(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2013/053372 Al 18 April 2013 (18.04.2013) P O P C T

(51) International Patent Classification: (81) Designated States (unless otherwise indicated, for every C07F 5/02 (2006.01) A61P 31/04 (2006.01) kind of national protection available): AE, AG, AL, AM, A61K 31/69 (2006.01) AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, (21) International Application Number: DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, PCT/EP20 11/005 142 HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, (22) International Filing Date: KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, 13 October 201 1 (13.10.201 1) MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SC, SD, (25) Filing Language: English SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, (26) Publication Language: English TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (71) Applicant (for all designated States except US): (84) Designated States (unless otherwise indicated, for every THERABOR PHARMACEUTICALS [IT/IT]; via kind of regional protection available): ARIPO (BW, GH, Campi 183, 1-41 125 Modena (IT). GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, SZ, TZ, UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, MD, RU, (72) Inventors; and TJ, TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, (75) Inventors/ Applicants (for US only): PRATI, Fabio DK, EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, [IT/IT]; via Antonelli 9/00, 1-42123 Reggio Emilia (IT). LV, MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, CASELLI, Emilia [IT/IT]; via della Pace 53, 1-41049 Sas- SM, TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, GQ, suolo (Mo) (IT). GW, ML, MR, NE, SN, TD, TG). (74) Agent: GRAF VON STOSCH, Andreas; Graf von Stosch Published: Patentanwaltsgesellschaft mbH, Prinzregentenstrasse 22, — with international search report (Art. 21(3)) 80538 Munchen (DE).

l

© o- (54) Title: BORONIC ACID INHIBITORS OF BETA-LACTAMASES (57) Abstract: The invention relates to novel boronic acid compounds, a method for the preparation of such compounds, intermedi - ate compounds for the preparation of such compounds, intermediate compounds for the use in a method for preparation of such com- pounds, a pharmaceutical composition, the use of one or more compounds discussed above or of a pharmaceutical composition in S the manufacture of a medicament for the treatment of a bacterial infection, and a screening method. Boronic acid inhibitors of beta-lactamases

The invention relates to novel boronic acid compounds, a method for the preparation of such compounds, intermediate compounds for the preparation of such compounds, intermediate compounds for the use in a method for preparation of such compounds, a pharmaceutical composition, the use of one or more compounds discussed above or of a pharmaceutical composition in the manufacture of a medicament for the treatment of a bacterial infection, and a screening method.

FIELD OF INVENTION

The present disclosure relates to boronic acids containing at least one heterocyle. These compounds act as beta-lactamase enzyme inhibitors.

BACKGROUND OF THE INVENTION

Bacterial resistance to represents today one of the most alarming aspects of the pharmacotherapy: in fact, the number of therapeutic options for serious, life-threatening bacterial infections are becoming increasingly limited because of multidrug resistance. Multidrug resistant pathogens are now characterized by their heterogeneity, increasing virulence, rsistance even to reserve agents and spread within and between hospitals and the community. Examples are the meticillin-resistance Staphylococcus aureus (MRSA) and enterococci, extended spectrum β-lactamases (ESBL) and carbapenemase- producing coliform, and toxin-hyperproducing Clostridium difficile. Resistance to β-lactams in Staphylococcus aureus is mainly caused by acquisition of a low affinity -binding protein, PBP 2a, but is accompained by other resistance factors that provide resistance to most of clinically used antibiotics.. A number of relatevly new drugs such as linezolid, and tigecycline are now available to treat infections caused by β-lactam-resistant Gram positive cocci. Thus attention has shfted to the drug resistant gram-negative bacteria that have been acquiring mobile genetic elements associated with multiple resistance determinants for most classes. In Gram negative bacteria, the most common β-lactam resistance

mechanism involves β-lactamase-mediated hydrolysis resulting in subsequent inactivation of the antibiotic.(Bush, K., Curr. Op. Microbiol., 2010, 13, 558-564) A recent compilation of β-lactamases cataloged over 950 unique, naturally occurring enzymes (see Lahey Clinic β-lactamase website). The simplest classification of this class of enzymes is based on the protein sequence whereby the β-lactamases are classified into four molecular classes A , B, C, and D, based on the conserved and distinguishing amino acids motif. Clas A , C and D include enzymes that hydrolyze their substrate by forming an acyl enzyme through an active site serine, whereas class B β-lactamases are metalloenzymes that utilize at least one-active site Zinc ion to facilitate hydrolysis. The concept of achieving antibacterial activity, without accelerating resistance development and without compromising safety, by synergism between a β-lactam antibiotic and a β-lactamase inhibitor (inactivator) is proven by decades of clinical success with the / (Augmentin®), /clavulanic acid (Timentin®), / (Unasyn®), / sulbactam (Sulperazone®) and / (Zosyn®). combinations. With the forward evolution of β-lactamase capability resulting in the diminished effectiveness of clavulate and the penam sulfone against these enzymes, there is a rapidly growing unmet medical need and hence a renewed opportunity for β-lactamase inhibitors discovery. Specifically, recognizing the emergence of β-lactamases capable of hydrolysis, future identification of an effective carbapenemase inhibitor has the potential to establish clinical longevity for existing , as clavulate and the penicillin sulfone have accomplished for the penicillin. Boronic acids have been known since the late '80 to inhibit β-lactamases

(Crompton, I. E., Cuthbert, B. K., Lowe, G., Waley, S. G., Biochem. J., 1988, 251, 453-9.). They are compounds that replace the β-lactam ring with boronic acid. The boron atom forms of a reversible, dative covalent bond with the active site serine of class A and C and D β-lactamases, assuming a geometry that resembles the tetrahedral transition state of the β-lactamase hydrolytic reaction. By modifying the boronic acid substituents to resemble in structure, distance, and stereochemical arrangement the R 1 side chains of natural substrates, affinities in the nM range against class C enzymes of Escherichia coli were achieved (U.S. Patent No. 7,271 ,186). Ness et al. (Biochemistry (2000) 39:5312-21) discloses β-lactamase inhibitors that target TEM-1 (a non-ESBL TEM variant from class A ; one of approximately 140 known TEM-type betalactamase variants). More recently patent WO201 0/1 30708 disclosed a-aminoacyl-p-boron (3-carboxyphenyl) as broad spectrum β-lactamases inhibitors.

Taking respect to the rapidly evolving β-lactamase capability of bacteria, it is the object of the invention to provide novel compounds for inhibition of β-lactamase activity.

As a solution the invention provides compounds featuring a boronic acid structure bearing a heterocycle.

These compounds act as β-lactamase inhibitors and are useful in several pathological conditions either used alone or in combination with a therapeutically effective agent.

A compound, particularly a boronic acid containing at least one heterocyle, having the general formula (I) is a bond or a C1-C8 saturated or unsaturated linear or branched aliphatic chain, optionally substituted with one or more groups chosen from hydroxyl, alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, amino, aminocarbonyl, carbonyl, aminosulfonyl, alkylaryl, aryl, aryloxy, carboxyl, cyano, guanidine, halogen, heteroaryl, heterocyclyl, sulfide, sulfonyl, sulfoxido, sulfonic acid, sulfate and thiol;

is chosen from hydrogen, linear or branched C1-C12 alkyl, linear or branched C1-C12 alkenyl, or C3-C8 cycloalkyi; said linear or branched C1-C12 alkyl, linear or branched C1-C12 alkenyl, or C3- C8 cycloalkyi being optionally substituted with one or more groups independently chosen from the group consisting of hydroxyl,

halogen, carboxyl, cyano, C(0)R 4, C(0)NR 4R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido;

is 0, 1, 2, 3, 4, 5, or 6; and X 2 are independently hydroxyl, halogen, NR1R2, C1-C6 alkoxy, or acyloxy; or and X 2 together form a cyclic boron ester, a cyclic boron amide, or a cyclic boron amide-ester, said cyclic boron ester, boron amide or boron amide-ester having a chain or a ring containing from 2 to 12 carbon atoms and, optionally, 1-3 heteroatoms which can be O, N , or S ;

is chosen from hydrogen, optionally substituted linear or branched C1-C12 alkyl, optionally substituted linear or branched C1-C12 alkenyl, OR3, SR3, or from the group consisting of aryl group NRiR2 > substituted with from 0 to 3 substituents, heteroaryl group substituted with from 0 to 3 substituents and heterocyclic group substituted with from 0 to 3 substituents, said substituents being independently selected from the group consisting of hydroxyl,

halogen, carboxyl, cyano, C(0)R4, C(0)NR R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido;

, R2, R3, R , R 5 are independently selected from the group consisting

of: (a) hydrogen; (b) C1-C10 alkyl any carbon of which can be substituted with 0 , 1, 2 or 3 substituents; (c) C3-C10 cycloalkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents; (d) heteroaryl group substituted with 0, 1, 2 or 3 substituents; (e) heterocyclic group substituted with 0, 1, 2 or 3 substituents; (f) C1-C12 alkyl, C1-C12 cycloalkyl, C1-C12 alkenyl; (g) aryl group substituted with 0, 1, 2, 3, 4 or 5 substituents;

(h) C(O)R6; -C(0)NR6R7; -C(0)OR6, C(=NR6) ; NR6R7, -

C(=NR6)R ; (i) C1-C6 alkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents; (j) C3-C7 cycloalkyi any carbon of which can be substituted with 0 , 1, 2 or 3 substituents; said substituents of Ri, R2, R3, R and R5 being independently selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(0)R6, C(0)NR 6 R7, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C10 carbons of (b), any of the carbons of the cycloalkyi group of (c) other than the one attached to the rest of the molecule, any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule, any of the C1-C6 carbons of (i) or any of the carbons of the cycloalkyi group of ) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, sulfoxido; and R 6 and R7 being independently selected from hydrogen, or from the group consisting of (a) C1-C6 alkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents, (b) C3-C7 cycloalkyi any carbon of which can be substituted with 0, , 2 or 3 substituents, (c) aryl group substituted with 0, 1, 2 or 3 substituents (d) heteroaryl group substituted with 0 , 1, 2 or 3 substituents and (e) heterocyclic group substituted with 0, 1, 2 or 3 substituents, the substituents of R 6 and R7 being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C6 carbons of (a), any of the carbons of the cycloalkyl group of (b) other than the one attached to the rest of the molecule, or any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, and sulfoxido or an oxyimino group as indicated above; and

is an heterocyclic or heteroaromatic ring system optionally substituted with 1 or more substituents selected from the group consisting of hydroxyl, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, amino, aminocarbonyl, carbonyl, aminosulfonyl, alkylaryl, aryl, aryloxy, carboxyl, cyano, guanidine, halogen, heteroaryl, heterocyclyl, sulfide, sulfonyl, sulfoxido, sulfonic acid, sulfate and thiol.

In another embodiment of the invention the heterocyclic or heteroaromatic ring system Z is a stable 3- to 8-membered monocyclic or stable 7- to 14-membered bicyclic heterocyclic ring which is either saturated or unsaturated, and which consists of carbon atoms and from 1 to 4 heteroatoms selected from the group consisting of N, O, and S, preferably N.

In another embodiment of the invention the heterocyclic or heteroaromatic ring system Z is a bicyclic group in which a heterocyclic ring is fused to a benzene ring.

In another embodiment of the invention the heterocyclic or heteroaromatic ring system Z is selected from the group comprising azetidinyl, oxetanyl, oxazolyl, pirazolyl, thiazolyl, triazolyl, tetrazolyl, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, 1,3-dioxolanyl, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, 2-oxopiperazinyl, 2-oxopiperdinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, 2-pyridinonyl, pyrazinyl, pyrazolidinyl.pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, thienyl and triazolyl.

