ANTIBACTERIAL PRODUCTS

The invention provides a combination of an antibacterial agent and a delivery agent, in which the delivery agent is bonded, or capable of binding, to the antibacterial agent, and in which the delivery agent is capable of binding to one or more structures on a bacterial cell membrane. The invention further provides the use of such combinations in treating or preventing bacterial infections.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE

This application is a continuation of U.S. Pat. Application No. 15/508,651, filed Mar. 3, 2017, as a national phase application under 35 U.S.C. § 371 of International Application No. PCT/GB2015/052564, filed Sep. 4, 2015, which claims priority to United Kingdom Application No. 1415776.2, filed Sep. 5, 2014, the entire contents of which are hereby incorporated by reference.

SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jun. 30, 2022, is named EPCL.P0056US.C1_ST25.txt and is 7.86 KB in size.

Field of the Invention

This invention relates to delivery agents that have affinity for bacterial cell walls, and combinations comprising such delivery agents together with antibacterial agents for treating and preventing bacterial infections.

Background

We are currently facing a worldwide pandemic of multidrug resistant bacteria, arising from the long-term use of antibacterials. Antibacterial overavailability and poor prescribing practices have allowed exposure to sub-optimal concentrations of antibacterials, promoting the evolution of environmental resistance mechanisms in bacteria.

There is therefore a continuing need to develop new ways of combating unwanted bacterial growth, particularly in bacteria that are resistant to existing drugs.

Some existing drugs target enzymes that are involved in the construction of bacterial cell walls. The bacterial cell wall is assembled by a glycosyltransferase (GT51) (http://www.cazy.org/GT51.html) and transpeptidase (TP) enzymes, which together catalyse the bond formation in the important cell wall polymer peptidoglycan. This is a mesh-like structure and is responsible for bacterial shape and strength. Due to their periplasmic location and lack of mammalian counterpart, both the GT51 and TP are excellent targets for inhibition. Indeed the transpeptidase is inhibited by the medically important ß-lactam group of antibacterials. The GT51 domains are membrane associated and catalyse the polymerisation of a lipid II pentapeptide substrate into b1,4-linked N-acetylmuramic acid, N-acetylglucosamine polymers [(MurNAc-GlcNAc)n] (Typas A., et al., Nat Rev Microbiol, 10, 123-136).

The GT51 enzymes are inhibited by moenomycins which cause accumulation of cell-wall intermediates leading to bacterial cell lysis and death. It is the only class of antibacterial which inhibits the GT51 enzyme. Moenomycins are extremely potent with minimal inhibitory concentration (MIC) against various Gram positive bacteria ranging from 1 ng/ml-100 ng/ml biological activity and is 10-1000 fold more potent than clinically useful glycopeptide vancomycin.

Bambermycin (also known as flavomycin, flavophospholipol) is a phosphoglycolipid antimicrobial produced by various strains of Streptomyces. Moenomycin A is a component of Bambermycin. It is active primarily against Gram-positive bacteria due to inhibition of transglycosylase and thus of cell wall synthesis (Pfaller M.A., et al. Diagnostic Microbiology and Infectious Disease 56 (2006) 115-121).

The antimicrobial spectrum of flavophospholipol is known to be limited. Both fermentative (e.g. Enterobacteriaceae) and nonfermentative (e.g., Pseudomonas spp.) Gram-negative bacilli are considered to be inherently resistant to flavophospholipol. It is believed that flavophospholipol is unable to penetrate the outer membrane of Gram-negative organisms and thus is unable to reach the target elements in these organisms (Pfaller M.A., ibid.).

Despite their potency, interest in developing moenomycins as novel drugs dissipated because of their poor bioavailability and suboptimal pharmacokinetic properties. (Ostash B. et al. Nat. Prod. Rep., 2010, 27, 1594-1617). Moenomycins show almost no curative or protective effects and no toxicity via the oral route, reflecting their extremely low absorption from the gastrointestinal tract. Moenomycin has a very long halflife in the bloodstream, some hemolytic activity and it is not orally bioavailable. Nevertheless, moenomycins have been successfully commercialised as animal growth promoters.

Ting-Jen et al. PNAS, 105, 2, 431-436 (2008) discusses the interactions between moenomycin and the penicillin binding proteins. However, there is no discussion of how the poor bioavailability and suboptimal pharmacokinetic properties of moenomycin may be overcome.

El-Abadla N. et al., Tetrahedron 55 (1999) 699-722 discloses the combination of moenomycin A with a cyclic polypeptide molecule. The polypeptide molecule is believed to disorganise the outer membrane of the target cells, however the potential for the polypeptide to similarly disrupt a host organism’s cells and thereby be toxic to the host organism is not addressed. The toxicity of polymyxin was reported in Falagas M. E. et al. Critical Care (2006) Vol 10, No. 1, 1-13.

The listing or discussion of a prior-published document in this specification should not necessarily be taken as an acknowledgement that the document is part of the state of the art or is common general knowledge.

DESCRIPTION OF THE INVENTION

According to the invention, there is provided a series of novel compositions of matter comprising a combination of an antibacterial agent and a delivery agent. The combinations have been developed in order to allow new and existing antibacterial agents, some of which may previously have been considered to be insufficiently active in the clinic, to be useful in treating and preventing bacterial infections. The approach involves the synthesis and use of new delivery agents which are capable of binding to both the antibacterial agent and certain components of a bacterial cell, thereby acting as an anchor for the antibacterial agent and significantly increasing its effectiveness.

According to a first aspect of the invention, there is provided a combination of an antibacterial agent and a delivery agent, wherein:

  • (i) the delivery agent is a moiety that is covalently bonded to the antibacterial agent, and wherein the delivery agent is either capable of covalently bonding to one or more structures on a bacterial cell membrane or comprises a hydrophilic portion capable of otherwise binding (e.g. by way of electrostatic interactions (i.e. ionic interactions) and/or hydrogen bonding) to one or more structures on a bacterial cell membrane;
  • (ii) the delivery agent is a compound of formula I,
  • or a pharmaceutically-acceptable salt thereof, wherein A1 represents a hydrogen atom or a terminating group; D1 represents a dendrimer fragment to which the X groups shown are attached, X represents -NH2, boronic acid or a boronic acid derivative; and n is 2 or more (e.g. from 2 to 20); or
  • (iii) the delivery agent is a polypeptide or a polypeptide derivative, or a pharmaceutically-acceptable salt thereof, that comprises at least two amino acid residues selected from the group consisting of arginine and lysine, and is capable of otherwise binding to one or more structures on a bacterial cell membrane.

Such combinations are hereinafter referred to as “compositions of the invention”.

In one embodiment, the composition is a combination of an antibacterial agent and a delivery agent, wherein:

  • (i) the delivery agent is a moiety that is covalently bonded to the antibacterial agent, and wherein the delivery agent is either capable of covalently bonding to one or more structures on a bacterial cell membrane or comprises a hydrophilic portion capable of otherwise binding (e.g. by way of electrostatic interactions (i.e. ionic interactions) and/or hydrogen bonding) to one or more structures on a bacterial cell membrane;
  • (ii) the delivery agent is a compound of formula I,
  • or a pharmaceutically-acceptable salt thereof, wherein A1 represents a hydrogen atom or a terminating group; D1 represents a dendrimer fragment to which the X groups shown are attached, X represents -NH2, boronic acid or a boronic acid derivative; and n is 2 or more (e.g. from 2 to 20); or
  • (iii) the delivery agent is a polypeptide, or a pharmaceutically-acceptable salt thereof, that comprises at least two arginine residues and is capable of otherwise binding to one or more structures on a bacterial cell membrane.

For the avoidance of doubt, the delivery agent must be bound or capable of binding to the antibacterial agent, and it must also be bound or capable of binding to one or more structures on a bacterial cell membrane.

By “antibacterial agent” we mean any substance which is capable of inhibiting bacterial growth and/or reproduction, or which is capable of killing bacteria following administration to the organism in vivo or in vitro. This includes known antibiotic compounds, and particularly includes compounds for which the development of bacterial resistance has been observed in the clinic. This also includes fragments of known antibiotic compounds where those fragments are themselves capable of inhibiting bacterial growth and/or reproduction, or capable of killing bacteria, under specific conditions (e.g. once the fragments are brought into association with a target structure in or on a bacterial cell).

By “delivery agent” we mean any substance which facilitates the binding of an antibacterial agent to a portion of a bacterial cell (preferably in the region of the bacterial cell wall), and which can thereby anchor the antibacterial agent in the vicinity of the biological target (e.g. an enzyme that is important for cellular activity).

In particular embodiments of the invention, the delivery agent is covalently bonded to the antibacterial agent. In alternative embodiments, the delivery agent and antibacterial agent are discrete molecules which are capable of binding to each other via non-covalent interactions.

Unless otherwise stated, terms such as “binding”, “bound”, etc., refer to the interaction between molecules or chemical structures which serve to hold those molecules or chemical structures in close proximity to one another. In the context of the present invention, the term “binding”, unless otherwise stated, particularly refers to the binding that occurs as a result of interactions between permanent dipoles or more preferably as a result of hydrogen bonding between the molecular structures involved.

By the term “hydrophilic portion which is capable of binding to one or more structures”, we include a molecular fragment which is more soluble in water or other polar solvents (e.g. protic solvents such as alcohols) than in oil or other hydrophobic solvents (e.g. hydrocarbons). The term specifically includes any structure which is capable of binding to one or more structures in bacterial cell membranes by way of one or more hydrogen bonds and/or electrostatic interactions (i.e. ionic bonds).

Delivery Agents

Where the delivery agent is a moiety that is covalently bonded to the antibacterial agent, particular delivery agents that may be mentioned include those which are capable of binding to one or more structures on a bacterial cell membrane via the formation of one or more covalent bonds with said structures, via the formation of one or more hydrogen bonds with said structures, or through electrostatic interactions between oppositely charged regions on the delivery agent and the bacterial cell membrane (i.e. a form of ionic bonding). In particular embodiments, the delivery agents are able to bind to the bacterial cell membrane via one or more such covalent bonds, a plurality of hydrogen bonds and/or a plurality of such electrostatic interactions. For example, the delivery agent moiety may be capable of forming 1, 2, 3, 4, 5, 6, 7, 8, 9 or more separate covalent bonds or hydrogen bonds with said structures, thereby greatly enhancing the extent to which the delivery agent is anchored to the cell wall. Delivery agents which are capable of forming at least 4, at least 6, or at least 8 of such linkages (particularly hydrogen bonding linkages) are preferred.

In embodiments of the invention in which the delivery agent is covalently bonded to the antibacterial agent, as well as embodiments in which the delivery agent and antibacterial agent are not covalently bonded together, covalent bonds between the delivery agent and the one or more structures on the bacterial cell membrane may be formed, for example, where the delivery agent comprises a boronic acid component or a pharmaceutically-acceptable salt thereof. Where such boronic acids or boronic acid derivatives are present, covalent bonding may occur between the boron atoms of the delivery agent and 1,2- and 1,3-diol groups within the saccharides on the surface of the bacterial cell.

In embodiments of the invention in which the delivery agent is covalently bonded to the antibacterial agent, as well as embodiments in which the delivery agent and antibacterial agent are not covalently bonded together, hydrogen bonds between the delivery agent and the one or more structures on the bacterial cell membrane may be formed through interactions of the delivery agent with saccharides on the surface of the bacterial cell, and particularly with other structures such as phosphate groups or sulphate groups in the lipopolysachharides or phospholipids of the cell membrane. Functional groups that are capable of participating in hydrogen bonding are well known to the skilled person. Particular functional groups that may be mentioned in this respect include primary amines amidines (including guanidines) and amides (including ureas), as well as pharmaceutically-acceptable salts thereof. Still further particular functional groups that should be mentioned include primary amines, amidines, guanidines, amides and ureas (and pharmaceutically-acceptable salts thereof).

In embodiments in which the delivery agent binds to the bacterial cell wall by way of one or more electrostatic interactions (optionally in combination with one or more hydrogen bonding interactions), the delivery agent may carry a plurality of positively charged regions. Such positively charged regions are able to interact with the negatively charged phosphate groups that are present in the phospholipids and lipopolysaccharides of the cell membranes. The positively charged regions on the delivery agent may be present due to the delivery agent molecule being provided in the form of a salt, or the delivery agent may exist as a zwitterion under physiological conditions. Accordingly, positive charges may be present as a result of the reaction of a free base form of the delivery agent with an acid to form an acid addition salt. In embodiments of the invention in which the delivery agent is covalently bonded to the antibacterial agent, as well as embodiments in which the delivery agent and antibacterial agent are not covalently bonded together, particular delivery agents that may be mentioned include those which contain a plurality (e.g. at least 4, at least 6, or at least 8 charged regions) of such charged regions.

Particular delivery agents that may be mentioned therefore include those which comprise one or more functional groups selected from the list consisting of boronic acids, boronic acid derivatives, primary amines, guanidines, and pharmaceutically-acceptable acid addition salts thereof. For example, when the delivery agent is covalently bonded to the antibacterial agent, the delivery agent may comprise one or more functional groups selected from the list consisting of boronic acids, boronic acid derivatives, primary amines, guanidines, and pharmaceutically-acceptable acid addition salts thereof, and when the delivery agent is a compound of formula I, X may represent a boronic acid group, a boronic acid derivative, a primary amine, a guanidine, or a pharmaceutically-acceptable acid addition salts of any such groups.

Where the delivery agent comprises one or more primary amine groups, it is preferred that the delivery agent is provided as an acid addition salt (thus containing one or more -NH3+ groups). Particular delivery agents that may be mentioned in this respect include delivery agents which are not covalently bonded to the antibacterial agent, and which comprise a polypeptide or a polypeptide derivative, or a pharmaceutically-acceptable salt thereof.

In other embodiments of the invention, the delivery agent comprises a plurality of said functional groups, particularly where the functional groups are intended to interact with the cell membrane via electrostatic or hydrogen bonding interactions. For example, the delivery agent may comprise 2, 3, 4, 5, 6, 7, 8, 9 or more of said functional groups. Delivery agents which comprise larger numbers of such functional groups are believed to be capable of binding more strongly to the structures in the bacterial cell wall, and thereby improve the efficacy of the associated antibacterial agent.

