[trans-(10E,Z)-20-oxo-8,13-dioxa-1,4- diazatetracyclo[12.6.2.14,7.017,21]tricosa-10,14(22),15,17(21),18-pentaen-6-yl]carbamate 1e
Figure imgf000066_0001
1d 1e
A solution of (±) benzyl N-(trans-1-(2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1- yl]ethyl)-4-(prop-2-en-1-yloxy)pyrrolidin-3-yl)carbamate 1d (560 mg, 1.11 mmol) in dry and degassed DCM (10 mL) and a solution of Grubbs Catalyst (1st Generation) (92 mg, 0.11 mmol) in dry and degassed DCM (10 mL) were added simultaneously to a stirred dry and degassed DCM (300 mL) over 5 h. The reaction mixture was further stirred at room temperature overnight then concentrated in vacuo. The resulting residue was purified by flash column chromatography using a gradient eluent system of 0-5% MeOH in EtOAc to give (±) benzyl N-[trans-(10E,Z)-20-oxo-8,13-dioxa-1,4- diazatetracyclo[12.6.2.14,7.017,21]tricosa-10,14(22),15,17(21),18-pentaen-6-yl]carbamate 1e (248 mg, 47 % yield) as a grey gum. LC-MS (Method C) 476.5 [M+H]+; RT 2.94 min and 476.5 [M+H]+; RT 2.75 min (f) (±) trans-6-amino-8,13-dioxa-1,4-diazatetracyclo[12.6.2.14,7.017,21]tricosa-14(22) ,15,17(21),18–tetraen-20-one 1f
Figure imgf000067_0001
To a solution of (±) benzyl N-[trans-(10E,Z)-20-oxo-8,13-dioxa-1,4- diazatetracyclo[12.6.2.14,7.017,21]tricosa-10,14(22),15,17(21),18-pentaen-6-yl]carbamate 1e (10 mg, 0.02 mmol) in EtOH (10 mL) under N2 was added rhodium on carbon (5 wt %, 2.2 mg). The reaction mixture was purged of N2 and exposed to a blanket of H2. Hydrogenation was continued at room temperature overnight. Ammonium formate (13 mg, 0.21 mmol) was added followed by palladium on carbon (10 wt %, 2.2 mg) and the mixture warmed to 60oC for 30 min. On cooling the mixture was filtered through celite and the celite washed with MeOH. The combined alcoholic filtrates were concentrated in vacuo to give a crude oil, which was taken up in DCM and washed with aqueous saturated NaHCO3 solution. The organic fraction was dried (MgSO4) and concentrated in vacuo to give (±) trans-6-amino- 8,13-dioxa-1,4-diazatetracyclo[12.6.2.14,7.017,21]tricosa-14(22),15,17(21),18–tetraen-20-one 1f as a brown oil, which was used without further purification. LC-MS (Method C) 344.5 [M+H]+; RT 1.99 min (g) (±) trans-6-[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-8,13- dioxa-1,4-diazatetracyclo[12.6.2.14,7.017,21]tricosa-14(22),15,17(21),18–tetraen-20-one 1
Figure imgf000068_0001
A solution of (±) trans-6-amino-8,13-dioxa-1,4-diazatetracyclo[12.6.2.14,7.017,21]tricosa- 14(22),15,17(21),18–tetraen-20-one 1f (17 mg, 0.05 mmol) and 3-oxo-2H,3H,4H- pyrido[3,2-b][1,4]oxazine-6-carbaldehyde (12 mg, 0.07 mmol) in anhydrous DCM (1 mL) / THF (1 mL) was stirred at room temperature for 15 min over 4Å molecular sieves. Sodium triacetoxyborohydride (21 mg, 0.1 mmol) was added and the mixture further stirred at room temperature overnight. The reaction mixture was diluted with DCM and washed with saturated aqueous NaHCO3 solution and brine, dried (MgSO4) and concentrated in vacuo. The resulting residue was purified by flash column chromatography eluting with a gradient system of 0-30% MeOH in DCM to give (±) trans-6-[({3-oxo-2H,3H,4H-pyrido[3,2- b][1,4]oxazin-6-yl}methyl)amino]-8,13-dioxa-1,4-diazatetracyclo[12.6.2.14,7.017,21]tricosa- 14(22),15,17(21),18–tetraen-20-one 1 (4 mg, 17 % yield) as a yellow solid. 1H NMR (Method A) (CDCl3): δ ppm 7.59 (d, J = 9.3 Hz, 1H), 7.51 (br s, 1H), 7.39 (d, J = 8.5 Hz, 1H), 7.19 (d, J = 8.0 Hz, 1H), 6.91 (d, J = 8.0 Hz, 1H), 6.78 (dd, J = 8.5, 2.2 Hz, 1H), 6.52 (d, J = 9.3 Hz, 1H), 4.64 (s, 2H), 4.43 (m, 2H), 4.24 (m, 2H), 3.79 (s, 2H), 3.71 (s, 1H), 3.60 (m, 1H), 3.41 (m, 2H), 3.28 (m, 2H), 3.04 (m, 1H), 2.79 (d, J = 12.3 Hz, 1H), 2.45 (m, 1H), 2.04 (m, 3H), 1.75 (m, 2H); LC-MS (Method C) 506.5 [M+H]+; RT 2.29 min Example 2: (±) trans-6-[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]- 8,14-dioxa-1,4-diazatetracyclo[13.6.2.14,7.018,22]tetracosa-15(23),16,18(22),19–tetraen- 21-one
Figure imgf000069_0001
Prepared using a similar procedure to that described in Example 1 but using Intermediate B in step (d). 1H NMR (Method A) (CDCl3): δ ppm 7.59 (d, J = 9.3 Hz, 1H), 7.41 (d, J = 8.6 Hz, 1H), 7.35 (d, J = 2.2 Hz, 1H), 7.19 (d, J = 8.1 Hz, 1H), 6.90 (d, J = 8.0 Hz, 1H), 6.80 (dd, J = 8.6, 2.2 Hz, 1H), 6.52 (d, J = 9.3 Hz, 1H), 4.63 (s, 2H), 4.24-4.06 (m, 3H), 3.77 (d, J = 3.7 Hz, 2H), 3.66 (dd, J = 4.9, 1.6 Hz, 1H), 3.49 (s, 2H), 3.45-3.36 (m, 2H), 3.33 (t, J = 8.0 Hz, 1H), 3.26- 3.16 (m, 2H), 3.00 (m, 1H), 2.72 (dt, J = 13.4, 4.1 Hz, 1H), 2.40 (dd, J = 10.5, 4.9 Hz, 1H), 1.96 (dd, J = 9.0, 7.2 Hz, 1H), 1.92-1.82 (m, 2H), 1.79-1.58 (m, 2H), 1.30-1.22 (m, 1H); LC- MS (Method C) 520.6 [M+H]+; RT 2.35 min
Example 3: (±) cis-6-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6- yl}methyl)amino]methyl}- 8,14-dioxa-1,4-diazatetracyclo[13.6.2.14,7.018,22]tetracosa-15 (23),16,18(22),19–tetraen-21-one
Figure imgf000069_0002
Prepared using a similar procedure to that described in Example 1 but using (±) tert-butyl cis-3-({[(benzyloxy)carbonyl]amino}methyl)-4-hydroxy-pyrrolidine-1-carboxylate (prepared as described in WO2006002047, Example 24c) in step (b) and Intermediate B in step (d). Example 4: 7-[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-14-oxa- 1,4,10-triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22),19–tetraen-9,21- dione a) benzyl N-{3-[(2-oxo-1,2-dihydroquinolin-7-yl)oxy]propyl}carbamate 4a
Figure imgf000070_0001
To a suspension of 7-hydroxy-1H-quinolin-2-one (2.10 g, 13.0 mmol) and Cs2CO3 (6.37 g, 19.6 mmol) in anhydrous DMF (40 mL) was added a solution of benzyl N-(3- bromopropyl)carbamate (3.55 g, 13.0 mmol) in anhydrous DMF (10 mL) and the resulting mixture heated to 90oC . After 2 h the reaction was diluted with EtOAc (100 mL) and H2O (50 mL). A precipitate formed in the organic phase. The organic and aqueous phases were separated and the organic phase filtered to remove the precipitate. The resulting filtrate was concentrated to give a brown residue and combined with the initial precipitate using DCM. Concentration of the DCM solution gave a residue which on trituration with EtOAc gave a precipitate. The precipitate was filtered and dried to give a yellow solid of benzyl N- {3-[(2-oxo-1,2-dihydroquinolin-7-yl)oxy]propyl}carbamate 4a (2.5 g, 54 % yield), which was used without further purification. LC-MS (Method A) 353.5 [M+H]+; RT 2.42 min b) benzyl N-(3-{[1-(2,2-diethoxyethyl)-2-oxo-1,2-dihydroquinolin-7-yl]oxy}propyl)carbamate 4b
Figure imgf000071_0001
A solution of benzyl N-{3-[(2-oxo-1,2-dihydroquinolin-7-yl)oxy]propyl}carbamate 4a (654 mg, 1.86 mmol) in anhydrous DMF (150mL) was heated to 50oC for 15 min. The reaction was cooled to 40oC and Cs2CO3 (907 mg, 2.78 mmol) added followed by bromoacetaldehyde diethyl acetal (279 µL, 1.86 mmol). The mixture was heated to 70oC for 21.5 h and further bromoacetaldehyde diethyl acetal (279 µL, 1.86 mmol) added. After 65.5 h the reaction was allowed to cool and concentrated in vacuo. The resulting crude was diluted with EtOAc (100 mL) and washed with saturated aqueous NaHCO3 (30 mL). The aqueous phase was extracted with EtOAc (3 x 20 mL). The combined organic extracts were washed with brine (20 mL), dried over MgSO4 and concentrated in vacuo. The resulting crude was purified by flash column chromatography using a gradient eluent system of 0 to 10% EtOAc in DCM to give benzyl N-(3-{[1-(2,2-diethoxyethyl)-2-oxo-1,2-dihydroquinolin-7- yl]oxy}propyl)carbamate 4b (520 mg, 64 % yield) as a colourless oil. c) benzyl N-(3-{[2-oxo-1-(2-oxoethyl)-1,2-dihydroquinolin-7-yl]oxy}propyl)carbamate 4c
Figure imgf000071_0002
4b 4c To a solution of benzyl N-(3-{[1-(2,2-diethoxyethyl)-2-oxo-1,2-dihydroquinolin-7- yl]oxy}propyl)carbamate 4b (400 mg, 0.91 mmol) in THF (20 mL) was added HCl (2.0 M, 15 mL) and the resulting mixture heated to 50oC. After 2 h the reaction was allowed to cool to room temperature. EtOAc (20 mL) was added and the aqueous phase was separated and extracted using EtOAc (3 x 20 mL). The combined organic extracts were washed with brine (30 mL), dried over MgSO4 and concentrated in vacuo. The resulting residue was purified by flash column chromatography using a gradient eluent system of 0 to 10% MeOH in DCM to give benzyl N-(3-{[2-oxo-1-(2-oxoethyl)-1,2-dihydroquinolin-7-yl]oxy}propyl)carbamate 4c (155 mg, 43 % yield) as a yellow solid. LC-MS (Method A) 395.5 [M+H]+; RT 2.50 min d) methyl 8-{2-[7-(3-{[(benzyloxy)carbonyl]amino}propoxy)-2-oxo-1,2-dihydroquinol-1- yl]ethyl}-1,4-dioxa-8-azaspiro[4.5]decane-6-carboxylate 4d
Figure imgf000072_0001