In another embodiment of the invention the heterocyclic or heteroaromatic ring system Z is a heteroaryl group, preferably a group having 5 to 14 ring atoms, more preferably a group having 5, 6, 9, or 10 ring atoms; said heteroaryl group preferably having 6, 10, or 14 π electrons shared in a cyclic array; and having 1, 2 or 3 heteroatoms independently selected from the group consisting of N, O, and S, even more preferably the heterocyclic or heteroaromatic ring system of Z is a heteroaryl group selected from the group comprising thienyl, benzothienyl, furyl, benzofuryl, dibenzofuryl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, indolyl, quinolyl, isoquinolyl, quinoxalinyl, tetrazolyl, oxazolyl, thiazolyl, isoxazolyl and triazolyl.

In another embodiment of the invention the heterocyclic or heteroaromatic ring system Z is fused to an aryl or heteroaryl group, preferably the heterocyclic or heteroaromatic ring system Z is selected from the group comprising tetrahydroquinolinyl, dihydrobenzofuranyl, benzoxazinyl, 1,2,3,4-tetrahydro- quinoxalinyl, benzoimidazolyl, benzothiazolyl, benzotriazolyl.

In another embodiment of the invention the heterocyclic or heteroaromatic ring system Z is substituted with 0, 1, 2 or 3 substituents selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(0)R6, C(O)NR 6 R7, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido, wherein

R 6 and R7 are independently selected from hydrogen, or from the group consisting of (a) C1-C6 alkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents, (b) C3-C7 cycloalkyl any carbon of which can be substituted with 0 , , 2 or 3 substituents, (c) aryl group substituted with 0, 1, 2 or 3 substituents (d) heteroaryl group substituted with 0, 1, 2 or 3 substituents and (e) heterocyclic group substituted with 0, 1, 2 or 3 substituents,

the substituents of R 6 and R being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C6 carbons of (a), any of the carbons of the cycloalkyl group of (b) other than the one attached to the rest of the molecule, or any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, and sulfoxido. In another embodiment of the invention L is a bond or a C1-C8 saturated or

unsaturated linear or branched aliphatic chain, preferably a bond, (CH2) or

(CH2 )2, more preferably (C ).

In another embodiment of the invention Z is an triazol optionally substituted with 1 or more substituents selected from the group consisting of hydroxyl, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, amino, aminocarbonyl, carbonyl, aminosulfonyl, alkylaryl, aryl, aryloxy, carboxyl, cyano, guanidine, halogen, heteroaryl, heterocyclyl, sulfide, sulfonyl, sulfoxido, sulfonic acid, sulfate and thiol,

preferably from

In another embodiment of the invention Y is a heteroaryl group substituted with from 0 to 3 substituents, said substituents being independently selected from the

group consisting of hydroxyl, halogen, carboxyl, cyano, C(O)R , C(0)NR R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido; preferably Y is selected from the group comprising thienyl, benzothienyl, furyl, benzofuryl, dibenzofuryl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, indolyl, quinolyl, isoquinolyl, quinoxalinyl, tetrazolyl, oxazolyl, thiazolyl, isoxazolyl and triazolyl.

Preferably the coumpound according to the invention is having the general formula (II), (V), (VI) or (VII) wherein Y, m, R , X and X 2 are defined as above and

Re, R9, R10, R 11 and R12 are independently selected from the group consisting of (a) hydrogen; (b) C1-C6 alkyl any carbon of which can be substituted with from 0 to 3 substituents, (c) C3-C7 cycloalkyi any carbon of which can be substituted with from 0 to 3 substituents, (d) aryl group substituted with from 0 to 3 substituents (e) heteroaryl group substituted with from 0 to 3 substituents and (f) heterocyclic group substituted with from 0 to 3 substituents, the substituents of R e and R g being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C6 carbons of (a), any of the carbons of the cycloalkyi group of (b) other than the one attached to the rest of the molecule, or any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, and sulfoxido; most preferably R 8, Rg, Rio, R and R12 are selected independently from either hydrogen or a compound according to one of the following formulas a., b., c , d., or e .

ther preferred that the compound is

, or a salt thereof.

In another embodiment the compound according to the invention is an inhibitor for β-lactamase, preferably for a class A , C or D β-lactamase, even more preferably for a β-lactamase selected from SHV-1 , TEM-1 , PDC-3 or KPC-2.

The invention further provides a compound according to the above said for use in the prevention or treatment of bacterial infections, preferably for the treatment of an bacterial infection caused by bacteria of the genus Staphylococcus, preferably Staphylococcus aureus or Staphylococcus epidermidis, Streptococcus, preferably Streptococcus agalactiae, Streptococcus pneumoniae or Streptococcus faecalis, Micrococcus, preferably Micrococcus luteus, Bacillus, preferably Bacillus subtilis, Listerella, preferably Listerella monocytogene, Escherichia, preferably Escherichia coli, Klebsiella, preferably Klebsiella pneumoniae, Proteus, preferably Proteus mirabilis or Proteus vulgaris, Salmonella, preferably Salmonella typhosa, Shigella, preferably Shigella sonnef, Enterobacter, preferably Enterobacter aerogenes or Enterobacter cloacae, Serratia, preferably Serratia marcescens, Pseudomonas, preferably , Acinetobacter, preferably Acinetobacter baumanii, Nocardia, preferably Nocardia autotrophica, or Mycobacterium, preferably Mycobacterium fortuitum.

The invention also provides a method for the preparation of a compound according to the above said wherein the heterocyclic or heteroaromatic ring system Z is synthesized by click chemistry, preferably by cycloaddition, more preferably by 1,3-dipolar cycloaddition, 2,4 cycloaddition or 2+2-cycloaddition, even more preferably by Huisgen 1,3-dipolar cycloaddition or strained- cycloaddtion.

In a preferred embodiment, the method according to the invention comprises the steps

a. providing an alkylboronate, preferably providing an azido- alkylboronate, more preferably a compound of the formula (III)

(III) Χ wherein Y, R, m, L, , X2 are defined as above, wherein are preferably a protectiongroup, more preferably X 1, X2 are pinanediol;

b. sythesis of Z by cycloaddtion, preferably by Cu/C-catalyzed click reaction, even more preferably by Huisgen 1,3-dipolar cycloaddtion, providing a compound of the formula IV wherein Y, m, R , L , Xi, X2 R8, R9 are defined in step a.,

c . optionally purification of the product;

d . optionally deprotection of the protected boronate-group.

In a further preferred embodiment the click reaction is a Cu/C-catalyzed click reaction comprising at least the steps a. providing a solution of an azido-alkylboronoate and an alkyne in dioxane; b. adding Cu(0)/C to the provided solution of step a); c . optionally providing inert gas-influx, preferably Argon-influx, into sealed reaction vessel; d. optionally exposure of the reaction mixture to irradiation, preferably microwave irradiation under stirring at 150°C.

The invention further provides a compound according to one of the formulas (III), (IV), (VIII), (IX).or (X) or a salt thereof, preferably according to one of the formulas III or IV, wherein Y is chosen from hydrogen or from the group consisting of aryl group substituted with from 0 to 3 substituents, heteroaryl group substituted with from 0 to 3 substituents and heterocyclic group substituted with from 0 to 3 substituents, said substituents being independently selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(0)R ,

C(0)NR 4R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxid; is a heteroaryl group substituted with from 0 to 3 substituents, said substituents being independently selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(O)R4, C(0)NR R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido; preferably Y is selected from the group comprising thienyl, benzothienyl, furyl, benzofuryl, dibenzofuryl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, indolyl, quinolyl, isoquinolyl, quinoxalinyl, tetrazolyl, oxazolyl, thiazolyl, isoxazolyl and triazolyl; wherein R and R5 are as defined above; is 0, 1, ,2, 3, 4, 5 or 6, preferably m is 0, 1, 2, more preferably m is 1;

are independently hydroxyl, halogen, NR1R2, C1-C6 alkoxy, or acyloxy; or X and X2 together form a cyclic boron ester, a cyclic boron amide, or a cyclic boron amide-ester, said cyclic boron ester, boron amide or boron amide-ester having a chain or a ring containing from 2 to 12 carbon atoms and, .optionally,

1-3 heteroatoms which can be O, N, or S; wherein R R2 are as defined above; preferably X and X2 are protection groups or do form together one protection group, wherein preferably the protection group is pinanediol; is a bond or a C1-C8 saturated or unsaturated linear or branched aliphatic chain, optionally substituted with one or more groups chosen from hydroxyl, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, amino, aminocarbonyl, carbonyl, aminosulfonyl, alkylaryl, aryl, aryloxy, carboxyl, cyano, guanidine, halogen, heteroaryl, heterocyclyl, sulfide, sulfonyl, sulfoxido, sulfonic acid, sulfate and thiol; R is chosen from hydrogen, linear or branched C1-C12 alkyl, linear or branched C1-C12 alkenyl, or C3-C8 cycloalkyi; said linear or branched C1-C12 alkyl, linear or branched C1-C12 alkenyl, or C3-C8 cycloalkyi being optionally substituted with one or more groups independently chosen from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(O)R4,

C(0)NR R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl,

alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy,

heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl,

aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl,

guanidino, sulfido, and sulfoxido; wherein R4 and R5 are defined as above; and wherein

Re, Rg are independently selected from the group consisting of

(a) hydrogen; (b) C1-C6 alkyl any carbon of which can be substituted with from 0 to 3 substituents, (c) C3-C7 cycloalkyi any carbon of which can be substituted with from 0 to 3 substituents, (d) aryl group substituted with from 0 to 3 substituents (e) heteroaryl group substituted with from 0 to 3 substituents and (f) heterocyclic group substituted with from 0 to 3 substituents,

the substituents being selected from the group consisting of

hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate,

optionally substituted: alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl,

aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl,

alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C6 carbons of (a), any of the carbons of the cycloalkyl group of (b) other than the one attached to the rest of the molecule, or any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, and sulfoxido; most preferably R8 and R9 are selected independently from either hydrogen or a compound according to one of the formulas a., b., c , d., or e.:

" a. > b. COO c. d. OH e.

In a preferred embodiment this compound is an inhibitor for β-lactamase, preferably for a class A , C or D β-lactamase, even more preferably for a β- lactamase selected from SHV-1 , TEM-1 , PDC-3 or KPC-2.

In a further preferred embodiment the invention provides said compound for use in preparation of a compound according to formula I as said above.

It is preferred that the compound is for use in a method according to the invention.

In a further embodiment the invention provides a compound according to the above said for use in manufacturing a medicament for the prevention or treatment of bacterial infections, preferably for the treatment of an bacterial infection caused by bacteria of the genus Staphylococcus, preferably Staphylococcus aureus or Staphylococcus epidermidis, Streptococcus, preferably Streptococcus agalactiae, Streptococcus pneumoniae or Streptococcus faecalis, Micrococcus, preferably Micrococcus luteus, Bacillus, preferably Bacillus subtilis, Listerella, preferably Listerella monocytogene, Escherichia, preferably Escherichia coli, Klebsiella, preferably Klebsiella pneumoniae, Proteus, preferably Proteus mirabilis or Proteus vulgaris, Salmonella, preferably Salmonella typhosa, Shigella, preferably Shigella sonnef, Enterobacter, preferably Enterobacter aerogenes or Enterobacter cloacae, Serratia, preferably Serratia marcescens, Pseudomonas, preferably Pseudomonas aeruginosa, Acinetobacter, preferably Acinetobacter baumanii, Nocardia, preferably Nocardia autotrophica, or Mycobacterium, preferably Mycobacterium fortuitum.