In certain embodiments, the delivery agent comprises a dendrimer-like structure. For example, compounds of formula I contain a dendrimer fragment at the position denoted as D1. For the avoidance of doubt, in embodiments of the invention in which the delivery agent is covalently bonded to the antibacterial agent, the delivery agent may also comprise a dendrimer-like structure. The term “dendrimer” is well known in the art, and refers to structures containing a branching, tree-like architecture. Preferably, such dendrimer structures are generally acyclic (e.g. they do not contain any cyclic structures having more than 6 members (i.e. the largest ring structures that may be present in the dendrimers are 6-membered rings such as phenyl groups)) or the dendrimer structure may be completely acyclic. A preferred delivery agent is an organic molecule (or a salt thereof) containing a dendrimer fragment (i.e. a fragment containing multiple, tree-like branches), and each of the branches of that dendrimer fragment may be linked to a relevant functional group. Thus, a single delivery agent may contain a plurality of functional groups capable of binding to a bacterial cell membrane via covalent bonds, hydrogen bonds and/or electrostatic interactions along the lines described above.

Particular delivery agents that may be mentioned include those which contain a dendrimer fragment. For example, delivery agents that may be mentioned include those of formula II:

or a pharmaceutically-acceptable salt thereof, wherein A represents a hydrogen atom, a terminating group or an antibacterial agent (or a derivative thereof); D1 represents a dendrimer fragment to which the NH2 groups shown above are attached; and n1 is 2 or more (e.g. from 2 to 20).

Other delivery agents that may be mentioned include those of formulae III and IV:

or a pharmaceutically-acceptable salt thereof, wherein each A independently represents a hydrogen atom, a terminating group or an antibacterial agent (or a derivative thereof); D2 and D3 each represent a dendrimer fragment to which the groups shown in parentheses are attached; Y represents O, NH or S; and each n1 is 2 or more (e.g. from 2 to 20).

Further delivery agents that may be mentioned include those of formulae V to VII:

or a pharmaceutically-acceptable salt thereof, wherein each A independently represents a hydrogen atom, a terminating group or an antibacterial agent; each of D4 to D6 represents a dendrimer fragment to which the groups shown in parentheses are attached; Y represents O, NH or S; each n1 is 2 or more (e.g. from 2 to 20); m, p, q and r each independently represent from 1 to 8 (e.g. from 1 to 6); R1 represents a C1-6 alkyl group.

In particular embodiments of each of the compounds of formulae I to VII, n1 represents from 3 to 20, from 4 to 18, from 6 to 16 or most particularly from 8 to 16.

Dendrimer fragments D1 to D6 may be polyglycerol-based structures or may be dendron-based structures. Particular dendrimer fragments that D1 to D6 (preferably D4 to D6) may represent include those of formulae A to F:

For each of the dendrimers of formula A to E, the single wavy line on the left-hand side of the structures as shown corresponds to the point of attachment of the A, A1 or A2 (which latter group is defined below) group (i.e. the hydrogen atom, the terminating group or the antibacterial agent). The remaining wavy lines indicate the points of attachment of the plurality of amine-, X- or boronic acid-containing portions of the delivery agent (i.e. the bracketed portions in formulae I to IX).

Dendrimer F is an example of a dendrimer that is not covalently bonded to an antibacterial agent, thus all of the wavy lines shown for dendrimer F represent points of attachment of the amine-containing portions of the delivery agent. The group denoted by A1 or A in compounds of formula I to VII is incorporated in the structure for dendrimer F and so is not shown.

In compounds of formulae II to IV, particular dendrimer fragments that D1 to D3 may represent include those of formulae A to F as defined above wherein the dendrimer fragments are linked to the plurality of amine-containing portions of the delivery agent (i.e. the bracketed portions in formulae II to IV) by way of direct bonds or, preferably, additional linker groups. Additional linker groups that may be mentioned in this respect include ester linkages (i.e. -C(O)-O-), amide linkages (i.e. -C(O)-NH-), sulfonamide linkages (i.e. -S(O)2-NH-), ether linkages (i.e. -O-), amine linkages (i.e. -N(Rx)- in which Rx represents hydrogen or a C1-6 alkyl group), a C1-12 (e.g. C1-6) alkylene linkage, or a plurality of such linkages in combination. In compounds of formulae II to IV which contain multiple such additional linker groups, the additional linker groups may be the same or different.

Particular preferred delivery agents include:

  • (i) compounds of formula V in which D4 represents a dendrimer fragment of any one of formulae A to F (particularly formula C);
  • (ii) compounds of formula VI in which D5 represents a dendrimer fragment of any one of formulae A to F (particularly formulae A or B);
  • (iii) compounds of formula VII in which D6 represents a dendrimer fragment of any one of formulae A to F (particularly formulae D, E or F).

Other dendrimer structures will be known to the skilled person. For example, various dendrimers that are known to the skilled person include those disclosed in J. Mater. Chem. B 2012, 2, 2153-2167. The disclosures in that document show that such dendrimer compounds may have low toxicities.

By the use of the term “terminating group” we include any structural feature that is largely inert under physiological conditions. Particular examples of such groups include hydrogen or a C1-30 (e.g. C8-22) alkyl chain. Where the terminating group is a C1-30 (e.g. C8-22) alkyl chain, that group may optionally be interrupted by one or more heteroatoms selected from O and N (e.g. so forming an alkyl-O-alkylene- group or an alkyl-N(Ry)-alkylene- group in which Ry represents hydrogen or a C1-6 alkyl group), and/or it may be bound to the dendrimer portion via an ester linkage, an amide linkage, a sulfonamide linkage, an ether linkage, an aryl group or heteroaryl group. Particular aryl or heteroaryl groups that may be mentioned in this respect include phenyl groups, and 5- or 6-membered heterocyclic aromatic rings in which the ring contains from 1 to 3 heteroatoms selected from O, N and S. For example, the heteroaryl group may be a 1,2,3-triazole, which is advantageous as it is amenable to synthesis via relatively simple chemical processes (known as “click”-chemistry).

Other delivery agents that may be mentioned include those of formulae VIIIa, VIIIb, and IX:

wherein each A2 independently represents an antibacterial agent (or a derivative thereof); L2 represents aliphatic linker (e.g. a C1-6 alkyl chain); D7 and D8 independently represent a direct bond or a dendrimer fragment to which the boron-containing groups shown are attached; n2 is 1 or more (e.g. from 2 to 20); and optionally wherein D7and D8 are attached to the boronic acid portions of the compound of formula VIIIa and IX, or boric acid portions of the compound of formula Vlllb, via a linker group. Thus, where the delivery agent is a compound of formula VIIIa, Vlllb or IX, it is covalently bonded to the antibacterial agent.

In embodiments of compounds of formulae VIIIa, Vlllb and IX, the dendrimer fragments that are represented by D7 and D8 may be polyglycerol-based structures or dendron-based structures as defined above in respect of dendrimer fragments D1 to D6. Similarly, particular dendrimer fragments that D7 and D8 may represent include those of formulae A to E as defined above.

In compounds of formulae VIIIa, Vlllb and IX, the linker group that may be present may comprise one or more groups selected from the list comprising C1-10 alkyl, -NH-, -O-, -C(O)-O-, and -C(O)-NH- (wherein the amide and ester linkers may each be attached in either of the two possible orientations). For example, the linker group may be a -(CH2)2-NH-(CH2)8-C(O)-NH- group.

Particular delivery agents that may be mentioned include those which contain a dendrimer fragment. That is, particular delivery agents that may be mentioned include compounds of formulae II, VIIIa, Vlllb and IX, or pharmaceutically-acceptable salts thereof (e.g. compounds of formulae III, IV, VIIIa, Vlllb and IX or pharmaceutically-acceptable salts thereof, or most preferably compounds of formulae V, VI, VII, VIIIa, VIIIb and IX or pharmaceutically-acceptable salts thereof).

Unless otherwise specified, alkyl groups and alkoxy groups as defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of three) of carbon atoms, be branched-chain and/or cyclic. Further, when there is a sufficient number (i.e. a minimum of four) of carbon atoms, such alkyl and alkoxy groups may also be part cyclic/acyclic. Such alkyl and alkoxy groups may also be saturated or, when there is a sufficient number (i.e. a minimum of two) of carbon atoms, be unsaturated. Unless otherwise specified, alkyl and alkoxy groups may also be substituted by one or more halo, and especially fluoro, atoms.

Unless otherwise specified, alkylene groups as defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of two) of carbon atoms, be branched-chain. Such alkylene chains may also be saturated or, when there is a sufficient number (i.e. a minimum of two) of carbon atoms, be unsaturated. Unless otherwise specified, alkylene groups may also be substituted by one or more halo atoms.

The term “aryl”, when used herein, includes C6-10 aryl groups such as phenyl, naphthyl and the like. When substituted, aryl groups are preferably substituted by between one and three substituents.

When used herein, the term “heteroaryl group” includes 4- to 12-membered (e.g. 5- to 10-membered) cyclic aromatic groups containing one or more heteroatoms selected from N, O and S. The term therefore includes such groups that are mono- or bicyclic. Preferred heterocyclic groups include groups such as pyrrolyl, furanyl, thienyl, imidazolyl, pyrrazolyl, thiazolyl, oxazolyl, triazolyl (e.g. 1,2,3-triazolyl), benzoxazolyl and pyridyl. Particularly preferred heterocyclic groups include pyrrolyl, imidazolyl, 1,2,3-triazolyl, and oxazolyl.

The term “halo”, when used herein, includes fluoro, chloro, bromo and iodo.

Pharmaceutically-acceptable salts that may be mentioned include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a delivery agent with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a delivery agent in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.

Examples of pharmaceutically acceptable addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric and sulphuric acids; from organic acids, such as tartaric, acetic, trifluoroacetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic and arylsulphonic acids; and from metals such as sodium, magnesium, or preferably, potassium and calcium. Particularly preferred salts include those derived from acetic, trifluoroacetic hydrochloric and tartaric acids.

Certain delivery agents disclosed herein may be novel and so may be of use in the treatments disclosed herein. Thus in a second aspect of the invention, there is provided a delivery agent as defined above. That is, the delivery agent may be a compound of any one of formulae I to IX as defined hereinbefore, or a pharmaceutically-acceptable salt thereof.

In particular embodiments of this aspect of the invention, the delivery agent is not covalently bonded to an antibacterial agent (but may be used in combination with an antibacterial agent in order to achieve the effects described herein).

In a first embodiment in which the delivery agent is not covalently bonded to an antibacterial agent, the delivery agent is a compound of formula I to VII, or a pharmaceutically-acceptable salt thereof, wherein D1 to D6, m, n, p, q, r and R1 are as hereinbefore defined and A, if present, represents A1 as hereinbefore defined.

In a second embodiment in which the delivery agent is not covalently bonded to an antibacterial agent, the delivery agent is a compound of formulae I to VII, or a pharmaceutically-acceptable salt thereof, wherein m, n, p, q, r and R1 are as hereinbefore defined, A represents A1 as hereinbefore defined, and D1 to D6 each represent a dendrimer fragment of formula A to F as hereinbefore defined.

In a third embodiment in which the delivery agent is not covalently bonded to an antibacterial agent, the delivery agent is a compound of formulae V to VII, as hereinbefore defined, wherein m, n, p, q, r and R1 are as hereinbefore defined, A represents A1 as hereinbefore defined, and D1 to D6 each represent a dendrimer fragment of formula A to F as hereinbefore defined (e.g. wherein, in the compounds of formulae VIIIa, Vlllb and IX, the optional linkers between the D7/D8 groups and the boronic acid or boric acid groups are absent).

In a fourth embodiment in which the delivery agent is not covalently bonded to an antibacterial agent, the delivery agent is a polypeptide or a polypeptide derivative, or a pharmaceutically-acceptable salt thereof. It is preferred (though not essential) that, when the delivery agent is a polypeptide or polypeptide derivative, or a pharmaceutically-acceptable salt thereof, then the polypeptide contains at least two residues selected from the group consisting of arginine and lysine. The amino acid residues may be provided in their naturally occurring stereochemical configuration (e.g. the L-configuration), or the alternative stereochemical configuration. It is preferred that the amino acid residues are provided in their naturally occurring stereochemical configuration.

In embodiments in which the delivery agent is a polypeptide, a polypeptide derivative or a pharmaceutically-acceptable salt thereof (e.g. a polypeptide, or a pharmaceutically-acceptable salt thereof), preferably the polypeptide contains at most 20 (e.g. no more than 15) amino acid residues. Particular polypeptides that may be mentioned include acyclic polypeptides (e.g. acyclic polypeptides containing at most 20 amino acid residues). For example, polypeptides of particular interest include those which contain at least four, at least six or at least eight arginine residues. In all cases, the term the amino acids that form the polypeptides that may be used in the delivery agents may be in either the D or L forms.

Other polypeptides and polypeptide derivatives of particular interest include compounds containing a sequence of at most 20 (e.g. between 5 and 15) amino acid residues. Polypeptide derivatives include polypeptide compounds which contain non-peptide moieties at one or both ends of the peptide chain. Alternatively or additionally, polypeptide derivatives include polypeptide compounds in which one or more of the amino acids is optionally provided in a chemically modified form. Examples of such modifications include replacing one or more -NH2 groups on side chains on said amino acids (e.g. the side chains of lysine or arginine) with amides and derivatives thereof (e.g. amides, ureas, thioamides or thioureas).

Particular polypeptides and polypeptide derivatives that may be mentioned in this respect include compounds containing a polypeptide sequence selected from the list consisting of formulae XX to XL:

RRRRRRRR XX KPLIYLLRLRGQF XXI KPLIYLLLRRGQF XXII KRRKRRKRR XXIII KRRRRRR XXIV PLIYLKLLKGQF XXV rrrrrrr XXVI PLIYLLGRR XXVII PLIYLLRGR XXVIII PLIYLLKGR XXIX PLIYLLRGK XXX PLIYLLKGK XXXI PLIYLKLLK XXXII KKKKKR XXXIII RRRR XXXIV RRRRRRRRR XXXV RRRRRRRRRR XXXVI KKRRRRRRRR XXXVII KKRKKKKR XXXVIII RRWWRRWRR XXXIX PLIYLRLLRGQF XL

In a further embodiment, the polypeptide derivative may consist of one amino acid selected from the group consisting of arginine and lysine.