A solution of benzyl N-(3-{[2-oxo-1-(2-oxoethyl)-1,2-dihydroquinolin-7- yl]oxy}propyl)carbamate 4c (525 mg, 1.33 mmol) and methyl 1,4-dioxa-8- azaspiro[4,5]decane-6-carboxylate (268 mg, 1.33 mmol) in DCM (8 mL) was treated with sodium triacetoxyborohydride (395 mg, 1.86 mmol). After stirring at room temperature for 4 h saturated aqueous NaHCO3 solution was added and the DCM layer separated. The aqueous was further extracted with DCM and the combined organics washed with brine and concentrated in vacuo to give a crude oil. The oil was purified by flash chromatography using a gradient eluent system of 0-5% MeOH in DCM to give methyl 8-{2-[7-(3- {[(benzyloxy)carbonyl]amino}propoxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl}-1,4-dioxa-8- azaspiro[4.5]decane-6-carboxylate 4d (418 mg, 92 % yield) as a colourless oil. LC-MS (Method C) 580.5 [M+H]+; RT 2.83 min e) 8-{2-[7-(3-{[(benzyloxy)carbonyl]amino}propoxy)-2-oxo-1,2-dihydroquinol-1-yl]ethyl}-1,4- dioxa-8-azaspiro[4.5]decane-6-carboxylic acid 4e
Figure imgf000073_0001
To methyl 8-{2-[7-(3-{[(benzyloxy)carbonyl]amino}propoxy)-2-oxo-1,2-dihydroquinolin-1- yl]ethyl}-1,4-dioxa-8-azaspiro[4.5]decane-6-carboxylate 4d (295 mg, 0.51 mmol) in H2O (20 mL) was added Et3N (5 mL). The resulting mixture was stirred vigorously at room temperature overnight and then concentrated under reduced pressure to give 8-{2-[7-(3- {[(benzyloxy)carbonyl]amino}propoxy)-2-oxo-1,2-dihydroquinol-1-yl]ethyl}-1,4-dioxa-8- azaspiro[4.5]decane-6-carboxylic acid 4e as a white solid, which was used without further purification. LC-MS (Method C) 566.5 [M+H]+; RT 2.60 min f) 8-{2-[7-(3-(aminopropoxy)-2-oxo-1,2-dihydroquinol-1-yl]ethyl}-1,4-dioxa-8- azaspiro[4.5]decane-6-carboxylic acid 4f
Figure imgf000074_0001
4e 4f To a stirred solution of 8-{2-[7-(3-{[(benzyloxy)carbonyl]amino}propoxy)-2-oxo-1,2- dihydroquinol-1-yl]ethyl}-1,4-dioxa-8-azaspiro[4.5]decane-6-carboxylic acid 4e (408 mg, 0.72 mmol) in MeOH (12 mL) under N2 was added palladium on carbon (10 wt %, 76 mg). The mixture was then purged of N2 and exposed to a blanket of H2. Hydrogenation was continued at room temperature for 20 h. The reaction mixture was then filtered through celite, which was washed with MeOH (3 x 5 mL). The organics were combined and solvent removed under reduced pressure to give 8-{2-[7-(3-(aminopropoxy)-2-oxo-1,2- dihydroquinol-1-yl]ethyl}-1,4-dioxa-8-azaspiro[4.5]decane-6-carboxylic acid 4f, which was used without further purification. LC-MS (Method C) 432.5 [M+H]+; RT 1.26 min (g) 14’-oxa-1’ ,4’ ,10’-triazaspiro[1,3-dioxolane-2,7’- tetracyclo[13.6.2.14,8.018,22]tetracosane]-15’(23’),16’,18’(22’),19’- tetraene-9’,21’-dione 4g
Figure imgf000074_0002
To a solution of DIPEA (0.38 mL, 2.16 mmol) and TBTU (602 mg, 1.59 mmol) in DCM (50 mL) was added 8-{2-[7-(3-(aminopropoxy)-2-oxo-1,2-dihydroquinol-1-yl]ethyl}-1,4-dioxa-8- azaspiro[4.5]decane-6-carboxylic acid 4f (311 mg, 0.72 mmol) and DIPEA (0.38 mL, 2,16 mmol) in DCM (50 mL) dropwise over 1 h. The mixture was then diluted with DCM, washed with aqueous saturated NaHCO3 solution, dried (Na2SO4) and concentrated in vacuo to give a crude residue, which was purified by flash column chromatography using a gradient eluent system of 0-10% MeOH in DCM to give 14’-oxa-1’ ,4’ ,10’-triazaspiro[1,3-dioxolane- 2,7’-tetracyclo[13.6.2.14,8.018,22]tetracosane]-15’(23’),16’,18’(22’),19’- tetraene-9’,21’-dione 4g (200 mg, 67 % yield) as a white solid. 1H NMR (Method A) (CDCl3): ^ ^ppm ^8.44 (bs, 1H), 7.59 (d, J = 9.4 Hz, 1H), 7.44 (d, J = 8.6 Hz, 1H), 7.06-6.94 (m, 1H), 6.88 (dd, J = 8.6, 2.2 Hz, 1H), 6.52 (d, J = 9.4 Hz, 1H), 4.40- 4.30 (m, 1H), 4.27-4.19 (m, 1H), 4.18-4.13 (m, 1H), 4.07-3.97 (m, 1H), 3.88-3.77 (m, 1H), 3.25-3.13 (m, 1H), 3.05-2.90 (m, 1H), 2.80 (s, 4H), 2.80-2.70 (m, 3H), 2.70-2.56 (m, 2H), 2.47-2.36 (m, 1H), 2.30-2.15 (m, 1H).2.14-2.00 (m, 1H), 1.80-1.65 (m, 2H); LC-MS (Method C) 414.4 [M+H]+; RT 2.04 min (h) 14-oxa-1,4,10-triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22),19- tetraene-7,9,21-trione 4h
Figure imgf000075_0001
To a solution of 14’-oxa-1’ ,4’ ,10’-triazaspiro[1,3-dioxolane-2,7’-tetracyclo[13.6.2.14,8.0 18,22]tetracosane]-15’(23’),16’,18’(22’),19’- tetraene-9’,21’-dione 4g (168 mg, 0.41 mmol) in DCM (30 mL) was added perchloric acid (60% aqueous solution, 1 mL). The resulting solution was stirred at room temperature for 1 h, then poured into aqueous saturated NaHCO3 solution and extracted with DCM (x3). The combined organic extracts were dried over Na2SO4 and concentrated in vacuo to give a crude product, which was purified by flash column chromatography using a gradient eluent system of 0-10% MeOH in DCM to give 14-oxa-1,4,10-triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22),19- tetraene-7,9,21-trione 4h (39 mg, 26 % yield) as a white solid. LC-MS (Method C) 370.4 [M+H]+; RT 1.30 min (i) 7-amino-14-oxa-1,4,10-triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22), 19-tetraene-9,21-dione 4i
Figure imgf000076_0001
4h 4i To 14-oxa-1,4,10-triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22),19-tetraene- 7,9,21-trione 4h (39 mg, 0.11 mmol) in EtOH (1 mL) was added ammonium acetate (123 mg, 1.58 mmol) followed by sodium cyanoborohydride (8 mg, 0.13 mmol). The reaction mixture was stirred for 5 min at room temperature, then heated in a microwave at 130oC for 2 min. On cooling the reaction mixture was concentrated in vacuo and purified by flash column chromatography to give 7-amino-14-oxa-1,4,10- triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22),19-tetraene-9,21-dione 4i (14 mg, 36 % yield) as a white solid. LC-MS (Method C) 371.5 [M+H]+; RT 1.30 min (j) 7-[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-14-oxa-1,4,10- triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22),19–tetraen-9,21-dione 4
Figure imgf000077_0001
To a solution of 7-amino-14-oxa-1,4,10-triazatetracyclo[13.6.2.14,8.018,22]tetracosa- 15(23),16,18(22),19-tetraene-9,21-dione 4i (14 mg, 0.04 mmol) in DCM (1 mL) was added 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazine-6-carbaldehyde (8 mg, 0.05 mmol) and the mixture stirred at room temperature for 15 min. Sodium triacetoxyborohydride (11 mg, 0.05 mmol) was added and stirring continued for a further 45 min. The reaction mixture was diluted with DCM and washed with saturated aqueous NaHCO3, dried over Na2SO4 and concentrated in vacuo. The resulting residue was purified by flash column chromatography using a gradient eluent system of 0-20% MeOH in DCM/3N NH3 to give 7-[({3-oxo- 2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-14-oxa-1,4,10- triazatetracyclo[13.6.2.14,8.018,22]tetracosa-15(23),16,18(22),19–tetraen-9,21-dione 4 (14 mg, 70 % yield) as a yellow solid. LC-MS (Method C) 533.5 [M+H]+; RT 2.06 min Example 5: (±) cis 7-[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-16- oxa-1,4,10- triazatetracyclo[15.6.2.14,8.020,24]hexacosa-17,19,21,24-tetraene-9,23-dione (a) (±) benzyl N-[cis-1-{2-[7-(but-3-en-1-yloxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl}-3- [(prop-2-en-1-yl)carbamoyl]piperidin-4-yl]carbamate 5a
Figure imgf000078_0001
A solution of N-[cis-3-[(prop-2-en-1-yl)carbamoyl]piperidin-4-yl]carbamate (147 mg, 0.46 mmol) and 2-[7-(but-3-en-1-yloxy)-2-oxo-1,2-dihydroquinolin-1-yl]acetaldehyde) Intermediate B (119 mg, 0.46 mmol) in DCM (8 mL) was treated with sodium triacetoxyborohydride (198 mg, 0.93 mmol). After stirring at room temperature for 1 h the reaction mixture was diluted with DCM (50 mL) and washed with saturated aqueous NaHCO3 solution, dried (MgSO4) and concentrated in vacuo. The residue was purified by flash chromatography using a gradient eluent system of 1-10% MeOH in DCM to give (±) benzyl N-[cis-1-{2-[7-(but-3-en-1-yloxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl}-3-[(prop-2-en- 1-yl)carbamoyl]piperidin-4-yl]carbamate 5a (121 mg, 47 % yield) as a colourless oil. LC-MS (Method A): 559.4 [M+H]+; RT 3.55 min (b) (±) benzyl N-[cis-(12E)-9,23-dioxo-16-oxa-1,4,10-triazatetracyclo[15.6.2.1 4,8.020,24]hexacosa-12,17,19,21,24-pentaen-7-yl]carbamate 5b
Figure imgf000078_0002
A dry degassed CHCl3 (100 mL) solution of (±) benzyl N-[cis-1-{2-[7-(but-3-en-1-yloxy)-2- oxo-1,2-dihydroquinolin-1-yl]ethyl}-3-[(prop-2-en-1-yl)carbamoyl]piperidin-4-yl]carbamate 5a (109 mg, 0.20 mmol) was treated with Grubbs Catalyst (2nd Generation) (12 mg, 0.02 mmol). The resulting mixture was stirred at room temperature overnight and recharged periodically with Grubbs Catalyst (2nd Generation) (7 x 10 mg) over a further 2 d. The reaction mixture was concentrated in vacuo and the resulting residue purified by flash chromatography using a gradient eluent system of 1-10% MeOH in EtOAc to give (±) benzyl N-[cis-(12E)-9,23-dioxo-16-oxa-1,4,10-triazatetracyclo[15.6.2.14,8.020,24]hexacosa- 12,17,19,21,24-pentaen-7-yl]carbamate 5b (45 mg, 44 % yield) as a straw coloured oil. LC-MS (Method A): 531.5 [M+H]+; RT 2.45min (c) (±) cis 7-amino-16-oxa-1,4,10-triazatetracyclo[15.6.2.14,8.020,24]hexacosa- 17,19,21,24-tetraene-9,23-dione 5c
Figure imgf000079_0001