In another aspect the invention provides a pharmaceutical composition comprising a compound according to the invention or a salt thereof.

In a preferred embodiment the pharmaceutical composition comprises a β-lactam antibiotic, preferably a β-lactam antibiotic selected from the group comprising , , , carbapenems, and .

It is further preferred if the pharmaceutical composition further comprises a pharmaceutically acceptable excipient.

In another embodiment the pharmaceutical composition is for use in the prevention or treatment of bacterial infection caused by gram-positive or gram- negative bacteria, preferably for the treatment of an infection caused by bacteria of the genus Staphylococcus, preferably Staphylococcus aureus or Staphylococcus epidermidis, Streptococcus, preferably Streptococcus agalactiae, Streptococcus pneumoniae or Streptococcus faecalis, Micrococcus, preferably Micrococcus luteus, Bacillus, preferably Bacillus subtilis, Listerella, preferably Listerella monocytogene, Escherichia, preferably Escherichia coli, Klebsiella, preferably Klebsiella pneumoniae, Proteus, preferably Proteus mirabilis or Proteus vulgaris, Salmonella, preferably Salmonella typhosa, Shigella, preferably Shigella sonnef, Enterobacter, preferably Enterobacter aerogenes or Enterobacter cloacae, Serratia, preferably Serratia marcescens, Pseudomonas, preferably Pseudomonas aeruginosa, Acinetobacter, preferably Acinetobacter baumanii, Nocardia, preferably Nocardia autotrophica, or Mycobacterium, preferably Mycobacterium fortuitum. In another aspect the invention provides a method for determining β-lactamase activity in a bacterial strain comprising the steps a. cultivation of a first colonie of the bacterial strain in a medium containing an β-lactam antibiotic; b. cultivation of a second colony of the bacterial strain in a medium containing an β-lactam antibiotic and a compound according to the invention; c. comparing the growth rate of the first and the second colony.

Taken together, the present invention relates to compounds having the general formula (I)

Where Y, m , R, and X2, L and Z are defined as above. Particularly, Z is an heterocylce comprising preferably at least one heteroatom selected from N, O and S, more preferably seleceted from N .

Another aspect of the invention is related to a pharmaceutical composition comprising: (a) one or more compounds discussed above; (b) one or more β- lactam antibiotics; and (c) one or more pharmaceutically acceptable carriers

A further aspect of the invention is related to a pharmaceutical composition comprising: (a) one or more compounds discussed above; and (b) one or more pharmaceutically acceptable carriers.

An additional aspect of the invention is related to a method of treating a bacterial infection in a mammal comprising administering to a mammal in need thereof: (a) one or more of the compounds discussed above and (b) and effective amount of β-lactam antibiotic.

Another aspect is for a method of treating a bacterial infection in a mammal comprising administering to a mammal in need thereof an effective amount of a compound discussed above.

An additional aspect of the invention is related to the use of one or more compounds discussed above in the manufacture of a medicament for the treatment of a bacterial infection.

In the context of this invention, when an amount, concentration, or other value or parameter is given as either a range, preferred range, or a list of upper preferable values and lower preferable values, this is to be understood as specifically disclosing all ranges formed from any pair of any upper range limit or preferred value and any lower range limit or preferred value, regardless of whether ranges are separately disclosed. Where a range of numerical values is recited herein, unless otherwise stated, the range is intended to include the endpoints thereof, and all integers and fractions within the range. It is not intended that the scope of the invention be limited to the specific values recited when defining a range.

The present invention relates generally to novel boronic acids and their derivatives which act as broad-spectrum inhibitors of betalactamase enzymes β- Lactamases hydrolyze β-lactam antibiotics, and are therefore an important cause of β-lactam antibiotic resistance.

The present invention also relates to pharmaceutical compositions comprising a compound of the present invention, or salt thereof, an optional β-lactam antibiotic, and a pharmaceutically acceptable excipient.

The present invention also relates to a method for treating a bacterial infection in a mammal by administration of a therapeutically acceptable amount of the aforementioned pharmaceutical compositions. The present invention also relates to a method for increasing the effectiveness of a β-lactam antibiotic in mammals by administering an effective amount of a compound of the present invention in combination with an effective amount of such β-lactam antibiotic.

DEFINITIONS

To facilitate understanding of the disclosure set forth herein, a number of terms are defined below.

Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.

The term "subject" refers to an animal, including, but not limited to, a primate (e.g., human), cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms "subject" and "patient" are used interchangeably herein in reference, for example, to a mammalian subject, such as a human subject, in one embodiment, a human.

The term "mammal" refers to a human, a non-human primate, canine, feline, bovine, ovine, porcine, murine, or other veterinary or laboratory mammal. Those skilled in the art recognize that a therapy which reduces the severity of a pathology in one species of mammal is predictive of the effect of the therapy on another species of mammal.

The terms "treat," "treating," and "treatment" are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or alleviating or eradicating the cause(s) of the disorder, disease, or condition itself. The terms "prevent," "preventing," and "prevention" are meant to include a method of delaying and/or precluding the onset of a disorder, disease, or condition, and/or its attendant symptoms; barring a subject from acquiring a disorder, disease, or condition; or reducing a subject's risk of acquiring a disorder, disease, or condition.

The term "pharmaceutically acceptable carrier," "pharmaceutically acceptable excipient," "physiologically acceptable carrier," or "physiologically acceptable excipient" refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material.

In one embodiment, each component is "pharmaceutically acceptable" in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice of Pharmacy, 21st ed.; Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 6th ed.; Rowe et ai, Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed.; Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd ed.; Gibson Ed.; CRC Press LLC: Boca Raton, FL, 2009.

The term "about" or "approximately" means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term "about" or "approximately" means within 1,2,3, or 4 standard deviations. In certain embodiments, the term "about" or "approximately" means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1 % , 0.5%, or 0.05% of a given value or range.

The terms "active ingredient" and "active substance" refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients, to a subject for treating, preventing, or ameliorating one or more symptoms of a condition, disorder, or disease. As used herein, "active ingredient" and "active substance" may be an optically active isomer, a diasteromeric mixture in any isomeric ratio, or an isotopic variant of a compound described herein.

The term "antibiotic" is used herein to describe a compound or composition which decreases the viability of a microorganism, or which inhibits the growth or reproduction of a microorganism. "Inhibits the growth or reproduction" means increasing the generation cycle time by at least 2-fold, preferably at least 10-fold, more preferably at least 100-fold, and most preferably indefinitely, as in total cell death. As used in this disclosure, an antibiotic is further intended to include an antimicrobial, bacteriostatic, or bactericidal agent. Non-limiting examples of antibiotics useful according to this aspect of the invention include penicillins, cephalosporins, aminoglycosides, sulfonamides, macrolides, tetracyclins, lincosides, quinolones, chloramphenicol, , metronidazole, rifampin, , spectinomycin, trimethoprim, sulfamethoxazole, and others.

The term "β-lactam antibiotic" is used to designate compounds with antibiotic properties containing a β-lactam functionality. Non-limiting examples of β-lactam antibiotics useful according to this aspect of the invention include penicillins, cephalosporins, penems, carbapenems, and monobactams. β-Lactam antibiotics are effective (in the absence of resistance) against a wide range of bacterial infections. These include those caused by both gram-positive and gram-negative bacteria, for example, bacteria of the genus Staphylococcus (such as Staphylococcus aureus and Staphylococcus epidermidis), Streptococcus (such as Streptococcus agalactiae, Streptococcus pneumoniae and Streptococcus faecalis), Micrococcus (such as Micrococcus luteus) , Bacillus (such as Bacillus subtilis), Listerella (such as Listerella monocytogenes), Escherichia (such as Escherichia coli), Klebsiella (such as Klebsiella pneumoniae), Proteus (such as Proteus mirabilis and Proteus vulgaris), Salmonella (such as Salmonella typhosa), Shigella (such as Shigella sonne ), Enterobacter (such as Enterobacter aerogenes and Enterobacter cloacae), Serratia (such as Serratia marcescens), Pseudomonas (such as Pseudomonas aeruginosa), Acinetobacter (such as Acinetobacter baumanii), Nocardia (such as Nocardia autotrophica), and Mycobacterium (such as Mycobacterium fortuitum).

The term "β-lactamase" means an enzyme produced by a bacterium that has the ability to hydrolyze the β-lactam ring of β-lactam antibiotics. Such enzymes are often classified into 4 major classes (Classes A , B, C, and D) according to the so- called Ambler classification scheme, based principally on protein homology.

The term "β-lactamase inhibitors with broad-spectrum functionality" as used herein refers to the ability of an inhibitor to inhibit a broad range of β-lactamase enzymes, spanning multiple subtypes from multiple classes (for example numerous enzyme subtypes from both Ambler Class A and Ambler Class C).

In some embodiments, β-lactamase enzyme(s) from at least two classes of β- lactamase enzymes are inhibited by a compound disclosed herein, with preferred embodiments being those where β-lactamase enzyme(s) from more than two classes of β-lactamase enzymes are inhibited by a compound disclosed herein.

The term "comprising" is intended to include embodiments encompassed by the terms "consisting essentially of and "consisting of.

Similarly, the term "consisting essentially of is intended to include embodiments encompassed by the term "consisting of. The terms "effective amount", "therapeutically effective amount", and "therapeutically effective period of time" are used to denote known treatments at dosages and for periods of time effective to show a meaningful patient benefit, i.e., healing of conditions associated with bacterial infection, and/or bacterial drug resistance.

Preferably, such administration should be parenteral, oral, sublingual, transdermal, topical, intranasal, intratracheal, or intrarectal. When administered systemically, the therapeutic composition is preferably administered at a sufficient dosage to attain a blood level of inhibitor of at least about lOC^g/mL, more preferably about 1 mg/mL, and still more preferably about 10mg/mL. For localized administration, much lower concentrations than this may be effective, and much higher concentrations may be tolerated.

CHEMICAL DEFINITIONS

The term alkyl means both straight and branched saturated hydrocarbon chain of 1-12 carbons, preferably of 1-8 carbon atoms, which may be optionally substituted. Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl (including all isomeric forms), n-propyl, isopropyl, butyl (including all isomeric forms), n-butyl, isobutyl, sec-butyl, t-butyl, pentyl (including all isomeric forms), and hexyl (including all isomeric forms).

The term alkenyl means both straight and branched chain containing at least a carbon-carbon double bond, of 1-12 carbon atoms, optionally substituted. Preferably the alkenyl moiety has 2-8 carbons and one or two double bonds.

Such alkenyl moieties may exist in the E or Z conformations; the compounds of this invention include both conformations. Examples of alkenyl groups include, but are not limited to, methlenyl, ethelenyl, propenyl (including all isomeric forms), n- propenyl, isopropenyl, butenyl (including all isomeric forms), n-butenyl, isobutenyl, pentenyl (including all isomeric forms), and hexenyl (including all isomeric forms).

The term alkynyl includes both straight chain and branched chain, optionally substituted, containing 2-12 carbon atoms and at least one triple bond, preferably the alkynyl moiety has 2-6 carbons and one or two triple bonds. Examples of alkynyl groups include, but are not limited to, ethynyl (-CECH), propynyl (including all isomeric forms, e.g., 1-propynyl (-CECCH3) and propargyl (-CH2CECH)), butynyl (including all isomeric forms, e.g., 1-butyn-1-yl and 2-butyn-1-yl), pentynyl (including all isomeric forms, e.g., 1-pentyn-1-yl and 1-methyl-2-butyn-1-yl), and hexynyl (including all isomeric forms, e.g., 1-hexyn-1-yl).