In each case, either the N-terminus or the C-terminus, or both, may be modified, e.g. one or both ends of the polypeptide may be modified with a PEG group, an azide, an alkyne group, an alkyl group, an -OH group, a -C(O)-NH2 group or a thiol. For example, the -C(O)OH at the C-terminus may be replaced with a -C(O)-C=CH group. As a further example, the N-terminus may be modified by the incorporation of a dye label (e.g. a TAMRA dye label) or a -C(O)-NH2 group.

Polypeptides and polypeptide derivatives which contain higher positive charge have been found to have increased effectiveness in enhancing the antibacterial potential (i.e. reducing the minimum inhibitory concentration) of existing antibacterial agents when the two agents are provided in combination. Therefore, in a preferred embodiment, the polypeptide or polypeptide derivative is a polypeptide-containing compound that bears a positive charge of at least 3 units. Particularly preferred embodiments include those in which the polypeptide or polypeptide derivative is a polypeptide-containing compound that bears a positive charge of at least 5 (e.g. at least 6) units.

The positive charge may be nominally determined by counting the number of lysine and arginine amino acids that are present in the polypeptide (each such amino acid providing one unit of positive charge). Other amino acids having side chains that are positively charged may also be mentioned in this respect.

Other polypeptides that may be used in the present invention include disulphide-based polypeptides. Examples of such polypeptides include poly-CBA peptides (e.g. as disclosed in Son et al. Accounts of Chemical Research, (2012) Vol 45, No. 7, 1100-1112 (see, in particular, poly(CBA-DAH) modified with arginine residues (poly(CBA-DAH-R), and poly(CBA-DAH) containing RGD peptide)).

Polypeptide delivery agents may be bound to the antibacterial agent, or alternatively may be provided as a separate agent. Salts (e.g. acid addition salts) of such polypeptide-based delivery agents are of particular interest.

Examples of polypeptide-based delivery agents which may be (covalently) bound to an antibacterial agent include delivery agents containing a polypeptide sequence selected from the list consisting of formulae XX to XXXII, as defined above. Particular such delivery agents include those containing a polypeptide sequence selected from the list consisting of formulae XX, XXIII and XXXIII. The polypeptide-based delivery agent may be bound to the antibacterial agent by any one of the structural linkages discussed hereinafter.

Without wishing to be bound by theory, it is believed that the delivery agents disclosed herein are able to facilitate the binding of an antibacterial agent close to the cell membrane in a bacterial cell due to the presence of functional groups on the delivery agent which are capable of hydrogen bonding to the cell wall, or bonding through electrostatic interactions. The delivery agent may be able to mask certain negative charges that may exist on the antibacterial fragment. For example, moenomycin derivatives contain a phosphate group and a carboxylate group, either of which may dissociate into anionic species under physiological conditions. The presence of similar charges on phosphate groups that are present in bacterial cell wall phospholipids and lipopolysaccharides (particularly in Gram negative bacteria) may prevent the antibacterial from entering the cell or approaching the target region.

It is also believed that the delivery agents disclosed herein are capable of reducing the extent to which certain antibacterials bind to blood plasma, thereby increasing the bioavailability of those antibacterials.

Antibacterial Agents

Antibacterial Agents that are of use in the context of the present invention are chemical agents that are bactericidal or bacteriostatic. These include known and naturally occurring antibacterial agents (include synthetic and semi-synthetic antibacterial agents), as well as fragments of those agents where those fragments retain at least a portion of the bactericidal or bacteriostatic activity.

Particular antibacterial agents that may be mentioned include those in which the agent is a molecule or fragment that inhibits the synthesis or repair of bacterial cell walls.

Other particular antibacterial agents that may be mentioned include those in which the agent is a molecule or fragment that modulates the activity of a penicillin binding protein. More particular antibacterial agents include those in which the agent is a molecule or fragment that inhibits a glycosyltransferase enzyme in a bacterium (e.g. a glycosyltransferase enzyme in a penicillin binding protein). In this context, compounds that inhibit a glycosyltransferase enzyme are capable of reducing the formation of peptidoglycan bonds in a bacterial cell, particularly reducing the formation of peptidoglycan bonds that are formed during bacterial cell wall synthesis.

Other antibacterial agents that may be mentioned include those which comprise a glycopeptide or a phosphoglycolipid fragment.

Most particular antibacterial agents that may be mentioned include moenomycin (including moenomycin A), vancomycin, β-lactam antibiotics, and derivatives thereof. The structure of moenomycin A is shown in FIG. 1.

In some embodiments of the invention, the antibacterial agent is covalently bonded to the delivery agent.

Where the antibacterial agent is moenomycin, or a derivative thereof, the delivery agent may be bonded to the moenomycin at any position on the molecular skeleton. For example, the moenomycin, or derivative thereof, may be bound to the delivery agent:

  • (i) such that the delivery agent (and any associated linker) replaces part or all of the 2-amido-cyclopentane-1 ,3-dione portion of the moenomycin; or particularly,
  • (ii) via the moenocinol portion of the moenomycin;
  • (iii) such that the delivery agent (and any associated linker) replaces part or all of the moenocinol portion of the moenomycin; or
  • (iv) via the 2-amino-cyclopentane-1,3-dione portion of the moenomycin.

Particular delivery agents that may be mentioned in this respect include delivery agents of formulae II to IX, or pharmaceutically-acceptable salts thereof, wherein D1 to D8, m, n, p, q, r and R1 are as hereinbefore defined and A and A2 each represent moenomycin or a derivative thereof.

Further delivery agents that may be mentioned in this respect include delivery agents of formulae II to IX, or pharmaceutically-acceptable salts thereof, wherein m, n, p, q, r and R1 are as hereinbefore defined, A and A2 each represents moenomycin or a derivative thereof, and D1 to D8 each represent a dendrimer fragment of formula A to E as hereinbefore defined.

Still further delivery agents that may be mentioned in this respect include delivery agents of formulae V to IX, or pharmaceutically-acceptable salts thereof, wherein m, n, p, q, r and R1 are as hereinbefore defined, A and A2 each represents moenomycin or a derivative thereof, and D1 to D8 each represent a dendrimer fragment of formula A to E as hereinbefore defined (e.g. wherein, in the compounds of formulae VIIIa, VIIIb and IX, the optional linkers between the D7/D8 groups and the boric acid or boronic acid groups are absent).

Still further delivery agents that may be mentioned in this respect include delivery agents which contain a polypeptide sequence as defined above bound to the antibacterial agent. In one embodiment, the delivery agent is an agent which contains a polypeptide sequence selected from the list consisting of formulae XX to XXXII as defined above (and, in particular, a polypeptide sequence selected from the list consisting of formulae XX, XXIII and XXXIII).

Polypeptide-based delivery agents may be bound to the antibacterial agent either directly or via a linker group. Suitable linker groups include any structural fragments that would be known to the skilled person. Such linker groups may include one or more fragments selected from the list comprising an optionally substituted alkyl chain, a carbonyl group, an amide group, an amine group, an ether group, and an optionally substituted aromatic (e.g, a phenyl or heteroaromatic ring).

Particular linkers that may be mentioned include linkers selected from formulae LA and LB:

wherein the wavy line on the left-hand side represents the point of attachment of the linker to the antibacterial agent, and the wavy line on the right-hand side represents the point of attachment of the linker to the polypeptide-based delivery agent (typically at the C-terminal end), and wherein X represents —O—, a C1—12 alkyl group or a polyethylene glycol group (e.g. a group containing —(CH2CH2O)n— wherein n is from 1 to 10 (such as from 2 to 5)).

When such linkers are used to join a polypeptide-based delivery agent to moenomycin, it is preferred that the triazole ring is bound to the moenomycin in place of the 2-amido-cyclopentane-1,3-dione constituent of moenomycin.

Covalent conjugates of moenomycin with in combination with particular polypeptide-based delivery agents and linkers have been found to exhibit an increased synergistic effect when used to combat particular pathogens. For example, an antibacterial agent containing moenomycin bound to a polypeptide sequence of formula XX via a linker of formula LA or LB (preferably LB) in which X represents a C1-6 alkyl-C(O)—NH—C1-6 alkyl chain is particularly effective when used to treat P. aeruginosa or K. pneumoniae.

An antibacterial agent containing moenomycin bound to a polypeptide sequence of formula XXXIII via a linker of formula LB in which X represents a —(CH2CH2O)2—(CH2)2—NH—C(O)—(CH2)2—C(O)— group is particularly effective when used to treat P. aeruginosa or A. baumannii.

An antibacterial agent containing moenomycin bound to a polypeptide sequence of formula XXIII via a linker of formula LB in which X represents a C5 alkyl chain is particularly effective when used to treat K. pneumoniae or A. baumannii.

Antibacterial agents containing moenomycin bound to a polypeptide sequence of formula XXXIII via a linker of formula LB in which X represents a direct bond, —O— or a C5 alkyl chain is also particularly effective when used to treat A. baumannii.

The antibacterial agents and delivery agents may exhibit tautomerism. All tautomeric forms and mixtures thereof are included within the scope of the invention.

The antibacterial agents and delivery agents may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism. Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation. The various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional (e.g. fractional crystallisation or HPLC) techniques. Alternatively the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation, or by derivatisation, for example with a homochiral acid followed by separation of the diastereomeric esters by conventional means (e.g. HPLC, chromatography over silica). All stereoisomers are included within the scope of the invention.

Particular embodiments of the invention that may be mentioned include the compounds of the Examples disclosed hereinafter.

PREPARATION

Delivery agents (including compounds of formulae I to IX) may be prepared in accordance with techniques known to those skilled in the art, for example as described hereinafter.

Thus, according to a third aspect of the invention there is provided a process for the preparation of a compound of formula I (or a compound of formula II, III, IV, V, VI, VII, VIIIa, VIIIb or IX, as appropriate), which comprises:

  • (a) for compounds of formula I in which X represents —NH2 and A1, D1 and n are as hereinbefore defined, or for compounds of formula II as hereinbefore defined, reaction of a compound of formula X:
  • wherein A, D1 and n are as defined hereinbefore and Ra1 represents a C1-12 alkyl group, with an acid (such as hydrochloric acid, trifluoroacetic acid or p-toluenesulfoic acid), for example under conditions known to those skilled in the art (such as in the presence of a suitable organic solvent (e.g. dioxane, THF, MeCN, diethyl ether, EtOAc, DCM or DMF));
  • (b) for compounds of formula IV wherein Y represents NH, reaction of a compound of formula I wherein X represents —NH2, and A1, D1 and n are as hereinbefore defined, or a compound of formula II as hereinbefore defined, with a compound of formula XI,
  • wherein L1 represents a suitable leaving group (such as an aryl group or a heteroaryl group (e.g. a pyrazole group)) under conditions known to those skilled in the art (such as in the presence of a suitable organic solvent (e.g. methanol, ethanol, MeCN, THF) and, optionally, in the presence of a suitable base (e.g. Et3N, pyridine, DMAP or DIPEA);
  • (c) for compounds of formula VII, reaction of a compound of formula XII,
  • wherein A, D6 and n1 are as defined hereinbefore, by an amide coupling reaction, i.e. the formation of an amide from a carboxylic acid, with a compound of formula XIII,
  • wherein r and Y are as defined hereinbefore, which reaction may be performed in the presence of a suitable coupling reagent (e.g. 1,1′-carbonyldiimidazole, N,N’-dicyclohexylcarbodiimide, N,N′-diisopropylcarbodiimide, 1-hydroxy-7-azabenzotriazole (HOAt), or the like) or, alternatively the —C(O)OH group may first be activated to the corresponding acyl halide (e.g —C(O)Cl, by treatment with oxalyl chloride, thionyl chloride, phosphorous pentachloride, phosphorous oxychloride, or the like) under standard conditions known to those skilled in the art (e.g. optionally in the presence of a suitable solvent, suitable base and/or in an inert atmosphere);
  • (d) for compounds of formula VII, reaction of a compound of formula XII in which the carboxylic acid group(s) is(are) provided as an ester derivative, with a compound of formula XIII, in the presence of, e.g. trimethylaluminium, for example in an inert atmosphere and in the presence of a suitable solvent (e.g. dichloromethane); or
  • (e) for compounds of formula III, in which the amidine (—C(═NH)—NH2) group is linked to D2 via a linker comprising a —CH2—NH— group, reaction of a compound of formula XIV,
  • wherein A, D2 and n1 are as defined hereinbefore, by reductive amination using a compound of formula XV,
  • wherein Q represents an appropriate linker group (such as a linker containing one or more groups selected from C1-6 alkylene, —NH—, —C(O)—NH, —C(O)—O— and —O), under appropriate reaction conditions, for example in a “one-pot” procedure in the presence of an appropriate reducing agent, such as a chemoselective reducing agent such as sodium cyanoborohydride or, preferably, sodium triacetoxyborohydride, or the like. Alternatively, such reactions may be performed in two steps, for example a condensation step (in the presence of e.g. a dehydrating agent such as trimethyl orthoformate or MgSO4 or molecular sieves, etc) followed by a reduction step (e.g. by reaction in the presence of a reducing agent such as a chemoselective one mentioned above or NaBH4, AlH4, or the like), for instance the conversion of -NH2 to -N(H)-isopropyl by condensation in the presence of acetone (H3C—C(O)—CH3) followed by reduction in the presence of a reducing agent such as sodium cyanoborohydride (i.e. overall a reductive amination).

Compounds of formulae III, V and VI may be prepared by processes analogous to process (a) above for compounds of formulae I or II, using appropriate starting materials (i.e. appropriate compounds of formula X in which the D1 group contains the relevant linker groups that are attached to the requisite —NH2 groups).

Compounds of formulae VIIIa, Vlllb and IX may be made, for example, by processes analogous to processes (c) to (e) above, e.g. via amide formation reactions or reductive amination processes. An example of a process analogous to process (e) above includes the reaction of a compound of formula XVI,

with a compound of formula XVII,

wherein A2, D8 and n2 are as hereinbefore defined under reductive amination conditions, for example as described above.