A MeOH (10 mL) solution of (±) benzyl N-[cis-(12E)-9,23-dioxo-16-oxa-1,4,10- triazatetracyclo[15.6.2.14,8.020,24]hexacosa-12,17,19,21,24-pentaen-7-yl]carbamate 5b (60 mg, 0.11 mmol) was treated with ammonium formate (71 mg, 1.13 mmol). Wet palladium on carbon (10 wt %, 30 mg) was added and the reaction heated to 50oC for 45 min. On cooling the reaction mixture was filtered through celite and the filtrate concentrated in vacuo. The resulting residue was taken up in DCM, washed with saturated aqueous NaHCO3, dried (MgSO4) and concentrated in vacuo to give (±) cis 7-amino-16-oxa-1,4,10- triazatetracyclo[15.6.2.14,8.020,24]hexacosa-17,19,21,24-tetraene-9,23-dione 5c (34 mg, 75 % yield) as a brown oil. LC-MS (Method A): 399.4 [M+H]+; RT 1.64 min (d) (±) cis 7-[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-16-oxa- 1,4,10- triazatetracyclo[15.6.2.14,8.020,24]hexacosa-17,19,21,24-tetraene-9,23-dione 5
Figure imgf000080_0001
5c 5 To a solution of (±) cis 7-amino-16-oxa-1,4,10-triazatetracyclo[15.6.2.14,8.020,24]hexacosa- 17,19,21,24-tetraene-9,23-dione 5c (34 mg, 0.09 mmol) in DCM (5 mL) was added 3-oxo- 2H,3H,4H-pyrido[3,2-b][1,4]oxazine-6-carbaldehyde (18 mg, 0.10 mmol) and the mixture stirred at room temperature for 10 min. Sodium triacetoxyborohydride (24 mg, 1.10 mmol) was added and stirring continued for a further 30 min. The reaction mixture was diluted with DCM and washed with saturated aqueous NaHCO3, dried (MgSO4) and concentrated in vacuo. The resulting residue was purified by flash column chromatography using a gradient eluent system of 1-10% 2M NH3/MeOH in DCM to give (±) cis 7-[({3-oxo-2H,3H,4H- pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-16-oxa-1,4,10-triazatetracyclo[15.6.2.1
4,8.020,24]hexacosa-17,19,21,24-tetraene-9,23-dione 5 (10 mg, 19 % yield) as a colourless oil. 1H NMR (Method A) (CDCl3): δ ppm: 8.92 (broad s, 1H), 8.43 (s, 1H), 7.58 (d, J = 9.4 Hz, 1H), 7.45 (d, J = 8.6 Hz, 1H), 7.21 (d, J = 8.1 Hz, 1H), 6.99 (d, J = 2.2 Hz, 1H), 6.94 (d, J = 8.1 Hz, 1H), 6.80 (dd, J = 8.6, 2.2 Hz, 1H), 6.51 (d, J = 9.4 Hz, 1H), 4.64 (s, 2H), 4.52-4.48 (m, 1H), 4.41-4.33 (m, 1H), 4.25-4.19 (m, 1H), 4.08-4.02 (m, 1H), 4.00 (d, J = 13.7 Hz, 1H), 3.80 (d, J = 13.7 Hz, 1H), 3.63 (dq, J = 13.7, 6.9 Hz, 1H), 3.39 (d, J = 11.7 Hz, 1H), 3.17- 3.11 (m, 1H), 3.00 (d, J = 11.3 Hz, 1H), 2.88-2.72 (m, 3H), 2.62-2.56 (m, 1H), 2.22– 2.09 (m, 2H), 2.01-1.77 (m, 5H), 1.76-1.68 (m, 2H), 1.66-1.58 (m, 2H); LC-MS (Method A): 561.5 [M+H]+; RT 2.11 min Example 6: (±) trans-6-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-7- yl}methyl)amino]methyl}-8,13-dioxa-1,4-diazatetracyclo[12.6.2.1⁴,⁷.0¹⁷,²¹]tricosa- 14(22),15,17(21),18-tetraen-20-one
Figure imgf000081_0001
Prepared using a similar procedure to that described in Example 1 but using (±) tert-butyl trans-3-({[(benzyloxy)carbonyl]amino}methyl)-4-hydroxypyrrolidine-1-carboxylate (prepared as described in WO2006002047, Example 42d).
1H NMR (Method A) (CDCl3): δ ppm 7.60 (d, J = 2.3 Hz, 1H), 7.57 (d, J = 9.4 Hz, 1H), 7.38 (d, J = 8.6Hz, 1H), 7.19 (d, J = 8.0 Hz, 1H), 6.92 (d, J = 8.0 Hz, 1H), 6.77 (dd, J = 8.6, 2.3 Hz, 1H), 6.50 (d, J = 9.3 Hz, 1H), 4.62 (s, 2H), 4.52-4.41 (m, 2H), 4.29-4.19 (m, 2H), 3.80 (s, 2H), 3.65-3.62 (m, 1H), 3.59-3.53 (m, 1H), 3.41-3.34 (m, 1H), 3.30-3.20 (m, 2H), 3.01- 2.94 (m, 1H), 2.77-2.65 (m, 2H), 2.59-2.53 (m, 1H), 2.39-2.29 (m, 1H), 2.18 (dd, J = 10.8, 4.4 Hz, 1H), 2.05-1.94 (m, 1H), 1.91-1.82 (m, 2H), 1.76-1.69 (m, 2H); LC-MS (Method C) 520.5 [M+H]+; RT 2.26 min Example 7: (±) cis-6-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]methyl}-8,13-diox a-1,4-diazatetracyclo[12.6.2.1⁴,⁷.0¹⁷,²¹]tricosa-14(22),15,17(21),18-tetraen-20-one
Figure imgf000082_0001
Prepared using a similar procedure to that described in Example 1 but using (±) tert-butyl cis-3-({[(benzyloxy)carbonyl]amino}methyl)-4-hydroxypyrrolidine-1-carboxylate (prepared as described in WO2006002047, Example 24c). 1H NMR (Method A) (CDCl3): δ ppm 7.63 (d, J = 2.3 Hz, 1H), 7.59 (d, J = 9.4 Hz, 1H), 7.41 (d, J = 8.6 Hz, 1H), 7.28 (s, 2H), 7.21 (d, J = 8.0 Hz, 1H), 6.95 (d, J = 8.0 Hz, 1H), 6.80 (dd, J = 8.6, 2.3 Hz, 1H), 6.53 (d, J = 9.3 Hz, 1H), 4.72-4.59 (m, 3H), 4.58-4.47 (m, 1H), 4.26- 4.14 (m, 1H), 4.13-4.03 (m, 1H), 3.90 (dd, J = 5.4, 3.0 Hz, 1H), 3.86-3.74 (m, 2H), 3.63- 3.56 (m, 1H), 3.44 (t, J = 9.3 Hz, 1H), 3.24 (d, J = 10.6 Hz, 1H), 3.00-2.77 (m, 4H), 2.70- 2.62 (m, 1H), 2.61-2.48 (m, 2H), 2.41 (dd, J = 10.5, 3.1 Hz, 1H), 2.18-2.04 (m, 1H), 1.87- 1.65 (m, 3H); LC-MS (Method C) 520.6 [M+H]+; RT 2.26 min
Example 8: (6S, 8S)-6-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6- yl}methyl)amino]methyl}-15-oxa-1,4,10-triazatetracyclo[14.6.2.04,8.019,23]tetracosa- 16,18,20,23-tetraene-9,22-dione (a) 1-tert-butyl 2-methyl (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)pyrrolidine-1,2- dicarboxylate 8a
Figure imgf000083_0001
Benzyl N-succinimidyl carbonate (3.67 g, 14.7 mmol) was added slowly to a solution of 1- tert-butyl 2-methyl (2S,4S)-4-(aminomethyl)pyrrolidine-1,2-dicarboxylate (3.8 g, 14.7 mmol) in DCM (30 mL) and the mixture stirred over 48 h. The reaction mixture was concentrated in vacuo to afford a yellow gum, which was purified by flash column chromatography eluting with Et2O to give 1-tert-butyl 2-methyl (2S,4S)-4- ({[(benzyloxy)carbonyl]amino}methyl)pyrrolidine-1,2-dicarboxylate 8a as a viscous clear gum (5.89 g, 100 % yield). 1H NMR (Method B) (CDCl3): δ ppm 7.41-7.28 (m, 5H), 5.08 (s, 2H), 4.80-4.70 (m, 1H), 4.35-4.30 (m, 1H), 3.75-3.60 (m, 4H), 3.30-3.30 (m, 3H), 2.55-2.45 (m, 1H), 2.19-1.82 (m, 2H), 1.40 (s, 9H) (b) tert-butyl (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)-2-[(prop-2-en-1- yl)carbamoyl]pyrrolidine-1-carboxylate 8b
Figure imgf000083_0002
To 1-tert-butyl 2-methyl (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)pyrrolidine-1,2- dicarboxylate 8a in MeOH (30 mL) was added H2O (5 mL). The resulting solution was stirred for 10 min before the addition of LiOH.H2O (1.09 g, 19.51 mmol). After stirring for a further 1 h the reaction mixture was evaporated to approximately 30% of its initial volume, then diluted with H2O (40 mL) and extracted with Et2O (50 mL). The remaining aqueous was carefully acidified to pH 1 with 2N HCl to afford a cloudy white solution, which was extracted with Et2O (2 x 75 mL). The combined organics were dried (MgSO4) and concentrated in vacuo to afford (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)-1-[(tert- butoxy)carbonyl]pyrrolidine-2-carboxylic acid (5.14 g, 91 % yield) as a clear glassy solid which was used without further purification.
To (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)-1-[(tert-butoxy)carbonyl]pyrrolidine-2- carboxylic acid (5.14 g, 13.58 mmol) and DIPEA (5.2 mL, 29.9 mmol) in DCM (60 mL) was added allyl amine (1.12 mL, 14.9 mmol) and HATU (6.58 g, 14.94 mmol). The resulting mixture was stirred at room temperature for 1 h, evaporated to approximately 90% of its initial volume and treated with aqueous 2N K2CO3 solution (100 mL). The mixture was extracted with Et2O (2 x 100 mL). The combined organics were dried (MgSO4) and concentrated in vacuo to afford a yellow gum, which was purified by flash column chromatography eluting with 50% Et2O in petroleum ether (40-60) to give tert-butyl (2S,4S)- 4-({[(benzyloxy)carbonyl]amino}methyl)-2-[(prop-2-en-1-yl)carbamoyl]pyrrolidine-1- carboxylate 8b (5.3 g, 93.5 % yield) as a white foam/gum. 1H NMR (Method B) (CDCl3): δ ppm 7.39-7.28 (m, 5H), 7.05 (s, 1H), 5.85-5.77 (m, 1H), 5.21-5.06 (m, 4H), 4.91-4.81 (m, 1H), 4.38-4.21 (m, 1H), 4.02-3.78 (m, 2H), 3.60-3.42 (m, 1H), 3.28-2.95 (m, 3H), 2.58-2.37 (m, 12H), 1.91-1.7 (m, 1H), 1.42 (s, 9H). (c) benzyl N-{[(3S,5S)-1-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]ethyl}- 5-[(prop-2-en-1-yl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8c
Figure imgf000084_0001
To a solution of tert-butyl (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)-2-[(prop-2-en-1- yl)carbamoyl]pyrrolidine-1-carboxylate 8b (5.3 g, 12.7 mmol) in Et2O (10 mL) was added 6N hydrogen chloride in propan-2-ol (5.57 mL, 126.9 mmol). The resulting solution was stirred overnight at room temperature and then evaporated under reduced pressure to afford the HCl salt of benzyl N-{[(3R,5S)-5-[(prop-2-en-1-yl)carbamoyl]pyrrolidin-3-yl]methy}carbamate (4.38 g, 97 % yield) as a glassy clear solid which was used without further purification. To benzyl N-{[(3R,5S)-5-[(prop-2-en-1-yl)carbamoyl]pyrrolidin-3-yl]methy}carbamate hydrochloride (3.0 g, 8.50 mmol) in DCM (35 mL) was added 2-[2-oxo-7-(prop-2-en-1- yloxy)-1,2-dihydroquinolin-1-yl]acetaldehyde Intermediate A (1.97 g, 8.11 mmol) and the mixture stirred at room temperature for 1 h. Sodium triacetoxyborohydride (1.89 g, 8.92 mmol) was added and stirring continued overnight. The resulting mixture was evaporated to 90% of its initial volume, quenched with aqueous 1N NaOH solution (50 mL) and extracted with EtOAc (2 x 75 mL). The combined organics were dried (MgSO4) and concentrated in vacuo to give a yellow gum, which was taken up in MeOH and purified using an SCX cartridge eluting with MeOH/NH3. Evaporation of relevant fractions gave a further residue, which was purified by flash column chromatography eluting with EtOAc to give benzyl N- {[(3S,5S)-1-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]ethyl}-5-[(prop-2-en-1- yl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8c (2.6 g, 56.5 % yield) as an off white foam. LC-MS (Method C) 545.7 [M+H]+; RT 2.94 min (d) benzyl N-{[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-5-[(prop-2- en-1-yl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8d