The term cycloalkyl means an alicyclic hydrocarbon group having 3-14 carbon atoms, optionally substituted, preferably the cycloalkyl moiety contain 3-8 carbon atoms. In one embodiment, cycloalkyl groups may be saturated or unsaturated but non-aromatic, and/or bridged, and/or non-bridged, and/or fused bicyclic groups. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptenyl, bicyclo[2.1 .1]hexyl, bicyclo[2.2.1]heptyl, decalinyl, and adamantyl.

The term aryl means a monovalent monocyclic aromatic group and/or monovalent multicyclic aromatic group that contain at least one aromatic carbon ring. Examples of aryl groups include, but are not limited to, phenyl, α-naphthyl, β- naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl. Aryl also refers to bicyclic or tricyclic carbon rings, where one of the rings is aromatic and the others of which may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetralinyl). In certain embodiments, aryl may be optionally substituted.

Heteroaryl is defined as an aromatic heterocyclic ring system (monocyclic or bicyclic) where the heteroaryl moieties are selected from, but not limited to,: (1) furan, thiophene, indole, azaindole, oxazole, thiazole, isoxazole, isothiazole, imidazole, N-methylimidazole, pyridine, pyrimidine, pyrazine, pyrrole, N- methylpyrrole, pyrazole, N-methyl pyrazole, 1,3,4-oxadiazole, ,2,4-triazole, 1- methyl-1 ,2,4-triazole, 1 H-tetrazole, 1-methyltetrazole, 1,2,4-thiadiazole, 1,3,4- thiadiazole, ,2,3-thiadiazole, 1,2,3-triazole, 1-methyl-1 ,2,3-triazole, benzoxazole, benzothiazole, benzofuran, benzisoxazole, benzimidazole, N- methylbenzimidazole, azabenzimidazole, indazole, quinazoline, quinoline, and isoquinoline; (2) a bicyclic aromatic heterocycle where a phenyl, pyridine, pyrimidine or pyridizine ring is: (a) fused to a 6-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom; (b) fused to a 5 or 6-membered aromatic (unsaturated) heterocyclic ring having two nitrogen atoms; (c) fused to a 5-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom together with either one oxygen or one sulfur atom; or (d) fused to a 5-membered aromatic (unsaturated) heterocyclic ring having one heteroatom selected from 0, Nor S.

Arylalkyl is defined as aryl-C1-C12alkyl~. Arylalkyl moieties include benzyl, 1- phenylethyl, 2-phenylethyl, 3-phenylpropyl, 2-phenylpropyl and the like. In certain embodiments, arylakyl are optionally substituted with one or more substituents.

Arylalkenyl is defined as aryl-C2-C12alkenyl-. Arylalkenyl moieties include, 1- phenylethenyl, 2-phenylethenyl, 1-phenylpropenyl, 2-phenylpropenyl and the like. Such arylalkenyl moieties may exist in the E or Z conformations and be optionally substituted with one or more substituents.

Arylalkynyl is defined as aryl-C2-C12alkynyl~. Arylalkynyl moieties include, phenylethynyl, phenylpropynyl, and the like.

Alkylaryl is defined as C1-C12alkyl-aryk

Alkenylaryl is defined as C2-C12 alkenylaryl-.

Alkynylaryl is defined as C2-C12alkynylaryl-.

Heteroarylalkyl is defined as heteroaryl-C 1-C12alkyl—.

Alkylheteroaryl is defined as C1-C12alkyl-heteroaryl~.

Heteroaryl is defined as a monovalent monocyclic aromatic group or monovalent multicyclic aromatic group that contain at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from 0, S, and N in the ring. Heteroaryl groups are bonded to the rest of a molecule through the aromatic ring. Examples of monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl. Examples of bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quinazolinyl, thiadiazolopyrimidyl, and thienopyridyl. Examples of tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl. In certain embodiments, heteroaryl may also be optionally substituted.

Heterocyclyl is defined as a monovalent monocyclic non-aromatic ring system or monovalent multicyclic ring system that contains at least one non-aromatic ring, wherein one or more of the non-aromatic ring atoms are heteroatoms independently selected from 0, S, and N ; and the remaining ring atoms are carbon atoms. In certain embodiments, the heterocyclyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may be fused or bridged, and in which nitrogen or sulfur atoms may be optionally oxidized, nitrogen atoms may be optionally quaternized, and some rings may be partially or fully saturated, or aromatic. The heterocyclyl may be attached to the main structure at any heteroatom or carbon atom which results in the creation of a stable compound. Examples of such heterocyclic groups include, but are not limited to, aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, azepinyl, hexahydroazepinyl, piperazinyl, morpholinyl, thiomorpholinyl, 1,4-dioxanyl, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl, benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, -carbolinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, decahydroisoquinolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, dihydrobenzisothiazinyl, dihydrobenzisoxazinyl, dihydrofuryl, dihydroisoindolyl, dihydropyranyl, dihydropyrazolyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dioxolanyl, dihydrotriazolyl, dihydroazetidinyl, dihydro-1 ,4-dioxanyl, 1,4-dithianyl, furanonyl, imidazolidinyl, imidazolinyl, indolinyl, isobenzotetrahydrofuranyl, isobenzotetrahydrothienyl, isochromanyl, isocoumarinyl, isoindolinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, oxazolidinonyl, oxazolidinyl, oxiranyl, piperazinyl, piperidinyl, 4-piperidonyl, pyrazolidinyl, pyrazolinyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydropyranyl, tetrahydrothienyl, thiamorpholinyl, thiazolidinyl, tetrahydroquinolinyl, and 1,3,5-trithianyl. In certain embodiments, heterocyclic may also be optionally substituted.

The term halogen is defined as CI, Br, F, and I.

Alkoxy is defined as C1-C6alkyl-O~.

Cycloalkoxy is defined as C3-C7cycloalkyl-O-.

Aryloxy is defined as aryl-O-.

Heteroaryloxy is defined as heteroaryl-O-.

Heterocyclyloxy is defined as C3-C7heterocyclyl-0-.

Sulfonic acid is defined as -S03H.

Sulfate is defined as -OS03H.

Amino is defined as -NH2.

Cyano is defined as --CN

Hydroxyl is defined as -OH

Thiol is defined as --SH

Carboxyl is defined as -CO2H.

Oxo is defined as double bonded oxygen. Trialkylammonium is defined as (A1 )(A2)(A3)N+- where A 1, A2 and A3 are independently alkyl, cycloalkyl, heterocyclyl and the nitrogen is positively charged.

Carbonyl is defined as -C(O)- where the carbon is optionally substituted and also attached to the rest of the molecule.

Aminocarbonyl is defined as -C(0)-N--, where the carbon is optionally substituted and the nitrogen is attached to the rest of the molecule.

Oxycarbonyl is defined as ~C(0)-0~, where the carbon is optionally substituted and the oxygen is attached to the rest of the molecule.

Aminosulfonyl is defined as —S(0)2-N- where the sulfur is optionally substituted and the nitrogen is attached to the rest of the molecule.

Sulfonyl is defined as -S(0)2- where the sulfur is bonded to an optional substituent and also to the rest of the molecule.

Guanidino is defined as ~N1 (H)-C(NH)-N2(H)~ where N 1 is optionally substituted and N2 is attached to the rest of the molecule.

Oxyimino is defined as (N-O-A) where the nitrogen is double bonded to a carbon which is attached to the rest of the molecule and A can be hydrogen, optionally substituted: alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl.

Sulfido is defined as -S- where sulfur is bound to an optional substituent and also to the rest of the molecule.

Sulfoxido is defined as ~S{O)~ where sulfur is bound to an optional substituent and also to the rest of the molecule.

Where a group or atom is described as "optionally substituted" one or more of the following substituents may be present on that group or atom: hydrogen, OH,

halogen, cyano, nitro, C(O)Ra, -C(O)ORa, -C(O)NRbRc, -C(=NRa)NRbRc, -ORa, -

OC(O)Ra, -OC(O)ORa, -OC(O)NR bRc, -OC(=NRa)NR Rc, -OS(O)R, -OS(O)nR a, -

OS(O)NR Rc, -OS(O)nNR bRc, _NRbRc, -NRaC(O)Rd, -NRaC(O)ORd, - NR C( )NR Rc, -NR C(=NR )NR R -NR S(O)R , -NR S(O)nR ,-NRaS(O)nR Rc, a O b a d b C a d a d b

-NRaS(O)nNRbRc, -SRa, -S(O)Ra, -S(O)nR a, -S(O)nR Rc alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, cycloalkoxy, heterocyclyloxy, aryloxy, arylamino, heteroarylamino, heteroaryloxy, arylakyl, alkylaryl, arylalkenyl, alkenylaryl, alkynylaryl, arylalkynyl, heteroarylalkyl, alkylheteroaryl, heteroarylalkenyl, alkenylheteroaryl, heteroarylalkynyl, alkynylheteroaryl, trialkylammonium. Optional substituents may be attached to the group or atom which they substitute in a variety of ways, either directly or through a connecting group of which the following are examples: alkyl, amine, amide, ester, ether, thioether, sulfonamide, sulfamide, sulfoxide, urea. As appropriate an optional substituent may itself be further substituted by another substituent, the latter being connected directly to the former or through a connecting group such as those exemplified above.

In certain embodiments, "optically active" and "enantiomerically active" refer to a collection of molecules, which has an enantiomeric excess of no less than about 50%, no less than about 70%, no less than about 80%, no less than about 90%, no less than about 9 1 %, no less than about 92%, no less than about 93%, no less than about 94%, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%. In certain embodiments, the compound comprises about 95% or more of one enantiomer and about 5% or less of the other enantiomer based on the total weight of the compound.

In describing an optically active compound, the prefixes R and S are used to denote the absolute configuration of the molecule about its chiral center( s). The (+) and ( -) are used to denote the optical rotation of the compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound. The (-) prefix indicates that the compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or counterclockwise. The (+) prefix indicates that the compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise. The term "isotopic variant" refers to a compound that contains an unnatural proportion of an isotope at one or more of the atoms that constitute such compounds. In certain embodiments, an "isotopic variant" of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen H), deuterium H , tritium 3H), carbon-1 (1 C), carbon-12 (12C), carbon-13 ( C), carbon-14 ( C , nitrogen-13 ( 3N), nitrogen-14 ( 4 N , nitrogen-15 (15 N), oxygen-14 ( 4 0), oxygen-15 (1 0), oxygen-16 ( 60), oxygen-17 ( 0), oxygen- 18 ( 80), fluorine-17 ( F), fluorine-18 ( F , phosphorus-31 (3 P), phosphorus-32 (32P), phosphorus-33 (33P), sulfur-32 (32S), sulfur-33 ( S), sulfur-34 (^S), sulfur- 35 ( S), sulfur-36 (36S), chlorine-35 (3 CI), chlorine-36 (36CI), chlorine-37 (37CI), bromine-79 (79Br), bromine-81 (8 1Br), iodine-123 ( 23l), iodine-125 (125l), iodine- 127 (127l), iodine-129 ( 29l), and iodine-131 (131 l). In certain embodiments, an "isotopic variant" of a compound is in a stable form, that is, non-radioactive. In certain embodiments, an "isotopic variant" of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen (1H), deuterium ( H), carbon-12 (1 C), carbon-13 ( C), nitrogen-14 ( 4 N), nitrogen-15 ( N), oxygen-16 (160 , oxygen-17 (170), oxygen-18 ( 0), fluorine-17 ( F), phosphorus-31 (3 P), sulfur-32 ( 2S), sulfur-33 ( S), sulfur-34 (^S), sulfur-36 (36S), chlorine-35 (35CI), chlorine-37 (37CI), bromine-79 ( Br), bromine-81 ( Br), and iodine-127 ( l). In certain embodiments, an "isotopic variant" of a compound is in an unstable form, that is, radioactive. In certain embodiments, an "isotopic variant" of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, tritium (3H), carbon-1 1 ( C), carbon-14 (1 C), nitrogen-13 (1 N), oxygen-14 ( 0), oxygen-1 5 ( 50), fluorine-18 ( F , phosphorus- 32 (32P), phosphorus-33 (33P), sulfur-35 ( S), chlorine-36 (3 CI), iodine-123 ( 23l), iodine-125 ( 5l), iodine-129 ( 29l), and iodine-131 ( 3 1l). It will be understood that, in a compound as provided herein, any hydrogen can be 2H , for example, or any carbon can be 1 C, as example, or any nitrogen can be 5N, as example, and any oxygen can be 1 0 , where feasible according to the judgment of one of skill. In certain embodiments, an "isotopic variant" of a compound contains unnatural proportions of deuterium. The term "solvate" refers to a complex or aggregate formed by one or more molecules of a solute, e.g., a compound provided herein, and one or more molecules of a solvent, which present in stoichiometric or non-stoichiometric amount. Suitable solvents include, but are not limited to, water, methanol, ethanol, n-propanol, isopropanol, and acetic acid. In certain embodiments, the solvent is pharmaceutically acceptable. In one embodiment, the complex or aggregate is in a crystalline form. In another embodiment, the complex or aggregate is in a noncrystalline form. Where the solvent is water, the solvate is a hydrate.