Other specific transformation steps (including those that may be employed in order to form compounds of formulae I to IX) that may be mentioned include:

  • (i) reductions, for example of a carboxylic acid (or ester) to either an aldehyde or an alcohol, using appropriate reducing conditions (e.g. -C(O)OH (or an ester thereof), may be converted to a —C(O)H or —CH2—OH group, using DIBAL and LiAlH4, respectively (or similar chemoselective reducing agents));
  • (ii) reductions of an aldehyde (—C(O)H) group to an alcohol group (—CH2OH), using appropriate reduction conditions such as those mentioned in point (i) above;
  • (iii) oxidations, for example of a moiety containing an alcohol group (e.g. —CH2OH) to an aldehyde (e.g. —C(O)H) or of a —S— moiety to a —S(O)— or —S(O)2— moiety (or the reverse reduction reaction), for example in the presence of a suitable oxidising agent, e.g. MnO2 or mcpba or the like;
  • (iv) conversion of a primary amide to a nitrile functional group, for example under dehydration reaction conditions, e.g. in the presence of POCl3, or the like;
  • (v) nucleophilic substitution (e.g. aromatic nucleophilic substitution) reactions, where any nucleophile replaces a leaving group, e.g. an amine may replace a -S(O)CH3 leaving group;
  • (vi) transformation of a methoxy group to a hydroxy group, by reaction in the presence of an appropriate reagent, such as boron fluoride-dimethyl sulfide complex or BBr3 (e.g. in the presence of a suitable solvent such as dichloromethane);
  • (vii) alkylation, acylation or sulfonylation reactions, which may be performed in the presence of base and solvent (such as those described hereinbefore);
  • (viii) specific deprotection steps, such as deprotection of a hydroxy group protected as a silyl ether (e.g. a tert-butyl-dimethylsilyl protecting group) by reaction with a source of fluoride ions, e.g. by employing the reagent tetrabutylammonium fluoride (TBAF).

Compounds of formulae X, XII, XIV and XVII can be prepared by methods analogous to those described above together with other reactions that are known to those skilled in the art.

Compounds of formulae I to XVII which contain dendrimer structures may be made according to methods known in the art, for example according to the methods described in J. Mater. Chem. B 2012, 2, 2153-2167.

Salts of compounds of formulae I to IX (and salts of other compounds) may be formed by conventional means, for example by reaction of a free acid or a free base form of a corresponding compound of formula I to IX with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.

Compounds of formula I (etc.) may be isolated from their reaction mixtures using conventional techniques. For example, compounds of formula I (etc.) may be isolated by conversion to an acid (e.g. hydrochloric acid) salt (e.g. by way of addition of acid to the crude product) and then recrystallisation of the salt from a suitable solvent (e.g. methanol or, particularly, ethanol). Alternatively, the salt can simply be washed with or slurried in the presence of such a suitable solvent in order to isolate the pure acid salt of the compound of formula I.

It will be appreciated by those skilled in the art that in the processes described above and hereinafter the functional groups of intermediate compounds may need to be protected by protecting groups.

Functional groups that it is desirable to protect include hydroxy, amino and carboxylic acid groups. Suitable protecting groups for hydroxy include optionally substituted and/or unsaturated alkyl groups (e.g. methyl, allyl, benzyl or tert-butyl), trialkylsilyl or diarylalkylsilyl groups (e.g. t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl) and tetrahydropyranyl. Suitable protecting groups for carboxylic acids include C1—6 alkyl or benzyl esters.

The protection and deprotection of functional groups may take place before or after coupling, or before or after any other reaction in the above-mentioned schemes.

Protecting groups may be removed in accordance with techniques that are well known to those skilled in the art and as described hereinafter.

Persons skilled in the art will appreciate that, in order to obtain compounds of formula I (etc.) in an alternative, and, on some occasions, more convenient, manner, the individual process steps mentioned hereinbefore may be performed in a different order, and/or the individual reactions may be performed at a different stage in the overall route (i.e. substituents may be added to and/or chemical transformations performed upon, different intermediates to those mentioned hereinbefore in conjunction with a particular reaction). This may negate, or render necessary, the need for protecting groups.

The type of chemistry involved will dictate the need, and type, of protecting groups as well as the sequence for accomplishing the synthesis.

The use of protecting groups is fully described in “Protective Groups in Organic Chemistry”, edited by J W F McOmie, Plenum Press (1973), and “Protective Groups in Organic Synthesis”, 3rd edition, T.W. Greene & P.G.M. Wutz, Wiley-Interscience (1999).

Protected derivatives of compounds of formula I (etc.) may be converted chemically to compounds of the invention using standard deprotection techniques (e.g. hydrogenation). The skilled person will also appreciate that certain compounds of formula I (etc.) may also be referred to as being “protected derivatives” of other compounds of formula I.

Those skilled in the art will also appreciate that certain compounds of formula I (etc.) will be useful as intermediates in the synthesis of certain other compounds of formula I.

Antibacterial agents may be commercially available, or may be synthesised according to known procedures. In many cases, antibacterial agents may be available from biological sources.

For example, moenomycin is produced by at least four streptomycete strains, Streptomycesghanaensis (ATCC14672), S.bambergiensis (ATCC13879), S.ederensis (ATCC15304) and S.geysiriensis (ATCC15303). The composition of the flavomycin complex produced by S.ghanaensis has been thoroughly studied. As a result, a number of phosphoglycolipids have been isolated and structures of moenomycins A, A12, C1, C3 and C4 have been determined. (Ostash B. et al. Nat. Prod. Rep., 2010, 27, 1594-1617). Moenomycin may be obtained by the processes disclosed in M. Adachi et al., J. Am. Chem. Soc. 2006, 128, 14012-14013.

Moenomycin may alternatively be synthesised by bench chemistry (J. G. Taylor, et al., J. Am. Chem. Soc., 2006, 128, 15084; J. G. Taylor, Ph.D. Thesis, Harvard University, 2006; and P. Welzel, Angew. Chem., Int. Ed., 2007, 46, 4825).

Antibacterial agents (whether naturally occurring or not) may be derivatised by way of chemical methods that are known to the skilled person.

For example, the cyclopentyl ring in moenomycin may be derivatised or replaced according to the procedures discussed in Ostash B. et al. ibid., and references cited therein. In particular, the antibacterial may be treated with an aryl diazonium salt, via a Japp-Klingemann reaction, to produce various substituted triazole moieties.

The lipid moiety in moenomycin (which moiety is known as moenocinol) may also be modified, removed or replaced according to methods known to the skilled person (e.g. those described in Ostash B. et al. ibid., and references cited therein). For example, moenomycin (optionally in a protected form) may be ozonised to give an aldehyde of formula XVIII,

wherein Rz represents the carbohydrate portion of the molecule (or a derivative thereof), thereby removing the majority of the lipid portion of the molecule.

The compound of formula XVIII may then be further modified, for example by reductive amination of the newly formed aldehyde moiety (e.g. along the lines of reaction (e) above), to give the antibacterial agents that are useful in the compositions of the invention.

USES AND PHARMACEUTICAL PREPARATIONS

Compositions of the invention are useful because they possess pharmacological activity. They are therefore indicated as pharmaceuticals.

Thus, according to a fourth aspect of the invention there is provided the compositions of the invention for use as pharmaceuticals.

In particular, compositions of the invention may bind to certain enzymes located in the cell walls of bacteria, thereby inhibiting the activity of those enzymes. Enzymes that may be mentioned in this respect include those necessary for synthesis and repair of bacterial cell walls (e.g. penicillin binding proteins), thus providing the effect of inhibiting bacterial growth, survival and reproduction. Particular enzymes of note are those involved in bond formation in peptidoglycan (a polymer component of bacterial cell walls).

In this respect, fifth, sixth, seventh and eighth aspects of the invention provide, respectively:

  • (a) the use of a composition of the invention, as hereinbefore defined, for the preparation of a medicament for the treatment or prevention of a bacterial infection;
  • (b) a method of treating or preventing a bacterial infection in a subject, the method comprising administering to said subject an antibacterially effective amount of a composition of the invention, as hereinbefore defined;
  • (c) a composition of the invention, as hereinbefore defined, for use in treating or preventing a bacterial infection;
  • (d) use (e.g. ex vivo use) of a composition of the invention to kill bacteria.

When used herein, the terms “ bacteria” (and derivatives thereof, such as “ bacterialinfection”) includes references to organisms (or infections due to organisms) of the following classes and specific types:

  • Gram-positive cocci, such as
    • Staphylococci (e.g. Staph. aureus, Staph. epidermidis, Staph. saprophyticus, Staph. auricularis, Staph. capitiscapitis, Staph. c. ureolyticus, Staph.caprae, Staph.cohniicohnii, Staph. c. urealyticus, S taph.equorum, Staph.gallinarum, Staph.haemolyticus, Staph.hominishominis, Staph. h. novobiosepticius, Staph.hyicus, Staph.intermedius, Staph.lugdunensis,Staph.pasteuri, Staph.saccharolyticus, Staph.schleiferischleiferi, Staph. s. coagulans, Staph.sciuri, Staph.simulans, Staph.warneri and Staph. xylosus) and
    • Streptococci (e.g. beta-haemolytic, pyogenic streptococci (such as Strept.agalactiae, Strept.canis, Strept.dysgalactiaedysgalactiae, Strept.dysgalactiaeequisimilis, Strept.equiequi, Strept.equizooepidemicus, Strept.iniae, Strept.porcinus and Strept.pyogenes), microaerophilic, pyogenic streptococci (Streptococcus “milleri”, such as Strept.anginosus, Strept.constellatusconstellatus, Strept.constellatuspharyngidis and Strept.intermediusy), oral streptococci of the “mitis” (alpha-haemolytic - Streptococcus “viridans”, such as Strept.mitis, Strept.oralis, Strept.sanguinis, Strept.cristatus, Strept.gordonii and Strept.parasanguinis), “salivarius” (non-haemolytic, such as Strept.salivarius and Strept.vestibularis) and “mutans” (tooth-surface streptococci, such as Strept.criceti, Strept.mutans, Strept.ratti and Strept.sobrinus) groups, Strept.acidominimus, Strept.bovis, Strept.faecalis, Strept.equinus, Strept.pneumoniae and Strept.suis, or Streptococci alternatively classified as Group A, B, C, D, E, G, L, P, U or V Streptococcus);
  • Gram-negative cocci, such as Neisseriagonorrhoeae, Neisseriameningitidis, Neisseriacinerea, Neisseriaelongata, Neisseriaflavescens, Neisserialactamica, Neisseriamucosa, Neisseriasicca, Neisseriasubflava and Neisseriaweaveri;
  • Bacillaceae, such as Bacillusanthracis, Bacillussubtilis, Bacillusthuringiensis, Bacillusstearothermophilus and Bacilluscereus;
  • Enterobacteriaceae, such as
    • Escherichiacoli,
    • Enterobacter (e.g. Enterobacteraerogenes, Enterobacteragglomerans and Enterobactercloacae)
    • Citrobacter (such as Citrob.freundii and Citrob.divernis),
    • Hafnia (e.g. Hafniaalvei),
    • Erwinia (e.g. Erwiniapersicinus),
    • Morganellamorganii,
    • Salmonella ( Salmonellaenterica and Salmonellatyphi),
    • Shigella (e.g. Shigelladysenteriae, Shigella flexneri, Shigellaboydii and Shigellasonnei),
    • Klebsiella (e.g. Klebs.pneumoniae, Klebs. oxytoca, Klebs.ornitholytica, Klebs.planticola, Klebs.ozaenae, Klebs.terrigena, Klebs.granulomatis ( Calymmatobacteriumgranulomatis) and Klebs.rhinoscleromatis),
    • Proteus (e.g. Pr.mirabilis, Pr.rettgeri and Pr.vulgaris),
    • Providencia (e.g. Providenciaalcalifaciens, Providenciarettgeri and Providenciastuartii),
    • Serratia (e.g. Serratiamarcescens and Serratialiquifaciens), and
    • Yersinia (e.g. Yersiniaenterocolitica, Yersiniapestis and Yersiniapseudotuberculosis);
  • Enterococci (e.g. Enterococcusavium, Enterococcuscasseliflavus, Enterococcuscecorum, Enterococcusdispar, Enterococcusdurans, Enterococcusfaecalis, Enterococcusfaecium, Enterococcusflavescens, Enterococcusgallinarum, Enterococcushirae, Enterococcusmalodoratus, Enterococcusmundtii, Enterococcuspseudoavium, Enterococcusraffinosus and Enterococcussolitarius);
  • Helicobacter (e.g. Helicobacterpylori, Helicobactercinaedi and Helicobacterfennelliae);
  • Acinetobacter (e.g. A.baumanii, A.calcoaceticus, A.haemolyticus, A.johnsonii, A.junii, A.Iwoffi and A.radioresistens);
  • Pseudomonas (e.g. Ps.aeruginosaPs.maltophilia, ( Stenotrophomonasmaltophilia), Ps.alcaligenes, Ps.chlororaphis, Ps.fluorescens, Ps.luteola, Ps.mendocina, Ps.monteilii, Ps.oryzihabitans, Ps.pertocinogena, Ps.pseudalcaligenes, Ps.putida and Ps.stutzeri);
  • Bacteriodesfragilis;
  • Peptococcus (e.g. Peptococcusniger);
  • Peptostreptococcus;
  • Clostridium (e.g. C.perfringens,C.difficile,C.botulinum,C.tetani,C.absonum,C.argentinense,C.baratii,C.bifermentans,C.beijerinckii,C.butyricum,C.cadaveris,C.carnis,C.celatum,C.clostridioforme,C.cochlearium,C.cocleatum,C. fallax, C.ghonii,C.glycolicum,C.haemolyticum,C.hastiforme,C.histolyticum,C.indolis,C.innocuum,C.irregulare,C.leptum,C.limosum,C.malenominatum,C.novyi,C.oroticum,C.paraputrificum,C.piliforme,C.putrefasciens,C.ramosum,C.septicum,C.sordelii,C.sphenoides,C.sporogenes,C.subterminale,C.symbiosumandC.tertium);
  • Mycoplasma (e.g. M.pneumoniae,M.hominis,M.genitaliumandM.urealyticum);
  • Mycobacteria (e.g. Mycobacteriumtuberculosis,Mycobacteriumavium,Mycobacteriumfortuitum,Mycobacteriummarinum,Mycobacteriumkansasii,Mycobacteriumchelonae,Mycobacteriumabscesses,Mycobacteriumleprae,Mycobacteriumsmegmitis,Mycobacteriumafricanum,Mycobacteriumalvei,Mycobacteriumasiaticum,Mycobacteriumaurum,Mycobacteriumbohemicum,Mycobacteriumbovis,Mycobacteriumbranderi,Mycobacteriumbrumae,Mycobacteriumcelatum,Mycobacteriumchubense,Mycobacteriumconfluentis,Mycobacteriumconspicuum,Mycobacteriumcookii,Mycobacteriumflavescens,Mycobacteriumgadium,Mycobacteriumgastri,Mycobacteriumgenavense,Mycobacteriumgordonae,Mycobacteriumgoodii,Mycobacteriumhaemophilum,Mycobacteriumhassicum,Mycobacteriumintracellulare,Mycobacteriuminterjectum,Mycobacteriumheidelberense,Mycobacteriumlentiflavum,Mycobacteriummalmoense,Mycobacteriummicrogenicum,Mycobacteriummicroti,Mycobacteriummucogenicum,Mycobacteriumneoaurum,Mycobacteriumnonchromogenicum,Mycobacteriumperegrinum,Mycobacteriumphlei,Mycobacteriumscrofulaceum,Mycobacteriumshimoidei,Mycobacteriumsimiae,Mycobacteriumszulgai,Mycobacteriumterrae,Mycobacteriumthermoresistabile, Mycobacterium triplex, Mycobacteriumtriviale,Mycobacteriumtusciae,Mycobacteriumulcerans,Mycobacteriumvaccae,MycobacteriumwolinskyiandMycobacterium xenopi);
  • Haemophilus (e.g. Haemophilusinfluenzae,Haemophilusducreyi,Haemophilusaegyptius,Haemophilusparainfluenzae,HaemophilushaemolyticusandHaemophilusparahaemolyticus);
  • Actinobacillus (e.g. Actinobacillusactinomycetemcomitans,Actinobacillusequuli,Actinobacillushominis,Actinobacilluslignieresii,ActinobacillussuisandActinobacillusureae);
  • Actinomyces (e.g. Actinomycesisraelii);
  • Brucella (e.g. Brucellaabortus,Brucellacanis,BrucellamelintensisandBrucellasuis);
  • Campylobacter (e.g. Campylobacterjejuni,Campylobactercoli,CampylobacterlariandCampylobacterfetus);
  • Listeriamonocytogenes;
  • Vibrio (e.g. V ibriocholeraeandVibrioparahaemolyticus,Vibrioalginolyticus,Vibriocarchariae,Vibriofluvialis,Vibriofurnissii,Vibriohollisae,Vibriometschnikovii,VibriomimicusandVibriovulnificus);
  • Erysipelothrix rhusopathiae;
  • Corynebacteriaceae (e.g. Corynebacteriumdiphtheriae,CorynebacteriumjeikeumandCorynebacteriumurealyticum);
  • Spirochaetaceae, such as Borrelia (e.g. Borreliarecurrentis,Borreliaburgdorferi,Borreliaafzelii,Borreliaandersonii,Borreliabissettii,Borreliagarinii,Borreliajaponica,Borrelialusitaniae,Borreliatanukii,Borreliaturdi,Borreliavalaisiana,Borreliacaucasica,Borreliacrocidurae,Borreliaduttoni,Borreliagraingeri,Borreliahermsii,Borreliahispanica,Borrelialatyschewii,Borreliamazzottii,Borreliaparkeri,Borreliapersica,BorreliaturicataeandBorreliavenezuelensis) and Treponema ( Treponemapallidumssp.pallidum,Treponemapallidumssp.endemicum,Treponemapallidumssp.pertenueandTreponemacarateum);
  • Pasteurella (e.g. Pasteurellaaerogenes,Pasteurellabettyae,Pasteurellacanis,Pasteurelladagmatis,Pasteurellagallinarum,Pasteurellahaemolytica,Pasteurellamultocidamultocida,Pasteurellamultocidagallicida,Pasteurellamultocidaseptica,Pasteurellapneumotropica and Pasteurellastomatis);
  • Bordetella (e.g. Bordetellabronchiseptica,Bordetellahinzii,Bordetellaholmseii,Bordetellaparapertussis,Bordetellapertussis and Bordetellatrematum);
  • Nocardiaceae, such as Nocardia (e.g. NocardiaasteroidesandNocardiabrasiliensis);
  • Rickettsia (e.g. Ricksettsii or Coxiella bumetii);
  • Legionella (e.g. Legionallaanisa,Legionallabirminghamensis,Legionallabozemanii,Legionallacincinnatiensis,Legionalladumoffii,Legionallafeeleii,Legionallagormanii,Legionallahackeliae,Legionallaisraelensis,Legionallajordanis,Legionallalansingensis,Legionallalongbeachae,Legionallamaceachernii,Legionallamicdadei,Legionallaoakridgensis,Legionallapneumophila,Legionallasainthelensi,Legionallatucsonensis and Legionallawadsworthii);
  • Moraxella catarrhalis;
  • Stenotrophomonasmaltophilia;
  • Burkholderiacepacia;
  • Francisellatularensis;
  • Gardnerella (e.g. Gardnerallavaginalis and G ardnerallamobiluncus);
  • Streptobacillusmoniliformis;
  • Flavobacteriaceae, such as Capnocytophaga (e.g. Capnocytophagacanimorsus,Capnocytophagacynodegmi,Capnocytophagagingivalis,Capnocytophagagranulosa,Capnocytophagahaemolytica,Capnocytophagaochracea and Capnocytophagasputigena);
  • Bartonella ( Bartonellabacilliformis,Bartonellaclarridgeiae,Bartonellaelizabethae,Bartonellahenselae,Bartonellaquintana and Bartonellavinsoniiarupensis);
  • Leptospira (e.g. Leptospira biflexa, Leptospiraborgpetersenii,Leptospirainadai,Leptospirainterrogans,Leptospirakirschneri,Leptospiranoguchii,Leptospirasantarosai and Leptospiraweilii);
  • Spirillium (e.g. Spirillumminus);
  • Baceteroides (e.g. Bacteroidescaccae,Bacteroidescapillosus,Bacteroidescoagulans,Bacteroidesdistasonis,Bacteroideseggerthii,Bacteroidesforsythus,Bacteroidesfragilis,Bacteroidesmerdae,Bacteroidesovatus,Bacteroidesputredinis,Bacteroidespyogenes,Bacteroidessplanchinicus,Bacteroidesstercoris,Bacteroidestectus,Bacteroidesthetaiotaomicron,Bacteroidesuniformis,Bacteroidesureolyticus and Bacteroidesvulgatus);
  • Prevotella (e.g. Prevotellabivia,Prevotellabuccae,Prevotellacorporis,Prevotelladentalis ( Mitsuokelladentalis), Prevotelladenticola,Prevotelladisiens,Prevotellaenoeca,Prevotellaheparinolytica,Prevotellaintermedia,Prevotellaloeschii,Prevotellamelaninogenica,Prevotellanigrescens,Prevotellaoralis,Prevotellaoris,Prevotellaoulora,Prevotellatannerae,Prevotellavenoralis and Prevotellazoogleoformans);
  • Porphyromonas (e.g. Porphyromonasasaccharolytica,Porphyromonascangingivalis,Porphyromonascanoris,Porphyromonascansulci,Porphyromonascatoniae,Porphyromonascircumdentaria,Porphyromonascrevioricanis,Porphyromonasendodontalis,Porphyromonasgingivalis,Porphyromonasgingivicanis,Porphyromonaslevii and Porphyromonasmacacae); Fusobacterium (e.g. F.gonadiaformans,F.mortiferum,F.naviforme,F.necrogenes,F.necrophorumnecrophorum,F.necrophorumfundiliforme,F.nucleatumnucleatum,F.nucleatumfusiforme,F.nucleatumpolymorphum,F.nucleatumvincentii,F.periodonticum,F.russii,F.ulcerans and F.varium); Chlamydia (e.g. Chlamydiatrachomatis); Chlamydophila (e.g. Chlamydophilaabortus(Chlamydiapsittaci),Chlamydophilapneumoniae ( Chlamydiapneumoniae) and Chlamydophilapsittaci ( Chlamydiapsittaci)); Leuconostoc (e.g. Leuconostoccitreum,Leuconostoccremoris,Leuconostoc dextranicum, Leuconostoclactis,Leuconostocmesenteroides and Leuconostocpseudomesenteroides); Gemella (e.g. Gemellabergeri,Gemellahaemolysans,Gemellamorbillorum and Gemellasanguinis); and Ureaplasma (e.g. Ureaplasmaparvum and Ureaplasmaurealyticum). Thus, compositions of the invention may be used to kill any of the above-mentioned bacterial organisms. Particular bacteria that may be mentioned in this respect include: Bacillaceae, such as Bacillusanthracis,Bacillussubtilis,Bacillusthuringiensis,Bacillusstearothermophilus and Bacilluscereus; Staphylococci, such as Staph.aureus (either Methicillin-sensitive (i.e. MSSA) or Methicillin-resistant (i.e. MRSA)) and Staph.epidermidis; Acinetobacter (e.g. A.baumanii,A.calcoaceticus,A.haemolyticus,A.johnsonii,A.junii,A.IwoffiandA.radioresistens); Enterobacteriaceae, such as Escherichiacoli, Klebsiella (e.g. Klebs.pneumoniae and Klebs. oxytoca) and Proteus (e.g. Pr.mirabilis,Pr.rettgeri and Pr.vulgaris); or Pseudomonas (e.g. Ps.aeruginosa,Ps.maltophilia ( Stenotrophomonasmaltophilia),Ps.alcaligenes,Ps.chlororaphis,Ps.fluorescens,Ps.luteola.Ps.mendocina,Ps.monteilii,Ps.oryzihabitans,Ps.pertocinogena,Ps.pseudalcaligenes,Ps.putida and Ps.stutzeri). Particular bacterial infections that may be mentioned in relation to the fifth to eighth aspects of the invention include infections with: Bacillaceae, such as Bacillusanthracis,Bacillussubtilis,Bacillusthuringiensis,Bacillusstearothermophilus and Bacilluscereus;
  • Staphylococci, such as Staph.aureus (either Methicillin-sensitive (i.e. MSSA) or Methicillin-resistant (i.e. MRSA)) and Staph.epidermidis;
  • Acinetobacter (e.g. A.baumanii,A.calcoaceticus,A.haemolyticus,A.johnsonii,A.junii,A.Iwoffi and A.radioresistens);
  • Enterobacteriaceae, such as Escherichiacoli, Klebsiella (e.g. Klebs.pneumoniae and Klebs. oxytoca) and Proteus (e.g. Pr.mirabilis,Pr.rettgeri and Pr.vulgaris); or
  • Pseudomonas (e.g. Ps.aeruginosa,Ps.maltophilia ( Stenotrophomonasmaltophilia), Ps.alcaligenes,Ps.chlororaphis,Ps.fluorescens,Ps.luteola.Ps.mendocina,Ps.monteilii,Ps.oryzihabitans,Ps.pertocinogena,Ps.pseudalcaligenes,Ps.putida and Ps.stutzeri).

The compositions of the present invention are particularly advantageous as they are capable of inhibiting the growth, survival and reproduction of Gram negative bacteria, something which few existing antibacterial agents are able to do effectively. Thus, in particular embodiments of all of the methods disclosed herein, the bacteria are Gram negative bacteria.

In this respect, particular conditions that the compositions of the invention can be used to treat include tuberculosis (e.g. pulmonary tuberculosis, non-pulmonary tuberculosis (such as tuberculosis lymph glands, genito-urinary tuberculosis, tuberculosis of bone and joints, tuberculosis meningitis) and miliary tuberculosis), anthrax, abscesses, acne vulgaris, actinomycosis, bacilliary dysentry, bacterial conjunctivitis, bacterial keratitis, botulism, Buruli ulcer, bone and joint infections, bronchitis (acute or chronic), brucellosis, burn wounds, cat scratch fever, cellulitis, chancroid, cholangitis, cholecystitis, cutaneous diphtheria, cystic fibrosis, cystitis, diffuse panbronchiolitis, diphtheria, dental caries, diseases of the upper respiratory tract, empymea, endocarditis, endometritis, enteric fever, enteritis, epididymitis, epiglottitis, erysipclas, erysipeloid, erythrasma, eye infections, furuncles, Gardnerella vaginitis, gastrointestinal infections (gastroenteritis), genital infections, gingivitis, gonorrhoea, granuloma inguinale, Haverhill fever, infected burns, infections following dental operations, infections in the oral region, infections associated with prostheses, intraabdominal abscesses, Legionnaire’s disease, leprosy, leptospirosis, listeriosis, liver abscesses, Lyme disease, lymphogranuloma venerium, mastitis, mastoiditis, meningitis and infections of the nervous system, mycetoma, nocardiosis (e.g. Madura foot), non-specific urethritis, opthalmia (e.g. opthalmia neonatorum), osteomyelitis, otitis (e.g. otitis externa and otitis media), orchitis, pancreatitis, paronychia, pelveoperitonitis, peritonitis, peritonitis with appendicitis, pharyngitis, phlegmons, pinta, plague, pleural effusion, pneumonia, postoperative wound infections, postoperative gas gangrene, prostatitis, pseudo-membranous colitis, psittacosis, pulmonary emphysema, pyelonephritis, pyoderma (e.g. impetigo), Q fever, rat-bite fever, reticulosis, Ritter’s disease, salmonellosis, salpingitis, septic arthritis, septic infections, septicameia, sinusitis, skin infections (e.g. skin granulomas), syphilis, systemic infections, tonsillitis, toxic shock syndrome, trachoma, tularaemia, typhoid, typhus (e.g. epidemic typhus, murine typhus, scrub typhus and spotted fever), urethritis, wound infections, yaws, aspergillosis, candidiasis (e.g. oropharyngeal candidiasis, vaginal candidiasis or balanitis), cryptococcosis, favus, histoplasmosis, intertrigo, mucormycosis, tinea (e.g. tinea corporis, tinea capitis, tinea cruris, tinea pedis and tinea unguium), onychomycosis, pityriasis versicolor, ringworm and sporotrichosis.

Further conditions that may be mentioned in this respect include infections with MSSA, MRSA, Staph.epidermidis,Strept.agalactiae,Strept.pyogenes,Escherichiacoli,Klebs.pneumoniae,Klebs. oxytoca, Pr.mirabilis,Pr.rettgeri,Pr.vulgaris,Haemophilisinfluenzae,Enterococcusfaecalis or Enterococcusfaecium.