Figure imgf000085_0001
To MeOH (50 mL) was added benzyl N-{[(3S,5S)-1-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2- dihydroquinolin-1-yl]ethyl}-5-[(prop-2-en-1-yl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8c (1.7 g, 3.12 mmol), K2CO3 (1.29 g, 9.36 mmol) and tetrakis(triphenylphosphine)palladium(0) (360 mg, 0.31 mmol). The resulting reaction mixture was stirred for over 48 h, then filtered and evaporated to 80% of its initial volume. The reaction mixture was then quenched with aqueous saturated ammonium chloride solution (50 mL) and extracted with Et2O (2 x 75 mL). The combined organic extracts were dried (MgSO4) and concentrated in vacuo to give a yellow gum. Purification of the gum through a SCX cartridge, eluting initially with MeOH followed by MeOH/NH3 gave benzyl N- {[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-5-[(prop-2-en-1- yl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8d (1.46 g, 2.89 mmol) as a yellow foam. LC-MS (Method C) 505.6 [M+H]+; RT 1.87 min (e) benzyl N-{[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-5-([(2E)-4- hydroxybut-2-en-1-yl]carbamoyl}pyrrolidin-3-yl]methyl}carbamate 8e
Figure imgf000086_0001
To dry degassed CHCl3 (80 mL) was added benzyl N-{[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2- dihydroquinolin-1-yl)ethyl]-5-[(prop-2-en-1-yl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8d (1.46 g, 2.89 mmol), methyl acrylate (1.97 mL, 28.94 mmol), allyl alcohol (2 mL) and Grubbs Catalyst (2nd Generation) (245 mg, 0.29 mmol). The resulting mixture was refluxed for 24 h, allowed to cool and solvent removed in vacuo. The resulting residue was taken back up in dry degassed CHCl3 (60 mL) and further quantities of methyl acrylate (2 mL), allyl alcohol (2 mL) and Grubbs Catalyst (2nd Generation) (245 mg, 0.29 mmol) added. The reaction mixture was then refluxed for a further 24 h. This process was repeated twice more. On final evaporation of the solvent the resulting residue was purified using flash column chromatography using a gradient eluent system of 0-10% MeOH in EtOAc to give benzyl N-{[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-5-([(2E)-4- hydroxybut-2-en-1-yl]carbamoyl}pyrrolidin-3-yl]methyl}carbamate 8e (620 mg, 38 % yield) as a yellow foam. LC-MS (Method C) 535.6 [M+H]+; RT 1.80 min (f) benzyl N-{[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-5-[(4- hydroxybutyl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8f
Figure imgf000087_0001
To a solution of benzyl N-{[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-5- ([(2E)-4-hydroxybut-2-en-1-yl]carbamoyl}pyrrolidin-3-yl]methyl}carbamate 8e (620 mg, 1.16 mmol) in MeOH (80 mL) under N2 was added rhodium on carbon (5 wt %, 12 mg). The mixture was then purged of N2 and exposed to a blanket of H2. Hydrogenation was continued at room temperature for 6 h. The reaction mixture was then filtered through celite, which was washed with MeOH (3 x 5 mL). The organics were combined and solvent removed under reduced pressure to give a gummy brown semi-solid, which was purified using flash column chromatography using a gradient eluent system of 5-10% MeOH in EtOAc to give benzyl N-{[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-5- [(4-hydroxybutyl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8f (485 mg, 78 % yield) as a white foam. LC-MS (Method C) 537.6 [M+H]+; RT 1.85 min (g) benzyl N-{[(6S,8S)-9,22-dioxo-15-oxa-1,4,10- triazatetracyclo[14.6.2.04,8.019,23]tetracosa-16,18,20,23-tetraen-6-yl]methyl}carbamate 8g
Figure imgf000087_0002
To DCM (50 mL) was added benzyl N-{[(3S,5S)-1-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin- 1-yl)ethyl]-5-[(4-hydroxybutyl)carbamoyl]pyrrolidin-3-yl]methyl}carbamate 8f (485 mg, 0.90 mmol) and triphenyldibromophosphorane (419 mg, 0.99 mmol). The resulting mixture was stirred overnight at room temperature. Further triphenyldibromophosphorane (168 mg, 0.40 mmol) was added and the reaction mixture stirred for a further 1 h. Evaporation of the solvent in vacuo gave a residue which was taken up in DMF (20 mL) and treated with K2CO3 (125 mg, 0.90 mmol). The resulting reaction mixture was stirred at 60oC for 2 h. On cooling H2O (70 mL) and Et2O were added sequentially resulting in precipitation of a solid, which was filtered. The solid was taken up in MeOH and purified using a SCX cartridge, eluting with MeOH/NH3. Evaporation of relevant fractions gave a solid, which was triturated with Et2O and filtered to give benzyl N-{[(6S,8S)-9,22-dioxo-15-oxa-1,4,10- triazatetracyclo[14.6.2.04,8.019,23]tetracosa-16,18,20,23-tetraen-6-yl]methyl}carbamate 8g (288 mg, 61 % yield) as a white solid. LC-MS (Method C) 519.6 [M+H]+; RT 2.60 min (h) (6S,8S)-6-(aminomethyl)-15-oxa-1,4,10-triazatetracyclo[14.6.2.04,8.019,23]tetracosa- 16,18,20,23-tetraene-9,22-dione 8h
Figure imgf000088_0001
To a solution of benzyl N-{[(6S,8S)-9,22-dioxo-15-oxa-1,4,10- triazatetracyclo[14.6.2.04,8.019,23]tetracosa-16,18,20,23-tetraen-6-yl]methyl}carbamate 8g (288 mg, 0.56 mmol) in EtOH (15 mL) under N2 was added palladium on carbon (10 wt %, 5.9 mg). The mixture was then purged of N2 and exposed to a blanket of H2. Hydrogenation was continued at 40oC for 10 h. On cooling the reaction mixture was filtered through celite, which was washed with EtOH. The combined organics were evaporated under reduced pressure and the residue taken up in MeOH and purified using an SCX cartridge eluting with MeOH/NH3. Evaporation of relevant fractions gave a further residue, which was purified by flash column chromatography eluting initially with 10% MeOH in EtOAc followed by 10% 7N NH3/MeOH in EtOAc to give (6S,8S)-6-(aminomethyl)-15-oxa-1,4,10- triazatetracyclo[14.6.2.04,8.019,23]tetracosa-16,18,20,23-tetraene-9,22-dione 8h (90 mg, 40 % yield) as a white foam. LC-MS (Method C) 385.6 [M+H]+; RT 2.59 min (i) (6S, 8S)-6-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]methyl}- 15-oxa-1,4,10-triazatetracyclo[14.6.2.04,8.019,23]tetracosa-16,18,20,23-tetraene-9,22-dione 8
Figure imgf000089_0001
A solution of (6S,8S)-6-(aminomethyl)-15-oxa-1,4,10- triazatetracyclo[14.6.2.04,8.019,23]tetracosa-16,18,20,23-tetraene-9,22-dione 8h (90 mg, 0.23 mmol) and 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazine-6-carbaldehyde (42 mg, 0.23 mmol) in CHCl3 (15 mL) was stirred over 3Å molecular sieves at 50oC for 2 h and then overnight at room temperature. Sodium borohydride (9 mg, 0.23 mmol) was then added, followed by dry MeOH (0.5 mL) and the mixture stirred for a further 20 min. Evaporation of the solvent in vacuo gave a residue, which was taken up in MeOH and purified using a SCX cartridge eluting with MeOH/NH3. Evaporation of relevant fractions gave a further residue, which was purified by flash column chromatography eluting with initially10% MeOH in EtOAc followed by 10% 7N NH3/MeOH in EtOAc. The resulting white foam was triturated with EtOAc and Et2O to give (6S, 8S)-6-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6- yl}methyl)amino]methyl}-15-oxa-1,4,10-triazatetracyclo[14.6.2.04,8.019,23]tetracosa- 16,18,20,23-tetraene-9,22-dione 8 (26 mg, 20 % yield) as a white solid. 1H NMR (Method B) (CDCl3): δ ppm 9.00 (brs, 1H), 7.59 (d, J = 9.2 Hz, 1H), 7.46 (d, J = 8.7 Hz, 1H), 7.34 (d, J = 2.6 Hz, 1H), 7.21 (d, J = 8.1 Hz, 1H), 6.89 (d, J = 8.1 Hz, 1H), 6.80 (d, J = 8.7, 1H), 6.72 (s, 1H), 6.61 (d, J = 9.2 Hz, 1H), 5.26-5.16 (m, 1H), 4.63 (s, 2H), 4.28- 4.16 (m, 2H), 3.81 (d, J = 12.2 Hz, 1H), 3.75 (d, J = 12.2 Hz, 1H), 3.75-3.68 (m, 2H), 3.62- 3.58 (m, 1H), 3.18-3.13 (m, 1H), 3.09-3.01 (m, 1H), 3.00-2.92 (m, 1H), 2.70-2.58 (m, 3H), 2.50-2.40 (m, 2H), 2.20-1.91 (m, 6H), 1.55-1.42 (m, 1H); LC-MS (Method C) 547.7 [M+H]+ RT 2.13 min Example 9: (±) (7R,8S)-7-[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6- yl}methyl)amino]-13,16-dioxa-1,4,10-triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa- 17,19,21,24-tetraene-9,23-dione (a) (±) 9-benzyl 7-methyl 1,5-dioxa-9-azaspiro[5.5]undecane-7,9-dicarboxylate 9a
Figure imgf000090_0001
A solution of (±) 1-benzyl 3-methyl 4-oxopiperidine-1,3-dicarboxylate (3.50 g, 12.02 mmol), 1,3-propane-diol (2.6 mL, 36.05 mmol) and p-toluenesulfonic acid monohydrate (114 mg, 0.60 mmol) in toluene (150 mL) was refluxed under Dean Stark conditions for 18 h. On cooling H2O (200 mL) was added and the mixture extracted with Et2O (4 x 50 mL). The combined organic extracts were washed with saturated aqueous NaHCO3, then brine, dried (MgSO4) and concentrated in vacuo to give an oil, which was purified by flash column chromatography using a gradient eluent system of 0-20% EtOAc in petroleum ether (40-60) to give (±) 9-benzyl 7-methyl 1,5-dioxa-9-azaspiro[5.5]undecane-7,9-dicarboxylate 9a (1.90 g, 45 % yield) as a colourless oil. LC-MS (Method A) 350.5 [M+H]+, RT 2.58 min (b) (±) methyl 1,5-dioxa-9-azaspiro[5.5]undecane-7-carboxylate 9b