Other objects and advantages will become apparent to those skilled in the art upon reference to the detailed description that hereinafter follows. Therein the figures show the following

Fig. 1 scheme of synthesis of a first compound according to the invention;

Fig. 2 scheme of synthesis of a second compound according to the invention with

a) (+)-pinanediol; b) LiChCI2, THF, -100°C, c) LiN(TMS) 2, THF, -80°C; d)

MeOH; then RCOCI, THF, 0°C, e) PhB(OH) 2, CH3CN, Hexane, HCI, rt, wherein SG = protection group, particularly pinanediol;

Fig. 3 scheme of synthesis of a third compound according to the invention with a)

BuLi, THF, -78°C, then B(OMe)3, then (+)-pinanediol; b) LiCHCI2, THF, -

100°C; c) LiN(TMS) 2, THF, -80°C; d) MeOH, then RCOCI, THF, 0°C; e)

PhB(OH) 2, CH3CN, Hexane, HCI, rt; wherein SG = protection group, particularly pinanediol;

Fig. 4 scheme of synthesis of a fourth compound according to the invention with

a) Catecholborane, THF, then (+)-pinandiol; b) LiCHCI2, THF, -100 °C; c)

LiN(TMS) 2, THF, -80 °C; d) MeOH, then RCOCI, THF, 0 °C; e) PhB(OH)2,

CH3CN, Hexane, HCI, rt; wherein SG = protection group, particularly pinanediol; Fig. 5 Residual velocities of nitrocefin hydrolysis together with 20 µΜ concentration of compounds 7a-e against the β-lactamases SHV-1 , TEM-1 , PDC-3, and KPC-2

A preferred embodiment relates to compounds of general formula II. Such a compound can be obtained by the synthesis shown in Fig. 1 and described as a retro-synthesis hereinafter.

In particular compounds of general formula XI, corresponding to said general

formula II, where Xi and X2 are OH, and Re, R g are as defined above, particularly R8 and R9 are selected from the group consisting of hydrogen; C1-C6 alkyl any carbon of which can be substituted with from 0 to 3 substituent; C3-C7 cycloalkyl any carbon of which can be substituted with from 0 to 3 substituents; aryl group substituted with from 0 to 3 substituents; heteroaryl group substituted with from 0 to 3 substituents and heterocyclic group substituted with from 0 to 3 substituents, the substituents being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido; most preferably R8 and R9 are selected independently from either hydrogen or a compound according to one of the formulas a., b., c , d., or e.:

can be obtained from compounds of general formula XII by using methods described in the literature for the conversion of boronates to afford boronic acids, for example treatment with a lewis acid like boron trichloride (Jin, S., Zhu, C. Y., Cheng Y. F., Li M. Y., Wang, B. H., Biorg. Med. Chem., 2010, 18, 1449-1455) sodium periodate (Courts, S. J., Adams, J., Krolikowski, D., Snow, R. J, Tetrahedron Lett, 1994, 35, 5109-51 12), via fluoroborane intermediates, (Inglis, S. R., Woon, E . C , Thompson, A . L , Schofield, C. J., J. Org. Chem., 2010, 75, 468- 471), by heating with hydrochloric acid (Martin, R., Bryan Jone, J., Tetrahedron Lett., 1995, 36, 8399-8402), through a biphasic transesterification method in

CH3CN/n-hexane or water/Et 20 with phenyl boronic acid (Zhu, Y., Zhu, X., Wu, Y., Ma, Y., Li, Y., Zhao, X., Yuan, Y., Yang, J., Yu, S., Shao, F., Li, R., Ke, Y., Lu, A., Liu, Z., Zhang L., J. Med Chem., 2010, 53, 1990-1999).

Χ Compounds of general formula XII where R e = H, and , X2 are a protection group, particularly (+)-pinanediol, can be obtained from compound of general formula XIII through an orthogonal cycloaddition reaction between compound XIII and a suitable alkyne. This reaction can be performed using Cu(l) as catalysis in a wide variety of conditions including acqueous or organic solvents and mixtures thereof like tetrahydrofurane (THF), acetonitrile (CH3CN), dioxane, water and ethanol, water and ethanol and tert-butylalcohol, etc... The reaction can be carried out at a temperature ranging from room to boiling temperatures, using a base such as N,N-diisopropylethylamine (DIPEA), triethylamine (TEA), lutidine, pyridine etc., a suitable ligand like Tris[(1-benzyl-1H-1 ,2,3-triazol-4-yl)methyl] amine (TBTA), proline, lutidine etc.... and a large choice of Cu(l) source such as copper halides (Cul, CuBr), copper sulfate (CuS0 4) and sodium ascorbate (NaAsc), Cu(0) with or without addition of CuS0 .(Hein J. E., Fokin, V.V.; Chem. Soc. Rev., 2010, 39, 1302-1315) Alternatively, compounds of general formula XII where R g = H can be obtained through catalysis of pentamethylcyclopentadienyl ruthenium chloride [Cp*RuCI] complexes (Boren, B. C , Narayan, S., Rasmussen, L . K., Zhang, L , Zhao, H., Lin, Z., Jia, G., Fokin, V. V., J. Amer. Chem. Soc, 2008, 130, 8923-8930).

XII XIII

Finally compound XIII can be obtained from compound XIV by conversion to the bis(trimethylsilyl)amine intermediate using conditions as described in Morandi F. et al., J. Amer. Chem. Soc, 2003, 125, 685-695, followed by coupling reaction with a suitable acid chloride or with an active ester such as that derived from the reaction of a carboxylic acid and isobutyl chloroformate or from the reaction of a carboxylic acid with a tetramethyluronium agent such as 0-(7-Azabenzotriazol-1- yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU). Compound XIV can be obtained from compound XV by treatment with chloromethyllithium at -100 °C according to the well known Matteson protocol (Sadhu, K. M., Matteson, D. S., Organometallics, 1985, 4, 1687-1689) whereas the a-azidomethane boronic ester XV can be obtained by nuleophilic substitution of the a-halogenated derivative XVI as described in Matteson D. S. et al, J. Org. Chem., 2008, 693, 2258-2262.

Within all compounds XIII, XIV, XV and XVI X2 are a protection group, particularly (+)-pinanediol; HAL is a halogen. XIII XIV XV XVI

The present invention relates to compounds of general formula I that can be prepared according but not limited to the procedures shown schematically in Fig. 1 and described herein as an example or obtained by any method known to a person skilled in the art.

Certain compounds of Formula (I) include compounds of Formula (II)

Wherein Y is chosen from hydrogen, optionally substituted linear or branched C1-

C12 alkyl, optionally substituted linear or branched C1-C12 alkenyl, NR1R2, OR3, SR3, or from the group consisting of aryl group substituted with from 0 to 3 substituents, heteroaryl group substituted with from 0 to 3 substituents and heterocyclic group substituted with from 0 to 3 substituents, said substituents being independently selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(0)R4, C(0)NR R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido;

Ri, R2, R3, R , R 5 are independently selected from the group consisting of: (a) hydrogen; (b) C1-C10 alkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents; (c) C3-C10 cycloalkyi any carbon of which can be substituted with 0, 1, 2 or 3 substituents;

(d) heteroaryl group substituted with 0, 1, 2 or 3 substituents; (e) heterocyclic group substituted with 0, 1, 2 or 3 substituents; (f) C1-C12 alkyl, C1-C12 cycloalkyi, C1-C12 alkenyl; (g) aryl group substituted with 0, , 2, 3, 4 or 5 substituents;

(h) C(0)R 6; -C(0)NR 6R7; -C(O)OR 6, C(=NR6) ; NR R7, -C(=NR6)R7; (i) C1-C6 alkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents; (j) C3-C7 cycloalkyi any carbon of which can be substituted with 0, 1, 2 or 3 substituents; said substituents of Ri, R2, R3, and R being independently selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(0)R 6, C(0)NR 6R , thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C10 carbons of (b), any of the carbons of the cycloalkyi group of (c) other than the one attached to the rest of the molecule, any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule, any of the C1-C6 carbons of (i) or any of the carbons of the cycloalkyi group of (j) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, sulfoxido; and R6 and R7 being independently selected from hydrogen, or from the group consisting of (a) C1-C6 alkyl any carbon of which can be substituted with 0 , 1, 2 or 3 substituents, (b) C3-C7 cycloalkyi any carbon of which can be substituted with 0, , 2 or 3 substituents, (c) aryl group substituted with 0, 1, 2 or 3 substituents (d) heteroaryl group substituted with 0, 1, 2 or 3 substituents and (e) heterocyclic group substituted with 0, 1, 2 or 3 substituents, the substituents of and R7 being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C6 carbons of (a), any of the carbons of the cycloalkyl group of (b) other than the one attached to the rest of the molecule, or any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, and sulfoxido;

m is 0, 1, 2 or 3;

L is CH2;

R is hydrogen;

Re R are selected from the group consisting of hydrogen; C1-C6 alkyl any carbon of which can be substituted with from 0 to 3 substituent; C3-C7 cycloalkyl any carbon of which can be substituted with from 0 to 3 substituents; aryl group substituted with from 0 to 3 substituents; heteroaryl group substituted with from 0 to 3 substituents and heterocyclic group substituted with from 0 to 3 substituents, the substituents being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido; most preferably R8 and R9 are selected independently from either hydrogen or a compound according to one of the formulas a., b., c , d., or e.: a. < b. - c. - . e.

X and X 2 are O H .

The compound provided herein can be prepared, isolated, or obtained by any method known to one of skill in the art.

A particularly preferred embodiment of the invention is related to compounds 7a-e Most preferred compounds are compounds 7d-e.

When the compound provided herein contains an acidic or basic moiety, it may also be provided as a pharmaceutically acceptable salt. See, Berge et al., 1. Pharm. Sci. 1977,66, 1-19; and Handbook of Pharmaceutical Salts, Properties, and Use; Stahl and Wermuth, Ed.; Wiley-VCR and VRCA: Zurich, Switzerland, 2002.

Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzene sulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+ )-camphoric acid, camphorsulfonic acid, (+ HiSJcamphor-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane-1 ,2-disulfonic acid, ethane sulfonic acid, 2-hydroxy- ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, D-gluconic acid, D-glucuronic acid, L-glutamic acid, uoxoglutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, hydroiodic acid, (+)-L-lactic acid, (±)-DL-lactic acid, lactobionic acid, lauric acid, maleic acid, (-)-L-malic acid, malonic acid, (±)-DL-mandelic acid, methane sulfonic acid, naphthalene-2-sulfonic acid, naphthalene-1 ,5-disulfonic acid, 1-hydroxy-2- naphthoic acid, nicotinic acid, nitric acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, perchloric acid, phosphoric acid, L-pyroglutamic acid, saccharic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p- toluenesulfonic acid, undecylenic acid, and valeric acid.

Suitable bases for use in the preparation of pharmaceutically acceptable salts, including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2-( diethylamino )-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, N-methyl-glucamine, hydrabamine, 1H- imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, 1-(2-hydroxyethyl)-pyrrolidine, pyridine, quinuclidine, quinoline, isoquinoline, secondary amines, triethanolamine, trimethylamine, triethylamine, N -methyl-D-glucamine, 2-amino-2-(hydroxymethyl)- 1 ,3-propanediol, and tromethamine.

The compound provided herein may also be provided as a prodrug, which is a functional derivative of the compound itself and is readily convertible into the parent compound in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound. A prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis.

ADMINISTRATION OF BETALACTAMASE INHIBITORS

β-Lactamase inhibitors can be administered to subjects in a biologically compatible form suitable for pharmaceutical administration in vivo to, e.g., increase antibacterial activity of β-lactam antibiotics. Administration of a β- lactamase inhibitor as described herein can be in any pharmacological form including a therapeutically active amount of a β-lactamase inhibitor alone or in combination with a pharmaceutically acceptable carrier.

A therapeutically active amount of a β-lactamase inhibitor may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the betalactamase inhibitor to elicit a desired response in the subject. Dosage regimes may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily, or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.

The therapeutic or pharmaceutical compositions can be administered by any suitable route known in the art including, for example, intravenous, subcutaneous, intramuscular, oral, buccal, intranasal, suppository, transdermal, intrathecal, or intracerebral or administration to cells in ex vivo treatment protocols. Administration can be either rapid as by injection or over a period of time as by slow infusion or administration of slow release formulation.

The pharmaceutical compositions can also be formulated as modified release dosage forms, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated-, fast-, targeted-, programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Delivery Technology, 2nd ed.; Rathbone et al, Eds.; Marcel Dekker, Inc.: New York, NY, 2008).

A β-lactamase inhibitor can also be linked or conjugated with agents that provide desirable pharmaceutical or pharmacodynamic properties. For example, a β- lactamase inhibitor can be coupled to any substance known in the art to promote penetration or transport across the blood-brain barrier such as an antibody to the transferrin receptor, and administered by intravenous injection (see, e.g., Friden PM et al, Science 259:373-77 (1993».

Furthermore, a β-lactamase inhibitor can be stably linked to a polymer such as polyethylene glycol to obtain desirable properties of solubility, stability, half-life, and other 5 pharmaceutically advantageous properties (see, e.g., Davis et al., Enzyme Eng. 4:169-73 (1978); Burnham NL, Am. J. Hosp. Pharm. 5 1 :210-18 (1994)). Furthermore, a β-lactamase inhibitor can be in a composition which aids in delivery into the cytosol of a cell. For example, the β-lactamase inhibitor may be conjugated with a carrier moiety such as a liposome that is capable of delivering the β-lactamase inhibitor into the cytosol of a cell. Such methods are well known in the art (see, e.g., Amselem S et al., Chem. Phys. Lipids 64:219-37 (1993)). Alternatively, a β-lactamase inhibitor can be modified to include specific transit peptides or fused to such transit peptides which are capable of delivering their β-

lactamase inhibitor into a cell. In addition, the β-lactamase inhibitor can be delivered directly into a cell by microinjection.

The compositions are usually employed in the form of pharmaceutical preparations. Such preparations are made in a manner well known in the pharmaceutical art. One preferred preparation utilizes a vehicle of physiological saline solution, but it is contemplated that other pharmaceutically acceptable carriers such as physiological concentrations of other non-toxic salts, five percent aqueous glucose solution, sterile water, or the like may also be used.

As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any standard media or agent is incompatible with the active compound, use thereof in the therapeutic compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions. It may also be desirable that a suitable buffer be present in the composition. Such solutions can, if desired, be lyophilized and stored in a sterile ampoule ready for reconstitution by the addition of sterile water for ready injection. The primary solvent can be aqueous or alternatively non-aqueous.

A β-lactamase inhibitor can also be incorporated into a solid or semi-solid biologically compatible matrix which can be implanted into tissues. The carrier can contain other pharmaceutically-acceptable excipients for modifying or maintaining the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation. Such excipients are those substances usually and customarily employed to formulate dosages for parenteral administration in either unit dosage or multi-dose form or for direct infusion by continuous or periodic infusion.

In some embodiments, the pharmaceutical compositions further comprise an effective amount of a β-lactam antibiotic. Exemplary β-lactam antibiotics include penicillins, cephalosporins, , carbapenems, monobactams, bridged monobactams, or a combination thereof. Pencillins include, but are not limited to, benzathine penicillin, , , procaine penicillin, , , dicloxaciliin, , temociilin, amoxicillin, ampicillin, co-amoxiclav, , , ticarcillin, , piperacillin, apalcillin, , bacampiciliin, , mecicilam, pevmecillinam, , talapiciliin, aspoxicillin, , , , or a combination thereof. Cephalosporins include, but are not limited to, cephalothin, cephaloridin, , , , , cephalexin, cephradine, , , cephacetrll, , , , cefoperazone, ceftizoxime, cefinenoxime, cefinetazole, cepha[oglycin, , , , , , , , , cefepim, , , , , , , , , axetil, cefpodoxime proxetil, pivoxil, pivoxil, pivoxil, pivoxil, , , loracarbacef, , anti-methicillin-resistant Staphylococcus aureus (MRSA) cephalosporins (e.g., or ceftaroline), FR264205 (see Takeda et al., Antimicrob. Agents Chemother. 5 1 :826-30 (2007)), or a combination thereof. Penem include but are not limited to . Carbapenems include, but are not limited to, , , , , , , anti-MRSA carbapenems (e.g., (PZ-601) or ME1 036, see Expert Rev. Anti-lnfect Ther. (2008) 6:39-49), or a combination thereof. Monobactams include, but are not limited to, , , BAL30072 or a combination thereof. See Figure 2 for structures of razupenem (PZ-601), ME1036, and BAL30072.

The β-lactamase inhibitors or their pharmaceutically acceptable salts may be administered at the same time as the dose of betalactam antibiotics or separately. This may be carried out in the form of a mixture of the two active ingredients or in the form of a pharmaceutical combination of the two separate active ingredients.

The dosage of the β-lactamase inhibitors and of their pharmaceutically acceptable salts may vary within wide limits and should naturally be adjusted, in each particular case, to the individual conditions and to the pathogenic agent to be controlled. In general, for a use in the treatment of bacterial infections, the daily dose may be between 0.10 g and 10 g per day, by the oral route in humans, or else between 0.10 g and 10 g per day by the intramuscular or intravenous route. Moreover, the ratio of the β-lactamase inhibitor or of the pharmaceutically acceptable salt thereof to the β-lactam antibiotic may also vary within wide limits and should be adjusted, in each particular case, to the individual conditions. In general, a ratio ranging from about 1 :20 to about 4: 1 is recommended.

Dose administration can be repeated depending upon the pharmacokinetic parameters of the dosage formulation and the route of administration used.

It is also provided that certain formulations containing a β-lactamase inhibitor are to be administered orally. Such formulations are preferably encapsulated and formulated with suitable carriers in solid dosage forms. Some examples of suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, gelatin, syrup, methyl cellulose, methyl- and propylhydroxybenzoates, talc, magnesium, stearate, water, mineral oil, and the like. The formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents, or flavoring agents. The compositions may be formulated so as to provide rapid, sustained, or delayed release of the active ingredients after administration to the patient by employing procedures well known in the art. The formulations can also contain substances that diminish proteolytic degradation and/or substances which promote absorption such as, for example, surface active agents.

It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage 5 unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals. The specific dose can be readily calculated by one of ordinary skill in the art, e.g., according to the approximate body weight or body surface area of the patient or the volume of body space to be occupied. The dose will also be calculated dependent upon the particular route of administration selected. Further refinement of the calculations necessary to determine the appropriate dosage for treatment is routinely made by those of ordinary skill in the art. Such calculations can be made without undue experimentation by one skilled in the art in light of the activity disclosed herein in assay preparations of target cells. Exact dosages are determined in conjunctiqn with standard dose-response studies. It will be understood that the amount of the composition actually administered will be determined' by a practitioner, in the light of the relevant circumstances including the condition or conditions to be treated; the choice of composition to be administered; the age, weight, and response of the individual patient; the severity of the patient's symptoms; and the chosen route of administration.

Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be

expressed as the ratio LD5o/ED50. Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.

The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds

lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the methods disclosed herein, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that

includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels In plasma may be measured, for example, by high performance liquid chromatography.

INHIBITION OF BACTERIAL GROWTH

The present disclosure also provides methods for inhibiting bacterial growth, by e.g. reducing bacterial resistance to a β-lactam antibiotic, such methods comprising contacting a bacterial cell culture, or a bacterially infected cell culture, tissue, or organism, with a β-lactamase inhibitor described herein.

Preferably, the bacteria to be inhibited by administration of a β-lactamase inhibitor of the invention are bacteria that are resistant to β-iactam antibiotics. More preferably, the bacteria to be inhibited are β-lactamase positive strains that are highly resistant to β-lactam antibiotics. The terms "resistant" and "highly resistant" are well-understood by those of ordinary skill in the art (see, e.g., Payne et al., Antimicrobial Agents and Chemotherapy 38:767-772 (1994); Hanaki et al., Antimicrobial Agents and Chemotherapy 30:1 120-1 126 (1995).

Preferably, highly resistant bacterial strains are those against which the MIC of methicillin is > 00 µg/mL Preferably, slightly resistant bacterial strains are those against which the MIC of methicillin is >25 µg/mL.

These methods are useful for inhibiting bacterial growth in a variety of contexts. In certain preferred embodiments, the compound of the invention is administered to an experimental cell culture in vitro to prevent the growth of β-lactam resistant bacteria. In certain other preferred embodiments the compound of the invention is administered to a mammal, including a human, to prevent the growth of betalactam resistant bacteria in vivo. The method according to this embodiment of the invention comprises administering a therapeutically effective amount of a betalactamase inhibitor for a therapeutically effective period of timeto a mammal, including a human. Preferably, the betalactamase inhibitor is administered in the form of a pharmaceutical composition as described above. In some embodiments, a betalactam antibiotic is co-administered with the β-lactamase inhibitor as described above.

Assays for the inhibition of β-lactamase activity are well known in the art. For instance, the ability of a compound to inhibit betalactamase activity in a standard enzyme inhibition assay may be used (see, e.g., Page, Biochem J. 295:295-304 (1993)). β-Lactamases for use in such assays may be purified from bacterial sources or, preferably, are produced by recombinant DNA techniques, since genes and cDNA clones coding for many β-lactamases are known (see, e.g., Cartwright & Waley, Biochem J. 221 :505-12 (1984)).