The use of certain compositions of the invention in medicine, as well as the use of certain delivery agents in medicine, is, to the knowledge of the inventors, novel. In certain embodiments of the invention, the subject of the treatment or prevention methods is a mammal, particularly a human.

For the avoidance of doubt, references herein to compositions of the invention include references to all embodiments described above in relation to delivery agents of formulae I to IX.

Compositions of the invention may comprise a delivery agent and an antibacterial agent as separate agents or those two agents may be covalently bonded together. Where the delivery agent and antibacterial agent are separate agents, they are distinct chemical entities and may be provided in a form in which they are mixed together or in which they are not intermixed. Additionally, where the delivery agent and antibacterial agent are separate agents, they be separately (e.g. sequentially) delivered to a bacterial cell, or they may be combined prior to delivery. For example, where a composition of the invention is to be used to treat or prevent a bacterial infection in a subject, that subject may be administered each agent either separately or simultaneously.

Because they have a different mode of action to many conventional anti-bacterial agents, compositions of the invention may be particularly useful in the treatment of bacterial infections where the infective agent is resistant to one or more anti-bacterial agents having a different mode of action. In this respect, according to a ninth aspect of the invention there is provided a method of treating a bacterial infection, where the infective agent is resistant to one or more anti-bacterial agents that do not act by inhibiting bond formation in peptidoglycan, which method comprises administration of a therapeutically effective amount of a composition of the invention to a person having that infection.

The use of compositions of the invention in medicine includes their use as pharmaceuticals (both for human and veterinary use). The compositions of the present invention may also be useful in other fields of industry. For example, the compositions may be useful as plant protection products (i.e. in agriculture), in cosmetic products (e.g. in creams, lotions and ointments), and hygiene and sterilisation procedures (e.g. in scientific laboratories).

The compositions of the invention may also be used to facilitate the binding of a pharmaceutically active substance to a penicillin binding protein in a bacterial cell. Therefore in a tenth aspect of the invention, there is provided a method of binding a pharmaceutically active substance to a penicillin binding protein in a bacterial cell, which method involves delivering the pharmaceutically active substance and a delivery agent to the bacterial cell;

  • wherein the delivery agent is covalently bonded to the pharmaceutically active substance or is capable of binding to the pharmaceutically active substance;
  • wherein the pharmaceutically active substance is capable of binding to a transglycosylase domain of the penicillin binding protein; and
  • wherein the delivery agent is capable of binding to a transmembrane domain of the penicillin binding protein.

In this context, the term binding means that the pharmaceutically active substance is held in close proximity to a transglycosylase domain of a penicillin binding protein in a bacterial cell wall, or may interact with the transglycosylase domain by way of one or more hydrogen bonds.

As a result of binding to a penicillin binding protein in a bacterial cell, or through other means, the compositions of the invention may also be used to modulate the activity of a penicillin binding protein in a bacterial cell. Therefore in an eleventh aspect of the invention, there is provided a method of modulating the activity of a penicillin binding protein in a bacterial cell, which method involves delivering a pharmaceutically active substance and a delivery agent to the bacterial cell;

  • wherein the delivery agent is covalently bonded to the pharmaceutically active substance or is capable of binding to the pharmaceutically active substance;
  • wherein the pharmaceutically active substance is capable of binding to a transglycosylase domain of the penicillin binding protein; and
  • wherein the delivery agent is capable of binding to a transmembrane domain of the penicillin binding protein.

In particular embodiments, the modulation of the activity of a penicillin binding protein in a bacterial cell may be the inhibition of the activity of that penicillin binding protein.

In certain embodiments, the methods of binding a pharmaceutically active substance to a penicillin binding protein and the methods of modulating the activity of a penicillin binding protein may be performed in vivo or ex vivo. For example, the methods may be used in the fields of agriculture, cosmetics, hygiene and general scientific research.

The delivery agents described hereinbefore may be capable of enhancing the efficacy of new and existing antibacterial agents due to their ability to bind to the structures in a bacterial cell membrane (thereby allowing them to facilitate in bringing an antibacterial agent into close proximity with structures in the bacterial cell wall, and potentially aiding in delivering the antibacterial agent into the cytoplasmic region of the cell). Thus in a twelfth aspect of the invention, there is provided a method of enhancing the antibacterial effectiveness of an antibacterial agent, which method involves bringing the antibacterial agent into association with a delivery agent as defined herein. By the use of the term “bringing into association” it is meant that the delivery agent and the antibacterial agent may be separate agents or may be covalently bonded together. Where they are separate agents, they be separately (e.g. sequentially) delivered to a bacterial cell, or they may be combined prior to delivery.

The compositions of the invention will normally be administered orally, subcutaneously, intravenously, intraarterially, transdermally, intranasally, by inhalation, or by any other parenteral route, in the form of pharmaceutical preparations comprising the active ingredient either as a free base or a non-toxic organic or inorganic acid addition salt, in a pharmaceutically acceptable dosage form. Depending upon the disorder and patient to be treated, as well as the route of administration, the compositions may be administered at varying doses.

According to a thirteenth aspect of the invention there is thus provided a pharmaceutical formulation including a composition of the invention in admixture with a pharmaceutically acceptable adjuvant, diluent or carrier. Such formulations may be used for the treatment or prevention of bacterial infections. Thus, in one embodiment of this aspect of the invention there is provided a pharmaceutical formulation including, in admixture with a pharmaceutically acceptable adjuvant, diluent or carrier, a composition of the invention and optionally one or more other chemical agents that are known to be effective in treating or preventing bacterial infections.

Compositions of the invention may comprise a delivery agent and an antibacterial agent as separate agents (i.e. where the two agents are not covalently bonded together).

According to an embodiment of this aspect of the invention, there is provided a combination product comprising:

  • (A) a delivery agent, as hereinbefore defined; and
  • (B) an antibacterial agent as hereinbefore defined, wherein each of components (A) and (B) is formulated in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier.

Such combination products provide for the administration of a delivery agent in conjunction with an antibacterial agent, and may thus be presented either as separate formulations, wherein at least one of those formulations comprises a delivery agent, and at least one comprises an antibacterial agent, or may be presented (i.e. formulated) as a combined preparation (i.e. presented as a single formulation including a delivery agent and an antibacterial agent).

Thus, there is further provided:

  • (1) a pharmaceutical formulation including a delivery agent as hereinbefore defined, an antibacterial agent as hereinbefore defined, and a pharmaceutically-acceptable adjuvant, diluent or carrier; and
  • (2) a kit of parts comprising components:
    • (a) a pharmaceutical formulation including a delivery agent, as hereinbefore defined, in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier; and
    • (b) a pharmaceutical formulation including an antibacterial agent, as hereinbefore defined, in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier, which components (a) and (b) are each provided in a form that is suitable for administration in conjunction with the other.

Suitable daily doses of the compositions of the invention in therapeutic treatment of humans are about 1 to 2000 mg/m2.

The most effective mode of administration and dosage regimen for the compositions of the invention depends on several factors, including the particular condition being treated, the extent and localisation of that condition in the patient being treated, as well as the patient’s state of health and their reaction to the compound being administered. Accordingly, the dosages of the compositions of the invention should be adjusted to suit the individual patient. Methods for determining the appropriate dose for an individual patient will be known to those skilled in the art.

Additionally, compositions of the invention may have the advantage that they may be more efficacious than, be less toxic than, have a broader range of activity than, be more potent than, produce fewer side effects than, or have other useful pharmacological properties over compositions known in the prior art. In particular, compositions of the invention may have the advantage that they are less toxic than compositions known in the prior art due to a reduction in the detrimental effects that the delivery agents may have on cell membrane (of the host organisms).

The invention will now be described in more detail by reference to the following, nonlimiting, Figures and Examples.

FIGURES

FIG. 1: The structure of moenomycin A.

FIG. 2: Showing the peptide delivery of TAMRA to E.coli labelled with GFP. Confocal microscope images of GFP labelled E. coli after treatment with TAMRA labelled cell penetrating peptides. A E. coli labelled with GFP, B The same image area looking at TAMRA labelled peptides. C an overlay of images A and B.

BIOLOGICAL TESTS

The effects of compounds of the invention in relation to inhibiting the growth of various microorganisms was determined using methods known to those skilled in the art, for example in vitro methods as described in J. Med. Chem. 47, 2133-2156 (2004) and in vivo methods as described in J. Med. Microbiol. 46, 208-213 (1997), the disclosures of which documents are hereby incorporated by reference.

MIC Testing

  • The bacteria were grown in 5 ml of Mueller Hinton Broth overnight at 37° C.
  • The inhibitor was tested on its own or in conjunction with a cell penetrating peptide (CPP).
  • When tested with a CPP they were incubated together at room temperature for 1 hour before use.
  • Delivery agents (Compound 5, Compound 6, and 7) were used at a 1:2 ratio. CPP Linker was used at a 1:3 ratio or a 1:10 ratio where stated.
  • All dilutions were carried out using Mueller Hinton Broth.
  • The MIC tests were carried out across two 96 well plates unless otherwise stated.
  • 100 µl of autoclaved Mueller Hilton Broth was added to wells 2-12 on plate 1 and 1-12 on plate 2.
  • 200 µl of the inhibitor was added to well 1 at a concentration of 512 µg/ml.
  • 100 µl of the inhibitor was taken from well 1 and pipetted into well 2.
  • The mixture was pipetted up and down three times before 100 µl was taken from well 2 and added to well 3.
  • This process was repeated until well 11 on plate 2.
  • 100 µl was then taken up from well 11 and discarded ensuring that there was no inhibitor present in well 12 of plate 2.
  • The pipette tips were changed between each well so that the concentration was not affected by any inhibitor on the outside of the tips.
  • Each well was inoculated with 100 µl of bacteria that had been diluted to an OD600nm of 0.1.
  • This was repeated three times.
  • The 96 well plates were then incubated for 24 hours at 37° C.
  • The minimum inhibitory concentration (MIC) was determined to be the lowest concentration at which there was no growth visible.
  • To determine the minimum bactericidal concentration (MBC) 10 µl of the sample was pipetted onto a Muller Hinton agar plate containing no inhibitor.
  • Thiswasdonefor each well in which there was no growth and the three wells above the MIC.
  • These were then incubated overnight and the MBC was determined by any growth from a sample corresponding to a well that had no visible growth.

Confocal Microscope

  • XL1blueE.coli was used for this experiment as it was transformed with pJF40 so that it produced GFP.
  • Once the transformation had been completed, 5 ml overnights were inoculated with a single colony and grown up overnight (37° C., 180 rpm).
  • 1mlofbacteria was then spun down and the supernatant was removed.
  • The bacteria pellet as then re-suspended in 0.5 ml HBSS.
  • The labelled peptides were then added to each sample to achieve a final concentration of 10 µM of peptide.
  • The samples were incubated at room temperature for three hours.
  • The samples were then washed 5 times in 0.5 ml PBS by re-suspended the pellet and re-suspending in PBS.
  • After washing 10 µl of each sample was smeared onto a microscope slide ready for viewing on the confocal.
  • No fixation was used as the GFP bleaches quite quickly so the samples will not be able to be used for than once.

Toxicity Test Methods Galleria Mellonella Model

This model was used to test Moenomycin A and peptides in vivo both for toxicity and antimicrobial activity. Conjugates were also tested using the same methods.

Galleriamellonella were selected on the basis of being free of infection and signs of trauma, not beginning to pupate and weighing between 225 mgs and 275 mgs. All calculations were on the basis of an average weight of 250 mgs. The selected Galleria mellonella were swabbed with 70% ethanol prior to injection using a Hamilton 10 µl syringe using disposable needles. Tips were changed between solutions and between each Galleria. All injections were into the front left proleg unless otherwise stated.

Preliminary Testing

Ringers solution was chosen as a control, and all dilutions of bacteria, antimicrobials and peptides were carried out in this. Ringers supplemented with 20% DMSO was also used as a control to account for the DMSO used to dissolve Compound 6. Groups of 10 Galleria were used for these control groups and injected with 10 µl of the appropriate solution as previously described. These were then observed for 96 hours. The Galleria were deemed to be alive if there was movement in response to physical stimuli (touch).

The kill kinetics of P.aeruginosa, S.aureus and A.baumannii were also determined by injecting a set CFU into the Galleria, the aim was to ensure that the Galleria died within 36 hours but not less than 24.

To determine the appropriate antimicrobial concentration, a range was set up from 40 mg/kg to 0.5 mg/kg. Moenomycin was tested both singularly and in conjunction with either Compound 6 or 8; these two compounds were also tested separately. Groups of 2 were used for each test with control groups of 5 used to cover the whole experiment. The Galleria were injected as previously described. For toxicity testing of Moenomycin conjugates, the same procedure was followed with a concentration range up to 80 mg/kg.

To determine the appropriate concentration of antimicrobial in regards to treatment of P. aeruginosa, the Galleria were first injected with 10 µl of the appropriate concentration of bacteria as previously described. They were then treated within half an hour by injection into the first right pro-leg with the compound(s) being tested.

EXAMPLES Example 1 - Purification of Moenomycin A

Flavomycin (100 g) was extracted twice with methanol (400 mL) at room temperature for 16 h. The combined methanol extract was concentrated in vacuo and the residue was purified by silica gel chromatography with a gradient of 2-propanol : 2 M ammonium hydroxide from 9.5 : 0.5 to 7 : 3. Moenomycin A eluted with the last gradient. The solvent was evaporated in vacuo and the obtained brown mass was lyophilized to give a brown solid. Further purification with RP-HPLC (Gemini C18) was performed with a gradient of ACN in water in 30 min to give moenomycin A as a white solid (400 mg).

Calculated mass: 1581.7. Found 789.5 (M/2 - H+); 1582.7 (M + H+); 1604.6 (M + Na+)

Example 2 - Synthesis of Dendrimer Delivery Agents

Compounds 1-4 shown below were prepared using reported procedures from J. Mater. Chem. B 2014, 2, 2153-2167 (see Schemes 1 and 2).

.