Figure imgf000090_0002
To a solution of (±) 9-benzyl 7-methyl 1,5-dioxa-9-azaspiro[5.5]undecane-7,9-dicarboxylate 9a (1.90 g, 5.44 mmol) in MeOH (50 mL) under N2 was added palladium on carbon (10 wt %, 289 mg). The mixture was then purged of N2 and exposed to a blanket of H2. Hydrogenation was continued at room temperature for 6 h, then filtered through celite and concentrated in vacuo to give (±) methyl 1,5-dioxa-9-azaspiro[5.5]undecane-7-carboxylate 9b (1.10 g, 94 % yield) as a colourless oil. 1H NMR (Method B) (CDCl3): δ ppm 4.03 (ddd, J = 11.4, 6.7, 4.3 Hz, 1H), 3.98-3.86 (m, 3H), 3.72 (s, 3H), 3.13-3.05 (m, 2H), 3.02-2.92 (m, 2H), 2.79 (ddd, J = 13.6, 11.4, 3.4 Hz, 1H), 2.08-2.00 (m, 1H), 1.94-1.88 (m, 1H), 1.81-1.67 (m, 2H), 1.56 (brs, 1H) (c) (±) methyl 9-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]ethyl}-1,5- dioxa-9-azaspiro[5.5] undecane-7-carboxylate 9c
Figure imgf000091_0001
To (±) methyl 1,5-dioxa-9-azaspiro[5.5]undecane-7-carboxylate 9b (4.38 g, 13.8 mmol) and 2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]acetaldehyde Intermediate A (1.24 g, 5.11 mmol) in DCM (20 mL) was added sodium triacetoxyborohydride (2.17 g, 10.22 mmol). The resulting mixture was stirred at room temperature for 2.5 h, quenched with saturated aqueous NaHCO3 and extracted with EtOAc (2 x 50 mL). The combined organics were dried (MgSO4) and concentrated in vacuo to give a residue, which was purified by flash column chromatography using a gradient eluent system of 0-10% MeOH in DCM to give (±) methyl 9-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]ethyl}-1,5-dioxa-9- azaspiro[5.5] undecane-7-carboxylate 9c (2.09 g, 92 % yield) as a colourless oil. LC-MS (Method A) 443.6 [M+H]+, RT 2.23 min (d) (±) methyl 9-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-1,5-dioxa-9- azaspiro[5.5]undecane-7-carboxylate 9d
Figure imgf000092_0001
9c 9d To MeOH (20 mL) under N2 was added (±) methyl 9-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2- dihydroquinolin-1-yl]ethyl}-1,5-dioxa-9-azaspiro[5.5] undecane-7-carboxylate 9c (2.08 g, 4.70 mmol) and tetrakis(triphenylphosphine)palladium(0) (360 mg, 0.31 mmol). After stirring at room temperature for 5 min K2CO3 (2.07 g, 15.04 mmol) was added and the reaction mixture further stirred overnight. The reaction mixture was filtered through celite and concentrated in vacuo to give a residue, which was purified by flash column chromatography using a gradient eluent system of 0-5% MeOH in DCM to give (±) methyl 9-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-1,5-dioxa-9-azaspiro[5.5]undecane-7- carboxylate 9d (1.49 g, 79 % yield) as a crystalline pale brown solid. LC-MS (Method A) 403.2 [M+H]+, RT 1.21 min (e) (±) methyl 9-(2-{7-[2-(2-{[(tert-butoxy)carbonyl]amino}ethoxy)ethoxy]-2-oxo-1,2- dihydroquinolin-1-yl}ethyl)-1,5-dioxa-9-azaspiro[5.5]undecane-7-carboxylate 9e
Figure imgf000093_0001
9d 9e A stirred mixture of tert-butyl N-[2-(2-bromoethoxy)ethyl]carbamate (1.49 g, 5.55 mmol), (±) methyl 9-[2-(7-hydroxy-2-oxo-1,2-dihydroquinolin-1-yl)ethyl]-1,5-dioxa-9- azaspiro[5.5]undecane-7-carboxylate 9d (1.49 g, 3.70 mmol) and Cs2CO3 (3.62 g, 11.11 mmol) in DMF (20 mL) was heated at 90oC for 1 h. The reaction was allowed to cool to room temperature and diluted with H2O. The product was then extracted with EtOAc and the aqueous layer further washed with EtOAc. The combined organics were concentrated in vacuo to give (±) methyl 9-(2-{7-[2-(2-{[(tert-butoxy)carbonyl]amino}ethoxy)ethoxy]-2-oxo- 1,2-dihydroquinolin-1-yl}ethyl)-1,5-dioxa-9-azaspiro[5.5]undecane-7-carboxylate 9e (870 mg, 40 % yield) as a yellow oil. LC-MS (Method A) 590.7 [M+H]+, RT 2.19 min (f) (±) methyl 9-(2-{7-[2-(2-aminoethoxy)ethoxy]-2-oxo-1,2-dihydroquinolin-1-yl}ethyl)- 1.5-dioxa-9-azaspiro[5.5]undecane-7-carboxylate HCl 9f
Figure imgf000093_0002
9e 9f A solution of (±) methyl 9-(2-{7-[2-(2-{[(tert-butoxy)carbonyl]amino}ethoxy)ethoxy]-2-oxo- 1,2-dihydroquinolin-1-yl}ethyl)-1,5-dioxa-9-azaspiro[5.5]undecane-7-carboxylate 9e (870 mg, 1.48 mmol) in MeOH (2 mL) was treated with HCl (4M in dioxane) (0.37 mL, 1.48 mmol). After stirring at room temperature for 1 h the reaction mixture was concentrated in vacuo to give (±) methyl 9-(2-{7-[2-(2-aminoethoxy)ethoxy]-2-oxo-1,2-dihydroquinolin-1- yl}ethyl)-1.5-dioxa-9-azaspiro[5.5]undecane-7-carboxylate 9f as its HCl salt (780 mg, 100 % yield) as a yellow gum. LC-MS (Method A) 490.5 [M+H]+, RT 1.21 min (g) (±) 9-(2-{7-[2-(2-aminoethoxy)ethoxy]-2-oxo-1,2-dihydroquinolin-1-yl}ethyl)-1,5- dioxa-9-azaspiro[5.5]undecane-7-carboxylic acid 9g
Figure imgf000094_0001
To (±) methyl 9-(2-{7-[2-(2-aminoethoxy)ethoxy]-2-oxo-1,2-dihydroquinolin-1-yl}ethyl)-1.5- dioxa-9-azaspiro[5.5]undecane-7-carboxylate hydrochloride 9f (780 mg, 1.48 mmol) in 1,4- dioxane (5 mL) and H2O (5 mL) was added LiOH.H2O (124 mg, 2.97 mmol). After stirring at room temperature for over 72 h the reaction mixture was loaded on to a SCX cartridge and eluted with MeOH followed by MeOH/NH3. Evaporation of relevant fractions afforded (±) 9- (2-{7-[2-(2-aminoethoxy)ethoxy]-2-oxo-1,2-dihydroquinolin-1-yl}ethyl)-1,5-dioxa-9- azaspiro[5.5]undecane-7-carboxylic acid 9g (705 mg, 100 % yield) as a colourless oil. LC-MS (Method A) 476.5 [M+H]+, RT 1.15 min (h) (±) 13',16'-Dioxa-1',4',10'-triazaspiro[1,3-dioxane-2,7'- tetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosane]-17',19',21',24'-tetraene-9',23'-dione 9h
Figure imgf000095_0001
9g 9h
To a solution of DIPEA (0.39 mL, 2.22 mmol) and TBTU (1.24 g, 3.26 mmol) in DCM (60 mL) at room temperature was added a mixture of (±) 9-(2-{7-[2-(2-aminoethoxy)ethoxy]-2- oxo-1,2-dihydroquinolin-1-yl}ethyl)-1,5-dioxa-9-azaspiro[5.5]undecane-7-carboxylic acid 9g (705 mg, 1.48 mmol) and DIPEA (0.39 mL, 2.22 mmol) in DCM (30 mL) dropwise over 1 h. After stirring for a further 30 min the reaction mixture was diluted with DCM and washed with saturated aqueous NaHCO3 solution, dried (MgSO4) and concentrated in vacuo to give a residue, which was purified by flash column chromatography using a gradient eluent system of 0-10% MeOH in DCM to give (±) 13',16'-dioxa-1',4',10'-triazaspiro[1,3-dioxane- 2,7'-tetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosane]-17',19',21',24'-tetraene-9',23'-dione 9h (680 mg, 100 % yield) as a colourless solid. LC-MS (Method A) 458.6 [M+H]+, RT 1.46 min (i) (±) 13,16-dioxa-1,4,10-triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa-17,19,21,24- tetraene-7,9,23-trione 9i
Figure imgf000096_0001
To a solution of (±) 13',16'-dioxa-1',4',10'-triazaspiro[1,3-dioxane-2,7'- tetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosane]-17',19',21',24'-tetraene-9',23'-dione 9h (680 mg, 1.49 mmol) in H2O (2.5 mL) and 2-propanol (1 mL) was added 6N HCl in 2-propanol (2.48 mL). The resulting mixture was heated to reflux overnight, allowed to cool and concentrated in vacuo. The resulting residue was purified by flash column chromatography using a gradient eluent system of 0-10% MeOH in DCM to give (±) 13,16-dioxa-1,4,10- triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa-17,19,21,24-tetraene-7,9,23-trione 9i (108 mg, 18 % yield) as a colourless solid. LC-MS (Method A) 400.5 [M+H]+, RT 1.53 min (j) (±) (7R,8S)-7-amino-13,16-dioxa-1,4,10-triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa- 17,19,21,24-tetraene-9,23-dione 9j
Figure imgf000096_0002
To a solution of (±) 13,16-dioxa-1,4,10-triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa- 17,19,21,24-tetraene-7,9,23-trione 9i (64 mg, 0.16 mmol) in MeOH (1 mL) was added ammonium acetate (185 mg, 2.40 mmol) followed by sodium cyanoborohydride (12 mg, 0.19 mmol). The reaction mixture was stirred for 5 min, then heated in the microwave at 130oC for 10 min. On cooling the mixture was diluted with EtOAc, then washed with saturated aqueous NaHCO3 (2 x 50 mL), dried (MgSO4) and concentrated in vacuo. The resulting residue was purified via column chromatography using a gradient eluent system of 0-10% MeOH in DCM to give (±) (7R,8S)-7-amino-13,16-dioxa-1,4,10- triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa-17,19,21,24-tetraene-9,23-dione 9j (58 mg, 90% yield) as a pale yellow solid. LC-MS (Method A) 401.6 [M+H]+, RT 1.44 min (k) (±) (7R,8S)-7-[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-13,16- dioxa-1,4,10-triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa-17,19,21,24-tetraene-9,23-dione 9
Figure imgf000097_0001