Alternatively, the sensitivity of bacteria known, or engineered, to produce a β- lactamase to an inhibitor may be determined. Other bacterial inhibition assays include agar disk diffusion and agar dilution (see, e.g., Traub & Leonhard, Chemotherapy 43:159-67 (1997)). Thus, a β-lactamase can be inhibited by contacting the beta-lactamase enzyme with an effective amount of an inventive compound or by contacting bacteria that produce the β-lactamase enzymes with an effective amount of such a compound so that the β-lactamase in the bacteria is contacted with the inhibitor. The contacting may take place in vitro or in vivo. "Contacting" means that the betalactamase and the inhibitor are brought together so that the inhibitor can bind to the β-lactamase. Amounts of a compound effective to inhibit a betalactamase may be determined empirically, and making such determinations is within the skill in the art. Inhibition includes both reduction and elimination of β-lactamase activity.

EXAMPLES

The disclosure herein is further defined in the following Examples. It should be understood that these Examples, while indicating preferred embodiments, are given by way of illustration only. From the above discussion and these Examples, one skilled in the art can ascertain the preferred features, and without departing from the spirit and scope thereof, can make various changes and modifications to adapt it to various uses and conditions.

Synthesis

General

All reactions were performed under argon using oven-dried glassware and dry solvents. Dry tetrahydrofuran (THF) and diethyl ether were obtained by standard methods and freshly distilled under argon from sodium benzophenone ketyl prior to use. The -100 °C bath was prepared by addition of liquid nitrogen to a pre- cooled (-78 °C) mixture of ethanol/methanol ( 1 :1). Reactions were monitored by TLC, which were visualized by UV fluorescence and by Hanessian's cerium molybdate stain. Chromatographic purification of the compounds was performed on silica gel (particle size 0.05-0.20 mm). Melting points were measured on a Buchi 510 apparatus. Optical rotations were recorded at +20 °C on a Perkin- Elmer 241 polarimeter and are expressed in 10 deg cm2 g-1 . 1H- and 13C-NMR spectra were recorded on a Bruker DPX-200 or Avance-400 spectrometer; chemical shifts (δ) are reported in ppm downfield from TMS as internal standard (s singlet, d doublet, t triplet, q quartet, m multiplet, br broad signal); coupling constants (J) are given in Hz. Two-dimensional NMR techniques (COSY, HMBC, HSQC) were used to aid in the assignment of signals in 1H and 13C spectra. Mass spectra were determined on a gas chromatography HP 5890 associated with mass spectrometer detector HP 5972 (El, 70 eV) or on an Agilent Technologies LC-MS (n) Ion Trap 631 OA (ESI, 70 eV).

In Fig. 1 synthesis of exemplary compounds according to formula II is shown, comprising the following steps:

(+)-pinanediol a-bromomethaneboronate 1 was synthesized according to the literature (Matteson et al., Organometallics, 1996, 15(1), 152).

(+)-pinanediol azidomethaneboronate 2 : A solution of 1 (3g, 11 mmol) in EtOAc

(18 ml_) was stirred with a solution of sodium azide (7.15 g , 110 mmol) and tetrabutylammonium bromide (2.03 g, 5.5 mmol) in water under argon at rt for 18 hrs. The ethyl acetate phase was diluted with n-hexane (120 ml_), extacted with a saturated solution of NH CI (2x 80mL), dried over anhydrous magnesium sulphate and filtered. Concentration under reduced pressure afforded the title compound 2 as a pale yellow oil (2.44 g, 94.2%). [a] D+26.5 (c, 2.9, CH2CI2). 1 H NMR (400 MHz, CDCI3): 0.88 (3H, s, pinanyl C 3), 1.15 ( 1H, d, J = 11. 1 , pinanyl H n 0), 1.34 (3 H, s, pinanyl C 3), 1.47 (3H, s, pinanyl C 3), 1.92- 2.42 3 (5H, s, pinanyl protons), 3.13 (2H, s, B-CH2) , 4.49 ( 1H, d, J = 8.6, CHOB). C

NMR (100 MHz, CDCI3): 23.9, 26.5, 27.0, 28.5, 35.2, 35.9 (CB), 38.1 , 39.5, 5 1.1 , 78.6, 87.0. GC-MS: m/z 235 (M)+, 220, 206, 192, 179, 166, 150, 138, 125, 110, 93, 67, 55. (+)-pinanediol 1-chloro-2-azidoethaneboronate 3 : (Dichloromethyl)lithium was prepared by addition of n-butyllithium (3.3 m l of a 2.5 M solution) to dichloromethane (0.92 mL, 14.4 mmol) in anhydrous THF (30 mL) at -100 °C under argon. A solution of azidoboronic ester 2 ( 1 .69 g, 7.2 mmol) in anhydrous THF (4 mL) was added dropwise with vigorous stirring. When the temperature reached -90 °C, a solution of ZnC (12.8 mL of a 1 M solution) was added and the mixture was allowed to warm to rt overnight. Diethylether (150 mL) was added to the mixture and the organic phase was washed with saturated NH CI (50 m L x 3), dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum. The crude product was filtered through a small silica gel column using 5% diethyl ether in n-hexane to afford the title compound 3 as a pale yellow oil

( 1.91 g, 93.8%). [o +29.23 (c, 1.7, MeOH).

1 H NMR (400 MHz, CDCI 3) : 0.87 (3H, s, pynanyl C 3), 1.19 (1H, d , J = 11.1 , pinanyl H d ), 1.32 (3 H, s, pinanyl C 3) , 1.46 (3H, s, pinanyl C 3), 1.91- 2.44

(5H, m , pinanyl protons), 3.60-4.02 (3H, m , BC -C 2), 4.40 ( 1H, dd, J = 1.6, 10.2 3 CHOB). C NMR (100 MHz, CDCI 3): 23.9, 26.3, 27.0, 28.4, 35.1 , 38.2, 39.3, + 4 1.4 (CB), 5 1.1 , 54.6, 78.9, 87.4. GC-MS m/z 268 (M - CH3) , 255, 240, 220, 186, 173, 161 , 145, 134, 119, 107, 93, 83, 67, 55, 4 1.

(+)-pinanediol 1-bis-trimethylsilylamino-2-azidoethaneboronate 4 : lithium hexamethyldisilazane (8.0 mL of a 1 M solution in THF) was added dropwise to a solution of 3 (2.27 g, 8.0 mmol) in anhydrous THF (20 mL) at -100°C under argon. The mixture was allowed to warm to rt overnight. Petroleum Ether (500 mL) was added and the abundant precipitate was filtered on a MgS0 pad. The solution was concentrated under vacuum to afford the title compound 4 as a pale yellow oil

(2.25 g, 69%).[oc]D +4.2 (c, 1.6 , CDCI 3).

H NMR (400 MHz, CDCI 3): £0.19 (18 H , s, [Si(CH) 3]2), 0.87 (3H, s, pynanyl C 3),

1.14 ( 1H, d, J = 11.0, pinanyl H d ), 1.32 (3 H, s, pinanyl C 3) , 1.43 (3H, s, pinanyl C 3), 1.80- 2.42 (5H, m , pinanyl protons), 2.90 ( 1H, dd, J = 5.9, 8.4,

CH), 3.33 (1H, dd, J = 8.4, 12.3, BCH-CH 2), 3.42 ( 1H, dd, J = 5.9, 12.3, BCH- 3 CH2) , 4.35 ( 1H, dd, J = 1.7, 8.7 CHOB). C NMR (100 MHz, CDCI 3): 2.2, 2.7, 24.0, 26.3, 27.0, 28.3, 35.2, 38.2, 39.4, 43.4 (CB), 5 1.4, 56.1 , 78.6, 86.1 .

2-azido-1-(2-thiophen-2-yl-acetylamino)-ethaneboronic acid 5 A solution of dried methanol (0.28 mL, 7.1 mmol) in anhydrous THF (2mL) was added to a solution of 4 (2.89 mL, 7.1 mmol) in anhydrous THF (10 mL) at - 0 °C under argon flow. After being stirred for 10 min at -10 °C, the cooling bath was

removed. The reaction mixture was stirred for 1 hr at rt. Thereafter the reaction mixture was cooled again at -10 °C and a solution of 2-tiopheneacetylchloride (0.96 mL, 7.1 mmol) was slowly added. The resulting mixture was allowed to react for four hours. Ethyl acetate (100 mL) was added, the organic phase was washed with water (30 mL) and saturated NaHC0 3 and the aqueous phase riestracted with EtOAc (2 x 50 mL). The combined organic phases were concentrated under

vacuum to afford a brownish solid which was crystallized from CH2CI2 and n- hexane to givethe title compound 5 as a beige solid ( 1 .659, 60%), p.f. 139-140

°C. [a]D- 6 1.5 (c, 1.0 , CHCI3) . 1 H NMR (400 MHz, CDCI3): 0.89 (3H, s, pynanyl C 3), 1.31 (3 H, s, pinanyl

C 3), 1.38 ( 1H, d , J = 10.1 , pinanyl H n 0), 1.43 (3H, s, pinanyl C 3), 1.77- 2.59 (5H, m , pinanyl protons), 2.98 ( 1H, m, BCH), 3.45 ( 1 H, dd, J = 10.1 , 12.9, BCH-

C 2) , 3.65 ( 1H, dd, J = 3.7, 12.9, BCH-CH 2), 3.94 (2H, s , C 2CO), 4.29 ( 1H, dd, J

= 2.0, 8.6 CHOB), 6.70 ( 1H, b, NH), 7.01 ( 1H, d, J = 2.4, a ), 7.06 ( 1 H, dd, J = 3 3.5, 5.1, ar), 7.34 ( 1H, dd, J = 1. 1 , 5.1, Har) . C NMR (100 MHz, CDCI3): £24.1 , 26.5, 27.3, 28.9, 34.1, 36.2, 38.2, 39.9, 4 1.5 (CB), 51.9, 53.9, 77.2, 84.7, 126.4, 127.7, 128.2, 133.3, 174.4. ESI-MS m/ 4 11 (M + Na)+, 389 (MH) +. MSMS 389: m/z 332, 290, 237, 180.

General Procedure for Cu/C-catalyzed "click" reaction: Cu(0) 10 (54 mg, 3 wt % , ca 0.03 mmol) was added to a solution of 5 (100 mg,

0.26 mmol) and the proper alkyne ( 1 .1 eq, 0.28mmol) in dioxane (3 mL), in a sealed test tube, and argon was fluxed in. The reaction mixture was exposed to microwave irradiation under stirring at 150 °C for 30 min. The mixture was filtered through a pad of celite to remove the catalyst and washed with EtOAc. The solution was concentrated under vacuum and triturated with Et20 to give the desired triazole as a solid which was further purified as indicated in the examples below.

(+)-Pinanediol 2-(4-Phenyl-[1 ,2,3]triazol-1 -yl)-1 -(2-thiophen-2-yl-acetylamino)- ethaneboronate ester 6a. The title compound was prepared according to the general procedure. After

concentration, the residue (104 mg) was purified by crystallization from CH2CI2

and n-hexane to afford 6a as white solid (66 mg, 53 %), p.f. 15-1 17 °C. [a] D -

63.1 (c, 1.03, CDCI3).