Example 3 - General Procedure for the Synthesis of Compounds 3 and 4

Each solution of the crude compounds 1 (500 mg, 0.43 mmol) or 2 (175 mg, 0.08 mmol), which were dissolved in dry DCM (120 mL), was added dropwise over 2 h at 0° C. into a solution of mono-Boc-DAPMA (1.24 g, 5.08 mmol, 12 eq., dissolved in 50 mL dry DCM) employing dry reaction conditions. Immediately, the solution turned yellow due to the displacement of p-nitrophenol. A solution of DMAP (0.20 g, 1.69 mmol, 0.5 eq. per p-nitrophenyl branch) and DIPEA (0.15 mL, 1.69 mmol, 1.0 eq. per p-nitrophenyl branch) in dry DCM (30 mL) was added and the reaction mixtures were stirred at room temperature for 72 h. The solvent was then removed under reduced pressure. Purification was performed both by column chromatography (CHCl3—MeOH—NH4OH 90 — 9 — 1) and size exclusion chromatography (SEC) using a Sephadex™ LH-20 (CHCl3—MeOH 1—1). Drying under high vacuum yielded the products 11 and 12 as yellowish oils.

Compound 3 was obtained as a yellowish viscous oil (1.57 g, 49%).

Compound 4 was obtained as a yellowish viscous oil (0.45 g, 40%).

Example 4 - General Procedure for the Synthesis of Compounds 5 and 6

Compounds 5 and 6 were synthesised from compounds 3 and 4, respectively, according to the procedure below.

TFA (6.0 mL, excess) was slowly added to a solution of compounds 3 (0.10 g, 0.06 mmol) or 4 (0.10 g, 0.03 mmol) in DCM (6.0 mL) and stirred overnight at room temperature. The solvent was removed in vacuo and the residue washed alternately with hexane and diethyl ether. Purification was accomplished via SEC (Sephadex™ LH20, MeOH) to remove any trace amounts of impurities. Freeze drying yielded the compounds 5 and 6 as white foams.

Compound 5 was obtained as a white foam (98 mg, quant.).

Compound 6 was obtained as a white foam (131 mg, quant.).

Example 5 - Guanidilation of PAMAM Dendrimer, 3,3’,3”,3”’-(Ethane-1,2-Divlbis(Azanetrivl)) Tetrakis(N-(2-Guanidinoethyl)Propanamide) (Compound 7)

To a solution of PAMAM dendrimer (200 mg, 0.387 mmol from a solution of 20% in MeOH) in EtOH (1.5 mL), was added 1-H-pyrazole-1-carboxamidine hydrochloride (348.3 mg, 2.322 mmol). The reaction mixture was refluxed at 86° C. and stirred overnight. After stirring, the solvent was removed in vacuo. Extraction in DCM (3x3 mL) was performed and supernatant solution was removed.

Crude mass: 430 mg; crude yield: 78%

Calculated mass: 684.6. Found: 685.6 (M + H+)

Example 6 - Cell Penetration Studies

TAMRA dye and Confocal Microscopy was used to demonstrate the principle of compound meditated delivery to Gram negative bacteria. E. coli cells were label by transforming with pJF40 which vector which harbours a gene for Green fluorescent protein (GFP) when viewed under confocal microscopy (excited at 555 nm emission at 570 nm). This was used to clearly identify bacterial cells.

The same cells were treated with TAMRA labelled cell penetrating peptides (sequence: PLIYLRLLRGQF; excited at 555 nm emission at 570 nm) which fluoresced under a different wavelength which was used to follow the penetration of the dye into the bacteria. Control experiment where peptides were omitted showed no fluorescence.

Confocal microscope images of GFP labelled E.coli after treatment with TAMRA labelled cell penetrating peptides. A E.coli labelled with GFP, B The same image area looking at TAMRA labelled peptides. C an overlay of images A and B.

Example 7 - General Procedure for the Synthesis of Compounds 8 to 19 General Procedure for Peptide Synthesis

Peptide syntheses was performed using standard Fmoc Solid Phase Peptide Synthesis (SPPS) protocols on Rink Amide Chemmatrix Resin, loading = 0.49 mmol/g on a 0.1 mmol scale using a Biotage Initiator + Alstra fully automated microwave peptide synthesizer. All amino acid couplings were performed using 5 eq. Amino Acid with 5 eq. DIC/Oxyma in DMF as a coupling cocktail by irradiating at 70° C. for 5 min. Fmoc deprotection was performed using 20% piperidine in DMF by shaking for 3 min, followed by shaking for 10 min. 4 x 45 s washes were performed after each coupling cycle and 3 x 30s washes were performed after each deprotection cycle.

Propiolic acid was coupled using 3 eq. of acid, 2.9 eq. of HATU and 6 eq. of DIPEA in DMF and shaking for 1 h.

Peptide cleavage was performed using TFA/TIS/H2O = 95:2.5:2.5 (3 mL/100 mg resin). For sequences containing 1 or 2 Arg groups, the cleavage time was 2 h. For 3 or higher Arg containing peptides, cleavage time was 5h. Peptides were precipitated using cold Et2O (-20° C.) by adding approximately 5x volume of the TFA used for cleavage and centrifuging at 7000 rpm at 0° C.

Analysis and Purification of Peptides/Conjugates

All peptides/conjugates were analysed on a Thermo Scientific Dionex Ultimate 3000 RP-HPLC equipped with a Phenomenex Gemini NX C18 110 Å (150 x 4.6 mm) column using the following buffer systems: A: 0.1% HCOOH in milliQ water. B: ACN using a flow rate of 1 ml/min. The use of TFA was avoided as it can damage the phosphate group of Moenomycin A. The column was flushed with 100% A for 5 min prior to an injection and was flushed for 5 min with 95% B and 5% A after the run was finished.

Peptides and conjugates were analysed using the following gradient: 100% A for 2 min. 0-95% B in 15 min. 95% B for 5 min. 100% A for 4 min.

Peptides and conjugates were purified using the same gradient as mentioned above, on a Thermo Scientific Dionex Ultimate 3000 RP-HPLC with a flow rate of 5 mL/min using a Phenomenex Gemini NX C18 110 Å (150 x 10 mm) semi-prep column.

LC-MS data were collected on an Agilent 1100 Series instrument with a Phenomenex Kinetex C18 100 Å column (150 x 4.6 mm, 5 µm at 35° C.) connected to an ESMSD type VL mass detector with a flow rate of 1.5 ml/min was used with the following solvent systems: (A): 0.1% HCOOH in H2O and (B) MeCN. The column was flushed with 100% A for 2 min, then a gradient from 0 to 100% B over 6 min was used, followed by 2 min of flushing with 100% B.

The following compounds were synthesised by this method.

Comp. number Peptide Derivative Chemical Formula Exact Mass Mass found 8 C51 H99N33O9 1317.83 1453.58[M+ CF3CO2- + Na+] 9 C81H130N22O16 1667.00 1667.87 10 C81H130N22O16 1667.00 1667.67 12 C45H87N27O8 1133.72 1133.9 [M + H+] 13 C72H118N16O14 1430.90 1432.94 [M+ H+] 14 C75H118N20O15 1538.91 1538.90 15 C75H118N20O15 1538.91 1674.51 [M+ CF3CO2 + Na+] 16 C75H118N20O15 1538.91 1674.55 [M+ CF3CO2 + Na+] 17 C72H118N20O14 1486.91 1487.4 18 C72H118N20O14 1486.91 1487.43 19 C81H130N22O16 1667.00 1667.87

Example 8 - General Procedure for the Synthesis of Peptide-Moenomycin A Conjugates Via a Ring Conjugation (Compounds 20 to 39) Peptide Synthesis

Peptide synthesis was performed using standard Fmoc Solid Phase Peptide Synthesis (SPPS) protocols on Rink Amide Chemmatrix Resin, loading = 0.49 mmol/g on a 0.1 mmol scale using a Biotage Initiator + Alstra fully automated microwave peptide synthesizer. All amino acid couplings were performed using 5 eq. Amino Acid with 5 eq. DIC/Oxyma in DMF as a coupling cocktail by irradiating at 70° C. for 5 min. Fmoc deprotection was performed using 20% piperidine in DMF by irradiating at 70° C. for 3 min, followed by shaking at r.t. for 10 min. 4 x 45s washes were performed after each coupling cycle and 3 x 30s washes were performed after each deprotection cycle.

Peptide cleavage was performed using TFA/TIS/H2O = 95:2.5:2.5 (3 mL/100 mg resin). For sequences containing 1 or 2 Arg groups, the cleavage time was 2 h. For 3 or higher Arg containing peptides, cleavage time was 5 h. Peptides were precipitated using cold Et2O (-20° C.) by adding approximately 5x volume of the TFA used for cleavage and centrifuging at 7000 rpm at 0° C.

Fmoc-ε-Ahx-OH and (9H-fluoren-9-yl)methyl (2-(2-(2-aminoethoxy)ethoxy)ethyl)-carbamate and 5-Amino-2-Nitrobenzoic acid were coupled using Amino acid, HATU and DIPEA in DMF.

General Procedure for Coupling Peptides to Moenomycin A (15 Mg Scale)

The peptides were coupled using the procedure described in Eur. J. Org. Chem. 2002, 1149-1162.

Analysis and Purification of Peptides/Conjugates

All peptides/conjugates were analysed on a Thermo Scientific Dionex Ultimate 3000 RP-HPLC equipped with a Phenomenex Gemini NX C18 110 Å (150 x 4.6 mm) column using the following buffer systems: A: 0.1% HCOOH in milliQ water. B: ACN using a flow rate of 1 ml/min. The use of TFA was avoided as it can damage the phosphate group of Moenomycin A. The column was flushed with 100% A for 5 min prior to an injection and was flushed for 5 min with 95% B and 5% A after the run was finished.

Peptides and conjugates were analysed using the following gradient: 100% A for 2 min. 0-95% B in 15 min. 95% B for 5 min. 100% A for 4 min.

Peptides and conjugates were purified using the same gradient as mentioned above, on a Thermo Scientific Dionex Ultimate 3000 RP-HPLC with a flow rate of 5 mL/min using a Phenomenex Gemini NX C18 110 Å (150 x 10 mm) semi-prep column.

LC-MS data were collected on an Agilent 1100 Series instrument with a Phenomenex Kinetex C18 100 Å column (150 x 4.6 mm, 5 µm at 35° C.) connected to an ESMSD type VL mass detector with a flow rate of 1.5 ml/min was used with the following solvent systems: (A): 0.1% HCOOH in H2O and (B) MeCN. The column was flushed with 100% A for 2 min, then a gradient from 0 to 100% B over 6 min was used, followed by 2 min of flushing with 100% B.

Complete list of peptides synthesized (Peptide Nos. 1 to 17):

Peptide No. Peptide-linker structure Chemical Formula Exact Mass Mass found 1. C37H66N20O8 918.54 919.54 2. C55H103N35O11 1429.86 1430 3. C61 H114N36O12 1542.94 1543 4. C67H125N37O13 1656.03 1655.8 5. C67H126N40O13 1699.04 1698.9 6. C73H138N44O14 1855.14 1855.5 7. C73H138N40O14 1799.13 1799.4 8. C61H115N33O12 1501.94 1502 9. C67H126N34O13 1615.02 1614.6 10. C73H137N35O14 1728.11 1728.6 11. C43H79N17O9 977.62 977.5 12. C49H90N18O10 1090.71 1090.6 13. C55H101N19O11 1203.79 1203.9 14. C66H123N25O13 1487.99 1487.8 15. C82H120N34O13 1788.98 1789.5 16. C79H122N22O17 1650.94 1651 17. C13H19N7O4 337.15 338.1 [M+ H+]

Complete list of conjugates synthesized (Compounds 20 to 34):

Comp. No. Conjugate description Chemical Formula Exact Mass Mass found 20. Conjugate with peptide 1 C106H171 N26O42P 2511.18 2512.2 21. Conjugate with peptide 2 C124H208N41O45P 3022.50 3022.8 22. Conjugate with peptide 3 C130H219N42O46P 3135.58 3137.1 23. Conjugate with peptide 4 C136H230N43O47P 3248.67 3249.6 24. Conjugate with peptide 5 C136H231 N46O47P 3291.68 3292.8 25. Conjugate with peptide 6 C142H243N50O48P 3447.78 3448.5 26. Conjugate with peptide 7 C136H231 N40O47P 3207.67 3208.2 27. Conjugate with peptide 8 C130H220N39O46P 3094.58 3094.8 28. Conjugate with peptide 9 C142H243N46O48P 3391.77 3392.1 29. Conjugate with peptide 10 C142H242N41O48P 3320.75 3322 30. Conjugate with peptide 11 C112H184N23O43P 2570.27 2570.8 31. Conjugate with peptide 12 C118H195N24O44P 2683.35 2683.8 32. Conjugate with peptide 13 C124H206N25O45P 2796.43 2797 33. Conjugate with peptide 16 C148H227N28O51P 3243.58 3244.4 34. Conjugate with peptide 17 C82H124N13O38P 1929.79 1930

General structure of a peptide-Moenomycin A conjugate via A ring conjugation

The molecular fragment shown below is present in all of peptides 1 to 17:

The general structure shown above for the peptide-Moenomycin A conjugates shows the nitrobenzene ring (as is present in all of peptides 1 to 17) attached to the Moenomycin skeleton.

Shown below are structures of peptide conjugates that have been synthesised.