A solution of (±) (7R,8S)-7-amino-13,16-dioxa-1,4,10- triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa-17,19,21,24-tetraene-9,23-dione 9j (53 mg, 0.13 mmol) and 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazine-6-carbaldehyde (23 mg, 0.13 mmol) in CHCl3 (11 mL) was stirred over 3Å molecular sieves at 50oC for 2 h. On cooling sodium borohydride (5 mg, 0.13 mmol) was then added, followed by dry MeOH (0.5 mL), and the mixture stirred for a further 1 h. Evaporation of the solvent in vacuo gave a residue, which was taken up in MeOH and purified using a SCX cartridge, eluting with MeOH/NH3. Evaporation of relevant fractions gave a further residue, which was purified by flash column chromatography eluting with 15% 1N NH3/MeOH in DCM to give (±) (7R,8S)-7-[({3-oxo- 2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]-13,16-dioxa-1,4,10- triazatetracyclo[15.6.2.1⁴,⁸.0²⁰,²⁴]hexacosa-17,19,21,24-tetraene-9,23-dione 9 (3 mg, 4 % yield) as a colourless solid. 1H NMR (Method B) (CDCl3): δ ppm 9.42 (s, 1H), 8.34 (s, 1H), 7.61 (d, J = 9.4 Hz, 1H), 7.44 (d, J = 8.5 Hz, 1H), 7.21 (m, 2H), 6.96 (d, J = 8.0 Hz, 1H), 6.84 (dd, J = 8.5, 2.2 Hz, 1H), 6.54 (d, J = 9.4 Hz, 1H), 5.30 (s, 2H), 4.63 (s, 2H), 4.42-4.31 (m, 2H), 4.18-4.11 (m, 1H), 4.08-4.04 (m, 1H), 3.91-3.60 (m, 5H), 3.53-3.47 (m, 1H), 3.32-3.22 (m, 1H), 3.06-3.00 (m, 1H), 2.92-2.87 (s, 1H), 2.85-2.75 (m, 1H), 2.67-2.61 (m, 1H), 2.42-2.17 (m, 2H), 1.92- 1.72 (m, 3H); LC-MS (Method A) 563.7 [M+H]+, RT 1.87 min Example 10: (6S,8S)-6-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6- yl}methyl)amino]methyl}-14-oxa-1,4,10-triazatetracyclo[13.6.2.04,8.018,22]tricosa- 15,17,19,22-tetraene-9,21-dione (a) tert-butyl N-(3-{[2-oxo-1-(2-oxoethyl)-1,2-dihydroquinolin-7-yl]oxy}propyl)carbamate 10a
Figure imgf000098_0001
To a solution of 1-(2,2-diethoxyethyl)-7-hydroxy-1,2-dihydroquinolin-2-one (2.78 g, 10.03 mmol) and tert-butyl N-(3-bromopropyl)carbamate (2.51 g, 10.53 mmol) in dry DMF (50 mL) was added K2CO3 (1.66 g, 12.04 mmol). The resulting reaction mixture was stirred at room temperature for 4 h. H2O (50 mL) was added followed by Et2O (75 mL). The organic layer was separated and the aqueous layer further extracted with Et2O (75 mL). The combined organics were dried (MgSO4) and concentrated in vacuo to give a yellow gum, which was dissolved in THF (30 mL) and treated with aqueous 2N HCl (30 mL). The resulting reaction mixture was stirred at room temperature for 4 h. H2O (50 mL) was added followed by solid K2CO3 until basic pH was attained. The mixture was then extracted with EtOAc (2 x 75 mL) and the combined organics dried (MgSO4) and concentrated in vacuo. The resulting yellow gum was purified by flash column chromatography using a gradient eluent system of 50- 100% EtOAc in Et2O to give tert-butyl N-(3-{[2-oxo-1-(2-oxoethyl)-1,2-dihydroquinolin-7- yl]oxy}propyl)carbamate 10a (1.47 g, 41 % yield) as a clear gum. 1H NMR (Method B) (CDCl3): δ ppm 9.67 (s, 1H), 7.68 (d, J = 9.3 Hz, 1H), 7.50 (d, J = 8.6 Hz, 1H), 6.81 (d, J = 8.6 Hz, 1H), 6.59 (d, J = 9.3 Hz, 1H), 6.47 (s, 1H), 5.10 (s, 2H), 4.71 (brs, 1H), 4.80 (t, J = 7.2 Hz, 2H), 3.40-3.27 (m, 2H), 2.05-1.95 (m, 2H), 1.45 (s, 9H). (b) methyl (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)-1-{2-[7-(3-{[(tert- butoxy)carbonyl]amino}propoxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl}pyrrolidine-2- carboxylate 10b
Figure imgf000099_0001
A solution of methyl (2S,4R)-4-({[(benzyloxy)carbonyl]amino}methyl)pyrrolidine-2- carboxylate (prepared by standard deprotection of the BOC group in 8a) (1.75 g, 5.99 mmol) and tert-butyl N-(3-{[2-oxo-1-(2-oxoethyl)-1,2-dihydroquinolin-7- yl]oxy}propyl)carbamate 10a (1.44 g, 4 mmol) in CHCl3 (50 mL) was stirred over 3Å molecular sieves at 44oC for 10 min. Sodium triacetoxyborohydride (1.1 g, 5.19 mmol) was added and the reaction stirred for a further 3 h at 44oC. On cooling the reaction was quenched with aqueous saturated NaHCO3 (50 mL) and extracted with EtOAc (2 x 50 mL). The combined organics were dried (MgSO4) and concentrated in vacuo. The resulting yellow gum was purified by flash column chromatography using a gradient eluent system of 80-100% EtOAc in heptane to give methyl (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)- 1-{2-[7-(3-{[(tert-butoxy)carbonyl]amino}propoxy)-2-oxo-1,2-dihydroquinolin-1- yl]ethyl}pyrrolidine-2-carboxylate 10b (0.7 g, 27% yield) as a yellow gum. LC-MS (Method C) 637 [M+H]+; RT 3.24 min (c) (2S,4S)-1-{2-[7-(3-aminopropoxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl}-4- ({[(benzyloxy)carbonyl]amino}methyl)pyrrolidine-2-carboxylate 10c
Figure imgf000100_0001
A solution of methyl (2S,4S)-4-({[(benzyloxy)carbonyl]amino}methyl)-1-{2-[7-(3-{[(tert- butoxy)carbonyl]amino}propoxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl}pyrrolidine-2- carboxylate 10b (699 mg, 1.1 mmol) in MeOH (1 mL) was treated with HCl (4M in dioxane) (20 mL). After stirring at room temperature for 1 h the reaction mixture was concentrated in vacuo. The resulting residue was dissolved in MeOH (20 mL) and H2O (4 mL). LiOH.H2O (138 mg) was added and the mixture stirred for 1 h, and then concentrated in vacuo to give (2S,4S)-1-{2-[7-(3-aminopropoxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl}-4- ({[(benzyloxy)carbonyl]amino}methyl)pyrrolidine-2-carboxylate 10c (550 mg, 67 % yield), which was used without further purification. LC-MS (Method C) 523.5 [M+H]+; RT 1.87 min (d) benzyl N-{[(6S,8S)-9,21-dioxo-14-oxa-1,4,10- triazatetracyclo[13.6.2.04,8.018,22]tricosa-15,17,19,22-tetraen-6-yl]methyl}carbamate 10d
Figure imgf000100_0002
To a solution of (2S,4S)-1-{2-[7-(3-aminopropoxy)-2-oxo-1,2-dihydroquinolin-1-yl]ethyl}-4- ({[(benzyloxy)carbonyl]amino}methyl)pyrrolidine-2-carboxylate 10c (574 mg, 1.1 mmol) in DCM (50 mL) was added Et3N (0.46mL, 3.3 mmol) and propylphosphoric anhydride (0.65 mL, 1.1 mmol). The resulting mixture was stirred at room temperature overnight and then concentrated in vacuo. The resulting residue was purified by flash column chromatography eluting with 10% MeOH in EtOAc to give benzyl N-{[(6S,8S)-9,21-dioxo-14-oxa-1,4,10- triazatetracyclo[13.6.2.04,8.018,22]tricosa-15,17,19,22-tetraen-6-yl]methyl}carbamate 10d (85 mg, 15 % yield) as a clear gum. LC-MS (Method C) RT 2.47 min (e) (6S,8S)-6-(aminomethyl)-14-oxa-1,4,10-triazatetracyclo[13.6.2.04,8.018,22]tricosa- 15,17,19,22-tetraene-9,21-dione 10e
Figure imgf000101_0001
10d 10e To a solution of benzyl N-{[(6S,8S)-9,21-dioxo-14-oxa-1,4,10- triazatetracyclo[13.6.2.04,8.018,22]tricosa-15,17,19,22-tetraen-6-yl]methyl}carbamate 10d (85 mg, 0.17 mmol) in DCM (50 mL) under N2 was added palladium on carbon (10 wt %, 18 mg). The mixture was then purged of N2 and exposed to a blanket of H2. Hydrogenation was continued at room temperature overnight. The reaction mixture was then filtered through celite. The solvent was removed in vacuo to give a residue that was purified by flash column chromatography eluting with 10% MeOH in EtOAc to give (6S,8S)-6- (aminomethyl)-14-oxa-1,4,10-triazatetracyclo[13.6.2.04,8.018,22]tricosa-15,17,19,22-tetraene- 9,21-dione 10e (59 mg, 95% yield) as a clear gum. LC-MS (Method C) 371 [M+H]+; RT 1.86 min (f) (6S,8S)-6-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]methyl}-14- oxa-1,4,10-triazatetracyclo[13.6.2.04,8.018,22]tricosa-15,17,19,22-tetraene-9,21-dione 10
Figure imgf000102_0001
A solution of (6S,8S)-6-(aminomethyl)-14-oxa-1,4,10- triazatetracyclo[13.6.2.04,8.018,22]tricosa-15,17,19,22-tetraene-9,21-dione 10e (59 mg, 0.16 mmol) and 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazine-6-carbaldehyde (29 mg, 0.16 mmol) in CHCl3 (11 mL) was heated at 50oC for 10 min. Sodium triacetoxyborohydride (152 mg, 0.72 mmol) was added and the reaction stirred at 50oC overnight. On cooling solvent was removed in vacuo and the resulting residue taken up in MeOH and purified using a SCX cartridge, eluting with MeOH/NH3. Evaporation of relevant fractions gave a further residue, which was purified by flash column chromatography eluting with 10% MeOH in EtOAc followed by 10% 6N NH3/MeOH in EtOAc to give a yellow foam, which was dissolved in DCM and triturated with petroleum ether (40-60) to give (6S,8S)-6-{[({3-oxo-2H,3H,4H- pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]methyl}-14-oxa-1,4,10- triazatetracyclo[13.6.2.04,8.018,22]tricosa-15,17,19,22-tetraene-9,21-dione 10 (20 mg, 22 % yield) as an off white solid. 1H NMR (Method B) (CDCl3): δ ppm 7.41 (brd, J = 8.2 Hz,1H), 7.58 (d, J = 9.1 Hz, 1H), 7.42 (d, J = 9.1 Hz, 1H), 7.20 (d, J = 9.3 Hz, 1H), 6.90 (d, J = 8.2 Hz, 1H), 6.80-6.71 (m, 2H), 6.52 (d, J = 8.2, 1H), 5.31-5.19 (brm, 1H), 4.69-4.59 (m, 2H), 4.46-4.35 (m, 1H), 4.30- 4.16 (m, 2H), 4.01-3.90 (m,1H), 3.81 (s, 2H), 3.68-3.49 (m, 2H), 3.35-3.22 (m, 1H), 3.00- 2.72 (m, 2H), 2.70-2.54 (m, 2H), 2.55-2.42 (m, 1H), 2.35 (t, J = 7.3 Hz, 1H), 2.17-2.05 (m, 1H), 2.00-1.82 (m, 3H), 1.55-1.42 (m, 2H); LC-MS (Method C) 533 [M+H]+; RT 1.92 min Example 11: (±) 7-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6- yl}methyl)amino]methyl}-10,15-dioxa-1,4-diazatetracyclo[14.6.2.14,7.019,23]pentacosa- 16,18,20,23-tetraen-22-one (a) (±) tert-butyl 3-cyano-3-[2-(prop-2-en-1-yloxy)ethyl]pyrrolidine-1-carboxylate 11a
Figure imgf000103_0001
To a solution of tert-butyl 3-cyano-3-(2-hydroxyethyl)pyrrolidine-1-carboxylate (0.93 g, 3.87 mmol) in THF (25 mL) was added allyl bromide (1 mL, 11.61 mmol) followed by NaH (60% dispersed in mineral oil) (278 mg, 5.81 mmol). The resulting mixture was stirred for 4 h at room temperature. Saturated aqueous ammonium chloride solution (50 mL) was added and the mixture extracted with Et2O (3 x 50 mL). The combined organic extracts were dried (MgSO4) and concentrated in vacuo. The resulting residue was purified by flash column chromatography eluting with 30-50% Et2O in heptanes to give (±) tert-butyl 3-cyano-3-[2- (prop-2-en-1-yloxy)ethyl]pyrrolidine-1-carboxylate 11a (900 mg, 83 % yield) as a clear gum. 1H NMR (Method B) (CDCl3): δ ppm 5.95-5.85 (m 1H), 5.29 (d, J = 17.2 Hz,1H), 5.19 (d, J = 12.1 Hz, 1H), 4.00 (s, 2H), 3.89 (dd, J = 17.2 Hz, J = 12.1 Hz, 1H), 3.71-3.65 (m, 2H), 3.60-3.49 (m, 2H), 3.28 (d, J = 12.1 Hz, 1H), 2.40-2.33 (m, 1H), 2.02-1.91 (m, 3H), 1.47 (s, 9H) (b) (±) 1-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]ethyl}-3-[2-(prop-2-en-1- yloxy)ethyl]pyrrolidine-3-carbonitrile 11b
Figure imgf000104_0001
A solution of (±) tert-butyl 3-cyano-3-[2-(prop-2-en-1-yloxy)ethyl]pyrrolidine-1-carboxylate 11a (1.26 g, 4.49 mmol) in HCl propan-2-ol solution (6N) (23.2 mL, 529 mmol) was stirred for 1 h at room temperature. This was then added to a mixture of Et2O (150 mL) and H2O (10 mL) containing solid K2CO3. After vigorous stirring for 30 min the mixture was filtered and concentrated in vacuo to give a clear gum, which was dissolved in DCM (40 mL) and treated with 2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]acetaldehyde Intermediate A (1.61 g, 6.66 mmol). The resulting mixture was stirred for 30 min at room temperature. Sodium triacetoxyborohydride (1.83 g, 8.65 mmol) was added and the mixture stirred for a further 1 h. H2O (50 mL) was added and the reaction mixture extracted with Et2O (2 x 75 mL). The combined organics were dried (MgSO4) and concentrated in vacuo. The resulting residue was purified by flash column chromatography using a gradient eluent system of 50-100% Et2O in heptanes to give (±) 1-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2- dihydroquinolin-1-yl]ethyl}-3-[2-(prop-2-en-1-yloxy)ethyl]pyrrolidine-3-carbonitrile 11b (1.5 g, 52 % yield) as a yellow gum. 1H NMR (Method B) (CDCl3): δ ppm 7.58 (d, J = 8.2 Hz,1H), 7.46 (d, J = 7.6 Hz,1H), 6.89 (d, J = 2.1 Hz, 1H), 6.82 (d, J = 7.6 Hz,1H), 6.53 (d, J = 8.2 Hz,1H), 6.14-6.05 (m, 1H), 5.94-5.86 (m, 1H), 5.48 (d, J = 12.2 Hz,1H), 5.35 (d, J = 8.4 Hz,1H), 5.27 (d, J = 12.2 Hz, 1H), 5.19 (d, J = 8.4 Hz,1H), 4.66 (d, J = 2.2 Hz, 2H), 4.39 (t, J = 7.2 Hz, 2H), 3.99 (d, J = 2.2 Hz, 2H), 3.65 (t, J = 7.2 Hz, 2H), 3.18 (d, J = 7.6 Hz, 1H), 3.00-2.93 (m, 1H), 2.87-2.68 (m, 4H), 2.44-2.35 (m, 1H), 2.02-1.94 (m, 3H) (c) (±) (12EZ)-22-oxo-10,15-dioxa-1,4-diazatetracyclo[14.6.2.14,7.019,23]pentacosa-12, 16(24), 17, 19(23), 20-pentaene-7-carbonitrile 11c
Figure imgf000105_0001
A solution of (±) 1-{2-[2-oxo-7-(prop-2-en-1-yloxy)-1,2-dihydroquinolin-1-yl]ethyl}-3-[2-(prop- 2-en-1-yloxy)ethyl]pyrrolidine-3-carbonitrile 11b (1.51 g, 3.71 mmol) in dry and degassed DCM (40 mL) was stirred for 10 min before the addition of Grubbs Catalyst (1st Generation) (629 mg, 0.74 mmol) in one portion. The resulting mixture was heated under reflux for 6 h, allowed to cool to room temperature and stirred over 48 h. The reaction mixture was concentrated to a low volume and purified using a SCX cartridge, eluting with MeOH/NH3. Evaporation of relevant fractions gave a further residue, which was purified by flash column chromatography using a gradient eluent system of 50-100% Et2O in heptanes to give (±) (12EZ)-22-oxo-10,15-dioxa-1,4-diazatetracyclo[14.6.2.14,7.019,23]pentacosa-12, 16(24), 17, 19(23), 20-pentaene-7-carbonitrile 11c (520 mg, 37 % yield) as a mixture of E and Z isomers. LC-MS (Method C) 380 [M+H]+; RT 2.58 min (d) (±) 7-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]methyl}-10,15- dioxa-1,4-diazatetracyclo[14.6.2.14,7.019,23]pentacosa-16,18,20,23-tetraen-22-one 11
Figure imgf000106_0001
11c 11 To a solution of (±) (12EZ)-22-oxo-10,15-dioxa-1,4- diazatetracyclo[14.6.2.14,7.019,23]pentacosa-12,16(24),17,19(23),20-pentaene-7-carbonitrile 11c (520 mg, 1.37 mmol) in dry THF (50 mL) under N2 was added Raney Nickel (8 mg). The mixture was then purged of N2 and exposed to a blanket of H2. Hydrogenation was continued at room temperature overnight. The reaction mixture was then flushed through a SCX cartridge and relevant fractions concentrated in vacuo to give a yellow gum, which was dissolved in DCM and treated with 3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazine-6- carbaldehyde (244 mg, 1.37 mmol). The reaction mixture was stirred at room temperature for 1 h, before the addition of sodium triacetoxyborohydride (377 mg, 1.78 mmol) in one portion. After 5 min the reaction mixture was quenched with MeOH, concentrated to a low volume and purified using a SCX cartridge, eluting with MeOH/NH3. Evaporation of relevant fractions gave a further residue, which was purified by flash column chromatography eluting with 10% MeOH in EtOAc followed by a gradient eluent system of 5-10% 7N NH3/MeOH in EtOAc to give (±) 7-{[({3-oxo-2H,3H,4H-pyrido[3,2-b][1,4]oxazin-6-yl}methyl)amino]methyl}- 10,15-dioxa-1,4-diazatetracyclo[14.6.2.14,7.019,23]pentacosa-16,18,20,23-tetraen-22-one 11 (75 mg, 10 % yield) as a white foam. 1H NMR (Method B) (CDCl3): δ ppm 8.45 (brs, 1H), 7.58 (d, J = 8.2 Hz,1H), 7.40 (d, J = 7.6 Hz,1H), 7.19 (d, J = 7.6 Hz,1H), 6.96 (d, J = 2.1 Hz,1H), 6.91 (d, J = 7.2 Hz,1H), 6.79 (dd, J = 7.2 Hz, J = 2.1 Hz, 1H), 6.52 (d, J = 8.2 Hz,1H), 4.65-4.55 (m, 3H), 4.25-4.14 (m, 3H), 3.78 (s, 2H), 3.60-3.40 (m, 4H), 3.06-2.96 (m, 1H), 2.80-2.50 (m, 6H), 2.07-1.51 (m, 10H); LC-MS (Method C) 548 [M+H]+; RT 2.53 min Example 12 - Antibacterial susceptibility testing Minimum Inhibitory Concentrations (MICs) versus planktonic bacteria are determined by the broth microdilution procedure according to the guidelines of the Clinical and Laboratory Standards Institute (Clinical and Laboratory Standards Institute. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically). The broth dilution method involves a two-fold serial dilution of compounds in 96-well microtitre plates, giving a final concentration range of 0.03-64 µg/mL or 0.25-128 µg/mL. Strains are grown in cation-adjusted Müller-Hinton broth (supplemented with 2% w/v NaCl in the case of methicillin-resistant S. aureus strains and 2% IsoVitalex in the case of Francisella tularensis) or on Müller-Hinton agar at 37°C in an ambient atmosphere. The MIC is determined as the lowest concentration of compound that inhibits growth following an incubation period of 16-24 h, of 12-24 h (Bacillus anthracis), of 46-50 h (Francisella tularensis) or of 24-48 h (Yersinia pestis). Table 1 - MIC values against Gram-negative and Gram-positive bacterial strains
A represents a concentration of 1 µg/mL or lower; B represents a concentration of from 1.1 to 8 µg/mL; C represents a concentration of from 9 µg/mL to 127 µg/mL; and D represents a concentration of 128 µg/mL or higher.
Figure imgf000107_0001
Figure imgf000108_0002
Table 2 - MIC values against a panel of biodefence microorganisms
A represents a concentration of 1 µg/mL or lower; B represents a concentration of from 1.1 to 8 µg/mL; C represents a concentration of from 9 µg/mL to 127 µg/mL; and D represents a concentration of 128 µg/mL or higher.
Figure imgf000108_0001
Example 13 - Human cell viability assay
Compounds are assessed for potential non-specific cytotoxic effects against a human hepatic cell line (HepG2 ATCC HB-8065). HepG2 cells are seeded at 20,000 cells/well in 96-well microtitre plates in minimal essential medium (MEM) supplemented with a final concentration of 10% FBS and 1 mM sodium pyruvate. After 24 h, compound dilutions are prepared in Dulbecco’s minimum essential media (DMEM) supplemented with final concentrations of 0.001% FBS, 0.3% bovine albumin and 0.02% HEPES and added to cells. Compounds are tested in two-fold serial dilutions over a final concentration range of 1-128 µg/mL in a final DMSO concentration of 1% vol/vol. Chlorpromazine is used as a positive control. Cells are incubated with compound at 37°C and 5% CO2 for a further 24 h, after which time the CellTiter-Glo reagent (Promega) is added. Luminescence is measured on a BMG Omega plate reader. Data are analysed using GraphPad Prism software to determine the concentration of compound that inhibits cell viability by fifty percent (IC50). Table 3 - IC50 values against HepG2
IC50 (in ^g/mL) of less than 1 is assigned the letter D; an IC50 of from 1 to 10 is assigned the letter C; an IC50 of from 10 to 100 is assigned the letter B; and an IC50 of over 100 is assigned the letter A.
Figure imgf000109_0001
The research leading to these results was conducted as part of the ENABLE consortium and has received support from the Innovative Medicines Joint Undertaking under Grant Agreement n° 115583, resources which are composed of financial contribution from the European Union’s seventh framework programme (FP7/2007-2013) and EFPIA companies in kind contribution.