Example 9 - Antibacterial Efficacy Test Results

Organism: A. Baumannii 19606 Test material MIC MBC Ampicillin A 128 µg/ml Growth in all wells B 128 µg/ml Growth in all wells C 128 µg/ml Growth in all wells Moenomycin A A 4 µg/ml 8 µg/ml B 4 µg/ml 8 µg/ml C 8 µg/ml 8 µg/ml Moenomycin A + Compound 5 A 4 µg/ml 8 µg/ml B 4 µg/ml 8 µg/ml C 4 µg/ml 8 µg/ml Compound 5 A Growth in all wells Growth in all wells B Growth in all wells Growth in all wells C Growth in all wells Growth in all wells Moenomycin A + Compound 6 A 2 µg/ml 2 µg/ml B 2 µg/ml 2 µg/ml C 2 µg/ml 2 µg/ml Compound 6 A 256 µg/ml 256 µg/ml B 256 µg/ml 256 µg/ml C 256 µg/ml 256 µg/ml Moenomycin A + Compound 7 A 4 µg/ml 8 µg/ml B 4 µg/ml 8 µg/ml C 4 µg/ml 8 µg/ml Compound 7 A Growth in all wells Growth in all wells B Growth in all wells Growth in all wells C Growth in all wells Growth in all wells Moenomycin A + Compound 8 2 16 Compound 8 Growth in all wells Growth in all wells Moenomycin A + Compound 9 16 128 Compound 9 Growth in all wells Growth in all wells Moenomycin A + Compound 10 16 128 Compound 10 Growth in all wells Growth in all wells Moenomycin A + Compound 11 16 32 Compound 11 Growth in all wells Growth in all wells Moenomycin A + Compound 12 4 8 Compound 12 Growth in all wells Growth in all wells Moenomycin A + Compound 13 16 16 Compound 13 Growth in all wells Growth in all wells Moenomycin A + Compound 15 8 32 Compound 15 Growth in all wells Growth in all wells Moenomycin A + Compound 16 2 64 Compound 16 Growth in all wells Growth in all wells Moenomycin A + Compound 17 8 64 Compound 17 Growth in all wells Growth in all wells Organism: K. Pneumonia 700603 Test material MIC MBC Ampicillin A Growth in all wells Growth in all wells B Growth in all wells Growth in all wells C Growth in all wells Growth in all wells Moenomycin A A 64 µg/ml 128 µg/ml B 64 µg/ml 128 µg/ml C 64 µg/ml 128 µg/ml Moenomycin A + Compound 5 A 32 µg/ml 16 µg/ml B 16 µg/ml 8 µg/ml C 16 µg/ml 16 µg/ml Compound 5 A 128 µg/ml 128 µg/ml B 64 µg/ml 64 µg/ml C 64 µg/ml 64 µg/ml Moenomycin A + Compound 6 A 4 µg/ml 4 µg/ml B 4 µg/ml 4 µg/ml C 4 µg/ml 4 µg/ml Compound 6 A Growth in all wells Growth in all wells B Growth in all wells Growth in all wells C Growth in all wells Growth in all wells Moenomycin A + Compound 7 A 64 µg/ml 64 µg/ml B 32 µg/ml 32 µg/ml C 32 µg/ml 32 µg/ml Compound 7 A Growth in all wells Growth in all wells B Growth in all wells Growth in all wells C Growth in all wells Growth in all wells Moenomycin A + Compound 8 4 4 Compound 8 Growth in all wells Growth in all wells Moenomycin A + Compound 9 32 64 Compound 9 Growth in all wells Growth in all wells Moenomycin A + Compound 10 32 64 Compound 10 Growth in all wells Growth in all wells Moenomycin A + Compound 11 32 64 Compound 11 Growth in all wells Growth in all wells Moenomycin A + Compound 12 8 32 Compound 12 Growth in all wells Growth in all wells Moenomycin A + Compound 13 32 128 Compound 13 Growth in all wells Growth in all wells Moenomycin A + Compound 15 32 32 Compound 15 Growth in all wells Growth in all wells Moenomycin A + Compound 16 16 64 Compound 16 Growth in all wells Growth in all wells Moenomycin A + Compound 17 8 64 Compound 17 Growth in all wells Growth in all wells

Organism: P. Aeruginosa 27853 Test material MIC MBC Ampicillin A Growth in all wells Growth in all wells B Growth in all wells Growth in all wells C Growth in all wells Growth in all wells Moenomycin A A 256 µg/ml Growth in all wells B 256 µg/ml Growth in all wells C 256 µg/ml Growth in all wells Moenomycin A + Compound 5 A 32 µg/ml 256 µg/ml B 32 µg/ml 256 µg/ml C 32 µg/ml 128 µg/ml Compound 5 A 256 µg/ml Growth in all wells B 256 µg/ml 256 µg/ml C 256 µg/ml 256 µg/ml Moenomycin A + Compound 6 A 4 µg/ml 16 µg/ml B 8 µg/ml 32 µg/ml C 4 µg/ml 16 µg/ml Compound 6 A 256 µg/ml 256 µg/ml B 256 µg/ml 256 µg/ml C 256 µg/ml 256 µg/ml Moenomycin A + Compound 7 A 128 µg/ml Growth in all wells B 128 µg/ml Growth in all wells C 128 µg/ml Growth in all wells Compound 7 A Growth in all wells Growth in all wells B Growth in all wells Growth in all wells C Growth in all wells Growth in all wells Moenomycin A + Compound 8 16 64 Compound 8 Growth in all wells Growth in all wells Moenomycin A + Compound 9 128 Growth in all wells Compound 9 Growth in all wells Growth in all wells Moenomycin A + Compound 10 128 Growth in all wells Compound 10 Growth in all wells Growth in all wells Moenomycin A + Compound 11 64 128 Compound 11 Growth in all wells Growth in all wells Moenomycin A + Compound 12 32 64 Compound 12 Growth in all wells Growth in all wells Moenomycin A + Compound 13 64 256 Compound 13 Growth in all wells Growth in all wells Moenomycin A + Compound 15 128 Growth in all wells Compound 15 Growth in all wells Growth in all wells Moenomycin A + Compound 16 256 Growth in all wells Compound 16 Growth in all wells Growth in all wells Moenomycin A + Compound 17 Growth in all wells Growth in all wells Compound 17 Growth in all wells Growth in all wells

Organism: E. Coli 25922 Test material MIC MBC Ampicillin 8 µg/ml 8 µg/ml Moenomycin A 32 µg/ml 128 µg/ml Moenomycin A + Compound 5 16 µg/ml 64 µg/ml Compound 5 64 µg/ml 128 µg/ml* Moenomycin A + Compound 6 4 µg/ml 8 µg/ml Compound 6 256 µg/ml Growth in all wells Moenomycin A + Compound 7 Not tested Not tested Compound 7 Not tested Not tested

Organism: B. Subtilis 2410 Test material MIC MBC Ampicillin 128 µg/ml 256 µg/ml Moenomycin A 256 µg/ml Growth in all wells Moenomycin A + Compound 5 8 µg/ml 64 µg/ml Compound 5 32 µg/ml 32 µg/ml Moenomycin A + Compound 6 4 µg/ml 64 µg/ml Compound 6 32 µg/ml 32 µg/ml Moenomycin A + Compound 7 Not tested Not tested Compound 7 Not tested Not tested

Example 9 - Antibacterial Efficacy Test Results for Conjugated Delivery Agents

Compound Number B. Subtilis 168 K. Pnumoniae ATCC 700603 A. Baumannii ATCC 19606 P. Aeruginosa ATCC 27853 MIC MBC MIC MBC MIC MBC MIC MBC Compound 20 32 GAW 64 32 64 GAW GAW Compound 21 128 GAW 8 256 128 GAW Compound 22 16 32 64 32 64 256 GAW Compound 23 2 64 8 256 64 128 16 256 Compound 24 2 64 GA W GAW 32 64 256 GAW Compound 25 1 64 GA W 256 64 128 256 256 Compound 26 4 GAW 16 64 8 32 128 256 Compound 27 GA W GAW 4 64 128 GAW Compound 28 8 64 GA W GAW 64 128 256 GAW Compound 29 256 GAW 16 64 128 GAW Compound 30 64 64 4 32 64 128 Compound 31 0.5 64 64 128 1 64 64 Compound 32 64 GAW 2 8 128 GAW Compound 33 GA W GAW 64 GAW GAW GAW Compound 34 64 64 4 256 256 GAW Compound number S. ATCC Aureus 25922 MIC MBC Compound 34 0.25 -

Example 10 - Antibacterial Efficacy Test Results for Peptide Delivery Agents

Peptide / anti-bacterial agent B. subtilis 168 K. pnumoniae ATCC 700603 A. baumannii ATCC 19606 P. aeruginosa ATCC 27853 MIC MBC MIC MBC MIC MBC MIC MBC Peptide 4 128 GAW GAW GAW GAW GAW GAW GAW Peptide 4 + Moenomycin A 1 GAW 16 32 2 4 64 256 Peptide 5 128 GAW GAW GAW GAW GAW GAW GAW Peptide 5 + Moenomycin A 0.25 8 32 GAW 1 4 32 256 Peptide 6 128 256 GAW GAW GAW GAW GAW GAW Peptide 6 + Moenomycin A 0.25 8 16 GAW 1 8 32 GAW Peptide 9 GAW GAW GAW GAW GAW GAW GAW GAW Peptide 9 + Moenomycin A 0.5 GAW 16 16 2 16 32 256 Peptide 15 GAW GAW GAW GAW GAW GAW GAW GAW Peptide 15 + Moenomycin A 0.25 16 8 16 1 4 32 64 “GAW” = growth in all wells

Peptide / anti-bacterial agent S. Aureus ATCC 25922 MIC MBC Peptide 17 256 Peptide 17 + Moenomycin A 0.25

Example 11 - Toxicity Data for Unbound Delivery Agents Wax Moth Galleria Mellonella

In vivo toxicity tests were conducted for Compounds 1 to 19 using wax moth Galleria mellonella. No toxicity was observed below 40 mg/kg (24-96 hr, 100 % survival).

Mammalian Cytotoxicity

Moenomycin A - no toxicity was observed up to 10 mg/mL.

Compound 8 - no toxicity was observed up to 110 µg/mL.

Compound 6 did not affect the cell viability in the tumor cell line 786-O in initial in vitro testing. Compound 6 was toxic in HeLa cells at 20 µg/ml.

Example 12 — Toxicity Data for Covalent Conjugates of Moenomycin A Wax Moth Galleria Mellonella

Compounds 26, 29, 32 were tested - no toxicity was observed below 80 mg/kg (24 hr, 100 % survival).

Mammalian Cytotoxicity

In vitro testing in HeLa cells showed no toxicity up to the concentrations mentioned in the table below.

Compound number 100% survival in µM 100% survival in µg/mL 29 10 33.23 26 2 6.42 32 10 27.98

ABBREVIATIONS

  • ACN = Acetonitrile
  • Boc = tert-butyloxycarbonyl
  • DCM = dichloromethane
  • DIBAL = diisobutylaluminium hydride
  • DIPEA = N,N-diisopropylethylamine
  • DMAP = 4-dimethylaminopyridine
  • DMF = N,N-dimethylformamide
  • eq. = equivalents
  • Et = ethyl
  • h = hour(s)
  • HCl = hydrochloric acid
  • HPLC = high performance liquid chromatography
  • IR = infra red (in relation to spectroscopy)
  • MBC = minimum bactericidal concentration
  • mcpba = meta-chloroperoxybenzoic acid
  • Me = methyl
  • MIC = minimum inhibitory concentration
  • min. = minute(s)
  • MS = mass spectrometry
  • PAMAM = polyamidoamine
  • RP = reverse phase
  • rt/RT = room temperature
  • THF = tetrahydrofuran
  • TFA = trifluoroacetic acid

Prefixes n-, s-, i-, t- and tert- have their usual meanings: normal, secondary, iso, and tertiary.

Claims

1. A combination of an antibacterial agent and a delivery agent, wherein; the delivery agent is a polypeptide or a polypeptide derivative, or a pharmaceutically-acceptable salt thereof, that bears a positive charge of at least 2 units and is capable of binding to one or more structures on a bacterial cell membrane.

2. The combination according to claim 1, wherein the antibacterial agent is a molecule or fragment that modulates the activity of a penicillin binding protein.

3. The combination according to claim 1, wherein the antibacterial agent is an inhibitor of a glycosyltransferase enzyme.

4. The combination according to claim 1, wherein the antibacterial agent is a molecule or fragment that inhibits the synthesis or repair of bacterial cell walls.

5. The combination according to claim 1, wherein the delivery agent is capable of binding to one or more structures on a bacterial cell membrane via the formation of one or more covalent bonds with said structures, or via the formation of one or more hydrogen bonds with said structures or through electrostatic interactions between oppositely charged regions on the delivery agent and the bacterial cell membrane.

6. The combination according to claim 1, wherein the delivery agent comprises one or more functional groups independently selected from the list consisting of, primary amines, amidines, amides, and salts thereof.

7. The combination according to claim 6, wherein the each of the one or more functional groups is independently selected from the list consisting of primary amines, amidines, guanidines, amides, ureas, thioamides, thioureas and acid addition salts thereof.

8. The combination according to claim 6, wherein the delivery agent comprises at least four of said functional groups.

9. The combination according to claim 1, wherein the delivery agent comprises at least two amino acid residues selected from the group consisting of arginine and lysine.

10. (canceled)

11. The combination according to claim 1, wherein the delivery agent is a compound selected from the group consisting of:.

12. The combination according claim 1, wherein the delivery agent is covalently bonded to the antibacterial agent.

13. The combination according to claim 1, wherein the antibacterial agent is a glycopeptide or a phosphoglycolipid molecule or fragment.

14. The combination according to claim 1, wherein the antibacterial agent is selected from the group consisting of moenomycin, moenomycin derivatives, vancomycin, vancomycin derivatives, β-lactam antibiotics and derivatives thereof.

15. The combination according to claim 14, wherein the antibacterial agent is a moenomycin A derivative which is covalently bound to the delivery agent:

(i) such that the delivery agent and any associated linker replaces part or all of the 2-amido-cyclopentane-1,3-dione portion of the moenomycin;
(ii) via the moenocinol portion of the moenomycin;
(iii) such that the delivery agent and any associated linker replaces part or all of the moenocinol portion of the moenomycin; or
(iv) via the 2-amino-cyclopentane-1,3-dione portion of the moenomycin.

16. The combination according to claim 14, wherein the delivery agent portion of the combination contains a structure selected from the list consisting of:.

17. A pharmaceutical formulation comprising the combination of claim 1 in admixture with a pharmaceutically acceptable adjuvant, diluent or carrier.

18-28. (canceled)

29. A delivery agent as defined in claim 16, or a pharmaceutically acceptable salt thereof.

30-40. (canceled)

Patent History
Publication number: 20230173082
Type: Application
Filed: Jun 30, 2022
Publication Date: Jun 8, 2023
Inventors: Ishwar SINGH (Lincoln), Edward TAYLOR (Lincoln)
Application Number: 17/855,222
Classifications
International Classification: A61K 47/64 (20060101); C07C 277/08 (20060101); A61K 38/14 (20060101); C07C 271/20 (20060101); C07C 279/12 (20060101); A61K 38/16 (20060101); C07H 13/00 (20060101); A61K 47/59 (20060101); A61P 31/04 (20060101); A61K 31/7034 (20060101); A61K 31/7056 (20060101);