Claims

CLAIMS 1. A compound of formula (I), or a pharmaceutically acceptable salt or N-oxide thereof:
Figure imgf000110_0001
(I) wherein
group A is selected from a 5-12heterocycloalkyl group comprising at least one nitrogen in the ring system and a 3-6heteroalkyl group comprising at least one nitrogen in the linking chain; L3 is attached by a covalent bond to an atom selected from the nitrogen and carbon atoms which form the group A ring system or linking chain; Z is independently selected from N and CR2; R1 and R2 are each independently selected from: H, C1-C4-alkyl, halogen, OR8, NR8R9 and C1-C4-haloalkyl; X1, X2, X3 and X4 are each independently selected from: N and CR10; wherein no more than two of X1, X2, X3 and X4 are N; wherein a single one of X3 and X4 is a carbon atom attached by a covalent bond to Y1; Y1 is independently selected from O, CR5R5, -C(R8)=C(R8)-, NR9, S and S(O)2; R5 is independently at each occurrence selected from: H, F, C1-C4-alkyl, NR8R9, OR8, C1-C4- haloalkyl and CO2R8; or two R5 groups attached to the same carbon together form =O; L1 is a linker group having the form -(CR5R5)r-; wherein r is an integer selected from 2 and 3; L5 is absent or is -L6-L2-; L6 is absent or is–L4NR6-; L2 is independently selected from–CR5R5- and a 3-, 4- or 5- membered cycloalkyl or heterocycloalkyl ring; L3 is independently –(CR5R5)s-Y3-(CR5R5)t-Y2-(CR5R5)u-; wherein s and t are each independently an integer selected from 1, 2, 3 and 4; u is an integer independently selected from 0 and 1; Y2 and Y3 are each independently selected from a bond, O, - C(R8)=C(R8)-, 1,2,3-triazole, NR9, S and S(O)2; and wherein L1, group A, r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size from 13 to 17 atoms; L4 is independently a bond or is–CR5R5-; R6 and R8 are independently at each occurrence selected from: H, C1-C4-alkyl and C1-C4- haloalkyl; R7 is a monocyclic aromatic or heteroaromatic ring or R7 is a bicyclic carbocyclic or heterocyclic ring system in which at least one of the two rings is aromatic or heteroaromatic; R9 is independently at each occurrence selected from H, C1-C4-alkyl, C1-C4-haloalkyl, S(O)2R8, C(O)NR8R8, C(O)R8 and C(O)OR8; R10 is independently at each occurrence selected from: H, halo, nitro, cyano, NR8R9, OR8; O-aryl, SR8, SOR8, SO3R8, SO2R8, SO2NR8R8, CO2R8, C(O)R8, CONR8R8, aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; wherein each of the aforementioned alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, carbocyclic, halocycloalkyl, heterocyclic, aryl (e.g. phenyl) and heteroaryl groups and aromatic and heteroaromatic rings is optionally substituted, where chemically possible, by 1 to 5 substituents which are each independently at each occurrence selected from the group consisting of: oxo, =NRa, =NORa, halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa , CO2Ra C(O)Ra, CONRaRa, CRaRaNRaRa, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; wherein Ra is independently at each occurrence selected from H and C1-C4-alkyl. 2. A compound of claim 1, wherein the compound of formula (I) is a compound of formula (XI):
Figure imgf000112_0001
(XI)
L3 is attached by a covalent bond to an atom selected from Ca, Cb and Cc; and wherein the positions on Ca, Cb and Cc to which L3 is not attached are occupied by R5 groups; R1, R2, R5, R6 and R8 are each independently at each occurrence selected from: H and C1-C4-alkyl; X1, X2 and X4 are each independently selected from: N and CR10; wherein no more than two of X1, X2 and X4 are N; L1 is a linker group having the form -(CR5R5)r-; wherein r is an integer selected from 2 and 3; L3 is independently–(CR5R5)s-Y3-(CR5R5)t-Y2-(CR5R5)u-; wherein s and t are each independently an integer selected from 1, 2, 3 and 4; u is an integer independently selected from 0 and 1; Y2 and Y3 are each independently selected from a bond, O, - C(R8)=C(R8)-, 1,2,3-triazole, NR9, S and S(O)2; and wherein r, s, t, u, Y2 and Y3 are selected such that the macrocyclic ring has a ring size from 13 to 17 atoms; L4 is independently a bond or is–CR5R5-; R9 is independently at each occurrence selected from H, C1-C4-alkyl, S(O)2R8, C(O)NR8R8, C(O)R8 and C(O)OR8; R10 is independently at each occurrence selected from: H, halo, nitro, cyano, NR8R9, OR8; O-aryl, SR8, SOR8, SO3R8, SO2R8, SO2NR8R8, CO2R8, C(O)R8, CONR8R8, aryl, C1-C4-alkyl, C2-C4-alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; m is an integer independently selected from 0 and 1; n is an integer selected from 1,
2 or 3; V1, V2 and V3 are each independently selected from: N and CR11; with the proviso that no more than two of V1, V2 and V3 are N; the ring B is a substituted or unsubstituted 5- or 6- membered saturated cycloalkyl or heterocycloalkyl ring; R11 is independently at each occurrence selected from: H, halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa, CO2Ra C(O)Ra, CONRaRa, CRaRaNRaRa, C1-C4-alkyl, C2-C4- alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl. wherein each of the aforementioned alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, carbocyclic, halocycloalkyl, heterocyclic, aryl (e.g. phenyl) and heteroaryl groups and aromatic and heteroaromatic rings is optionally substituted, where chemically possible, by 1 to 5 substituents which are each independently at each occurrence selected from the group consisting of: oxo, =NRa, =NORa, halo, nitro, cyano, NRaRa, NRaS(O)2Ra, NRaC(O)Ra, NRaCONRaRa, NRaCO2Ra, ORa; SRa, SORa, SO3Ra, SO2Ra, SO2NRaRa, CO2Ra C(O)Ra, CONRaRa, CRaRaNRaRa, C1-C4-alkyl, C2-C4- alkenyl, C2-C4-alkynyl and C1-C4-haloalkyl; wherein Ra is independently at each occurrence selected from H and C1-C4-alkyl.
3. A compound of claim 2, wherein L3 is attached to Ca.
4. A compound of claim 2, wherein L3 is attached to Cb.
5. A compound of claim 2, wherein L3 is attached to Cc.
6. A compound of any one of claims 2 to 5, wherein X1, X2 and X4 are each CR10, wherein R10 is independently selected from: H, halo, C1-C4-alkyl and C1-C4-haloalkyl.
7. A compound of any one of claims 2 to 6, wherein R1 is H.
8. A compound of any one of claims 2 to 7, wherein R2 is H.
9. A compound of any one of claims 2 to 8, wherein r is 2.
10. A compound of any one of claims 2, 3 and 5 to 9, wherein m is 0.
11. A compound of any one of claims 2, 3 and 5 to 9, wherein m is 1.
12. A compound of any one of claims 2 to 11, wherein L4 is a bond.
13. A compound of any one of claims 2 to 11, wherein L4 is–CR5R5-.
14. A compound of any one of claims 2 to 11, wherein n is 2.
15. A compound of claim 1, wherein the compound of formula (I) has a structure selected from:
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
16. A pharmaceutical composition comprising a compound of any one of claims 1 to 15 and a pharmaceutically acceptable excipient.
17. A compound of any one of claims 1 to 15 for therapeutic use.
18. A compound of any one of claims 1 to 15 for use in treating a bacterial infection.
19. A compound of any one of claims 1 to 15 for the use of claim 18, wherein the bacterial infection is caused by a Gram negative bacterial strain.
20. A compound of any one of claims 1 to 15 for the use of claim 18, wherein the bacterial infection is caused by a Gram positive bacterial strain.
21. A compound of any one of claims 1 to 15 for the use of any one of claims 18 to 20, wherein the bacterial strain is a resistant strain.
22. A compound of any one of claims 1 to 15 for the use of any one of claims 18 to 20, wherein the bacterial strain is a strain used in biowarfare.
PCT/GB2016/051514 2015-05-26 2016-05-25 Antibacterial compounds WO2016189304A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1509006.1 2015-05-26
GBGB1509006.1A GB201509006D0 (en) 2015-05-26 2015-05-26 Antibacterial compounds

Publications (1)

Publication Number Publication Date
WO2016189304A1 true WO2016189304A1 (en) 2016-12-01

Family

ID=53540914

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2016/051514 WO2016189304A1 (en) 2015-05-26 2016-05-25 Antibacterial compounds

Country Status (2)

Country Link
GB (1) GB201509006D0 (en)
WO (1) WO2016189304A1 (en)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0154142A1 (en) 1984-02-02 1985-09-11 Merck & Co. Inc. Substituted hexahydro arylquinolizines
WO2000047207A1 (en) 1999-02-09 2000-08-17 Bristol-Myers Squibb Company LACTAM INHIBITORS OF FXa AND METHOD
WO2003068790A2 (en) * 2002-02-13 2003-08-21 Abbott Laboratories Macrolide antibacterial compounds
US6645980B1 (en) 2000-05-25 2003-11-11 Sepracor Inc. Heterocyclic analgesic compounds and methods of use thereof
WO2006002047A2 (en) 2004-06-15 2006-01-05 Glaxo Group Limited Antibacterial agents
EP1900732A1 (en) * 2005-06-24 2008-03-19 Toyama Chemical Co., Ltd. Novel nitrogenated heterocyclic compound and salt thereof
US20110092495A1 (en) * 2005-06-16 2011-04-21 Astrazeneca Ab Compounds for the treatment of multi-drug resistant bacterial infections
WO2013139697A1 (en) * 2012-03-17 2013-09-26 Polyphor Ag Conformationally constrained, fully synthetic macrocyclic compounds

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0154142A1 (en) 1984-02-02 1985-09-11 Merck & Co. Inc. Substituted hexahydro arylquinolizines
WO2000047207A1 (en) 1999-02-09 2000-08-17 Bristol-Myers Squibb Company LACTAM INHIBITORS OF FXa AND METHOD
US6645980B1 (en) 2000-05-25 2003-11-11 Sepracor Inc. Heterocyclic analgesic compounds and methods of use thereof
WO2003068790A2 (en) * 2002-02-13 2003-08-21 Abbott Laboratories Macrolide antibacterial compounds
WO2006002047A2 (en) 2004-06-15 2006-01-05 Glaxo Group Limited Antibacterial agents
US20110092495A1 (en) * 2005-06-16 2011-04-21 Astrazeneca Ab Compounds for the treatment of multi-drug resistant bacterial infections
EP1900732A1 (en) * 2005-06-24 2008-03-19 Toyama Chemical Co., Ltd. Novel nitrogenated heterocyclic compound and salt thereof
WO2013139697A1 (en) * 2012-03-17 2013-09-26 Polyphor Ag Conformationally constrained, fully synthetic macrocyclic compounds

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
E. L. ELIEL; S. H. WILEN: "Stereochemistry of Organic Compounds", 1994, WILEY
FA CAREY; RJ SUNDBERG: "Advanced Organic Chemistry, Part B, Reactions and Synthesis", 2001, KLUWER ACADEMIC/PLENUM PUBLICATIONS
M. E. AULTON, PHARMACEUTICALS - THE SCIENCE OF DOSAGE FORM DESIGNS, 1988
MB SMITH; J. MARCH: "March's Advanced Organic Chemistry - Reactions, Mechanisms and Structure 5th edition", WILEY
RC LAROCK: "Comprehensive Organic Transformations - A Guide to Functional Group Transformations", 1999, WILEY-VCH
RK MACKIE; DM SMITH: "Guidebook To Organic Synthesis", 1982, LONGMAN
S WARREN: "Designing Organic Syntheses", 1983, WILEY
S WARREN: "Organic Synthesis - The Disconnection Approach", 1982, WILEY
TW GREENE; PGM WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS INC

Also Published As

Publication number Publication date
GB201509006D0 (en) 2015-07-08

Similar Documents

Publication Publication Date Title
US11884674B2 (en) Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra- hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11866419B2 (en) Antibacterial compounds
EP1611133B1 (en) Condensed n-heterocyclic compound and use thereoff as crf receptor antagonist
EP3601282B1 (en) Novel pyrido[2,3-b]indole compounds for the treatment and prophylaxis of bacterial infection
WO2016067009A1 (en) Compounds with activity against bacteria and mycobacteria
AU2014324535A1 (en) Aryl substituted aminomethyl spectinomycin analogs as antibacterial agents
WO2016189304A1 (en) Antibacterial compounds
EP3887370B1 (en) Tricyclic compounds for the treatment and prophylaxis of bacterial infection
US10487093B2 (en) Heterocyclic compounds, in particular 2-oxo-4,4,5,5,6,6,7,7-octahydrobenzoxazole derivatives, and their use as antibacterial compounds
CA3012567A1 (en) Antibacterial compounds
WO2016024096A1 (en) Antibacterial compounds
US11970500B1 (en) Crystalline form of (s)-7-(1-acryloylpiperidin-4-yl)- 2-(4-phenoxyphenyl)-4,5,6,7-tetra- hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
CA3012569A1 (en) Antibacterial compounds
US20240132470A1 (en) Antibacterial Compounds
WO2017046606A1 (en) Antibacterial compounds
GB2547740A (en) Antibacterial compounds

Legal Events

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

Ref document number: 16730459

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16730459

Country of ref document: EP

Kind code of ref document: A1