H NMR (400 MHz, CDCI3): 0.92 (3H, s , pinanyl C 3) , 1.29 (3 H, s, pinanyl

do), C 3), 1.40 ( 1H, d, J = 11.7, pinanyl H e n 1.47 (3H, s, pinanyl C 3), 1.52- 2.57

(5H, m, pinanyl protons), 3.33 ( 1H, t, J = 6.4, BCH), 3.98 (2H, s, C 2CO), 4.32

(1H, dd, J = 2.2, 8.7, CHOB), 4.54 (2H, d, J = 6.4, BCH-C 2), 6.95-7.99 (2H, m ,

Har.tioph), tioph), 7.23 ( 1H , dd, J = 2.4, 4.1 , a 7.35-7.44 (3H, m, ar), 7.62-7.67 (2H,

Har), az), 13 δ m, 7.75 (1H, s, C tri 8.13 ( 1H, b, NH) . C NMR (100 MHz, CDCI3): 24.2, 26.8, 27.4, 29.2, 33.0, 38.2, 39.9, 43.0 (CB), 52.2, 54.0, 69.2, 76.9, 84.2, 120.3, 125.5, 126.1 , 127.5, 128.0, 128.2, 128.8, 130.2, 132.9, 147.5, 175.4. ESI- MS m/z 513 (M + Na)+, 491 (MH) +. MSMS 389: m/z 346, 339, 324, 194.

(+)-Pinanediol 2-[4-(3-carboxy-phenyl)-[1,2,3]triazol-1-yl]-1-(2-thiophen-2-yl- acetylamino)-ethaneboronate ester 6b The title compound was prepared according to the general procedure. After concentration, the residue (mg) was purified by chromatography (Si0 2, EtOAc/

EtPet 90:10) to afford 6c as white solid (90.6 mg, 66 %), p.f. 228 °C dec. [c ] D - 8 1.5 (c, 0.7 , MeOD). H NMR (400 MHz, CDCI3): 0.891 (3H, s, pinanyl C H 3) , 1.29 (3 H, s, pinanyl

C H 3) , 1.39 (3H, s, pinanyl C 3) , 1.44 ( 1H, d, J = 10.3, pinanyl H e ndo). 1 78- 2.43

(5H, m, pinanyl protons), 3.23 ( 1H, dd, J = 4.4, 10.2, BCH), 3.98 (2H, s, C H 2 CO),

4.24 ( 1H, dd, J = 1.6, 8.7, CHOB), 4.44 ( 1 H, dd, J = 10.2, 14.5, BCH-CH 2 ) , 4.60

( 1H , dd, J = 4.4, 14.5, BCH-CH 2 ) , 6.93-7.01 (2H, m, H a r tioph), 7.31 ( 1H, dd, J = 1.5,

5.0, a tioph), 7.56 (2H, t , J = 7.8, H a r) , 8.03 (2H, t, J = 8.2, H ar) , 8.43 ( 1H, s, CHtriaz), 8.49 ( 1H, b, NH) . ESI-MS m/z 557 (M + Na)\ 535 (MH)+. MSMS 535: m/z 383, 368, 346.

(+)-Pinanediol 2-[4-(3-amino-phenyl )-[ 1,2,3]triazol-1 -yl] -1-(2-thiophen-2-yI- acetylamino)-ethaneboronate ester 6c The title compound was prepared according to the general procedure. After concentration, the residue (mg) was purified by chromatography (SiO2, EtOAc/

EtPet 90:10) to afford 6c as white solid (96 mg, 74%), p.f. 87-90 °C. [a] D - 47.8 (c,

0.96 , CDCI3).

H NMR (400 MHz, CDCI 3 ) : 0.91 (3H, s , pynanyl CH3), 1.32 (3 H, s, pinanyl

C 3), 1.45 (3H, s, pinanyl C 3), 1.50 ( 1H , d , J = 10.3, pinanyl H e ndo), 1-84- 2.39

(5H, m, pinanyl protons), 3.29 (1H, d, J = 8.6, BCH), 3.50 (2H, b, N 2) , 3.96 (2H, s, C 2 CO), 4.30 (1H, d , J = 8.0, CHOB), 4.41-4.60 (2H, m, BCH-CH 2), 6.65 ( 1H, d , J = 7.8, Har), 7.35-7.44 (4H, m, Ha ), 7.14 ( 1H, t, J = 7.9, Ha ), 7.20 ( 1H, d, J = 4.9, Hartioph), 7.61 ( 1H, s, CHtriaz), 8.89 ( 1 H, b, NH) . 3C NMR (100 MHz, CDCI3):

£24.2, 26.8, 27.4, 29.2, 33.0, 36.7, 38.2, 40.1 , 43.4 (CB), 52.3, 52.6, 84.0, 112.2, 115.1 , 115.9, 120.2, 125.9, 127.3, 127.9, 129.7, 131 .0, 133.1 , 146.7, 147.6, 175.7. ESI-MS m/z 528 (M + Na , 506 (MH) +. MSMS 506: m/z 354, 346, 339.

(+)-Pinanediol 2-[4-(1 -Hydroxy-1 -methyl-ethyl)-[1 ,2,3]triazol-1 -yl]-1 -(2- thiophen-2-yl-acetylamino)-ethaneboronate ester 6d The title compound was prepared according to the general procedure but decreasing the temperature to 120 °C.

After concentration, the residue (121 mg) was purified by crystallization from Et20 and n-hexane to afford the title compound 6d as pale yellow solid (76 mg, 63%), p.f. 76-78 °C. [a] D - 54.9 (c,

1 H NMR (400 MHz, CDCI 3): 0.90 (3H, s, pinanyl C 3), 1.31 (3 H, s, pinanyl

C 3), 1.43 (3H, s, pinanyl endo), 1.43 (3H, s, C 3), 1.46 ( 1H, d , J = 10.3, C 3),

1.49 (3H, s, pinanyl C 3) , 1.81- 2.47 (5H, m , pinanyl protons), 3.19 ( 1H, d, J =

10.2, BCH), 3.95 (2H, s, C 2CO), 4.26 ( 1H, dd, J = 2.1 , 8.6, CHOB), 4.43 ( H, dd,

J = 3.4, 14.6, BCH-CH 2), 4.51 ( 1H , dd, J = 11.4, 14.6, BCH-CH 2), 6.93-6.97 (2H, m , Har.tioph), 7.22 ( 1 H, dd, J = 3.2, 6.5, a tioph), 7.66 ( 1 H, s , C triaz) 9.29 ( 1H, b, NAT) . 3C NMR (100 MHz, CDCI3): 24.2, 26.8, 27.4, 29.2, 29.5, 30.9, 32.6, 36.8, 38.0, 38.2, 40.1 , 44.3 (CB), 52.3, 52.8, 76.5, 83.7, 120.3, 125.8, 127.2, 127.7, 133.2, 155.5, 175.9.

(+)-Pinanediol 2-(4-carboxy-[1,2,3]triazol-1-yl)-1-(2-thiophen-2-yl- acetylamino)-ethaneboronate ester 6e The title compound was prepared according to the general procedure. After concentration the residue (125 mg) was crystallized from Et20 and n-hexane to afford the title compound 6e as a beige solid (70 mg, 60%), pf 121 °C dec. [a ] -

7 1.6 ( .3 , MeOD). H NMR (200 MHz, CDCI3) : 0.88 (3H, s, pinanyl C 3), 1.29 (3 H, s, pinanyl

C 3) , 1.37 (3H, s, pinanyl C 3) , 1.40 ( 1 H, d , J = 12.5, pinanyl endo),1 52- 2.47

(5H, m, pinanyl protons), 3.20 (1H, dd, J = 4.2, 9.9, BCH), 3.96 (2H, s, C 2CO),

4.21 (1H, d, = 2.2, 8.8, CHOB), 4.43 ( 1H, dd, J = 9.9, 14.4, BCH-CH 2) , 4.60 ( H, dd, = 4.2, 14.4, BCH-CH 2) , 6.92-7.80 (2H, m , a ,tioph) . 7.32 ( 1H, dd, J = 1.6, 4.9,

Hartioph), 8.79 ( 1H, s, CHtriaz). 3C NMR (50 MHz, CDCI3): 23.1, 26.2, 26.4, 28.3,

3 1.1 , 36.3, 37.7, 40.0, 40.4 (CB), 52.2, 52.4, 76.2, 83.2, 125.3, 126.6, 127.2, 128.7, 133.2, 177.2. ESI-MS m/z 471 (M + Na)\ 459 (MH)+. MSMS 459: m/z 346, 307, 212.

General Procedure for pinanediol deprotection

The pinanediol esters were dissolved in C H 3C N (3 mL) and HCI (3 equivalent of a

1M solution in degassed H20), phenylboronic acid ( 1 equivalent) and n-exane (3 mL) were sequentially added and the resulting biphasic solution was vigourously stirred. After 30 min the n-hexane solution (containing the pinanediol phenylboronate) was removed and n-hexane (3 mL) was added. This last procedure was repeated several times until a TLC analysis revealed no more presence of 6a-e. The acetonitrile phase was concentrated affording the desired compounds 7a-e.

2-(4-Phenyl-[1 ,2,3]triazol-1 -yl)-1 -(2-thiophen-2-yl-acetylamino)-ethaneboronic acid 7a

Compound 7 a was obtained in 85% yield as white solid (mp 159 °C dec), [α] = - 96.5 (c 1.07%; MeOH), de > 98%. H NMR (400 MHz, MeOD): H NMR (400 MHz, MeOD): 3.26 ( 1 H, d , J = 8.5,

BCH), 4.02 (2H, s, C 2CO), 4.33-4.65 (2H, m , BCH-CH 2), 6.96-7.03 (2H, m , Har tioph), 7.33 ( 1 H, d, J = 5.0, a tioPh), 7.38 (1H, d , J = 7.2), 7.45 (1H, t, J = 7.4, H„),

13 7.82 ( 1H, s, Har), 7.84 ( 1H, s , Har), 8.40 (1H, s, C ria ). C NMR (100 MHz, MeOD): £30.7, 45.7, 52.5, 121 .6, 125.4, 125.4, 126.7, 127.4, 128.1 , 128.6, 129.9, 133.2, 147.2, 177.7 2-[4-(3-carboxy-phenyl)-[1 ,2,3]triazol-1 -yl]-1 -(2-thiophen-2-y l-acetylamino)- ethane boronic acid 7b Compound 7b was obtained in 74% yield using MeOH in place of CH3CN in the transesterification process, as white solid, [ ] = -71 .4 (c, 0.94, MeOD), de ≥ 98%.

7b

1 H NMR (400 MHz, MeOD): £ 3.32 ( 1H, d, J = 1.4, BCH), 4.05 (2H, s, C 2CO),

4.48-4.66 (2H, m, BCH-CH 2), 6.99 (1 H, dd, J = 3.5, 5.0, a tioph), 7.05 ( 1 , d, J =

ioph), 3.5, a r t 7.35 ( 1H, d , J = 5.0, a tioPh), 7.64 ( 1H, t, J = 7.6, a ) , 8.08 ( 1 H, d, J 3 = 10.9, Ha ), 8.10 ( 1H, d, J = 8.2, Ha ), 8.51 ( H, s, C tria ) , 8.69 ( 1H, s, Har) . C NMR (100 MHz, MeOD): £ 30.8, 45.3, 53.5, 123.3, 125.5, 126.7, 126.8, 127.5, 128.5, 129.2, 129.9, 130.0, 131 .7, 133.1 , 145.1 , 167.5, 177.9. ESI-MS m/z 423 (M + Na)+, 401 (MH)+, 387, 373, 364. 2-[4-(3-amino-phenyl)-[1 ,2,3]triazol-1 -yl]-1 -(2-thiophen-2-yl-acetylamino)- ethane boronic acid 7c Compound 7c was obtained in 66% yield as white solid (mp 215°C dec), [ ] = - 77.3 (c 0.73, MeOD), de > 98%.

7c

H NMR (400 MHz, MeOD): 3.24 ( 1H, d , J = 8.5, BCH), 4.02 (2H, s, C 2CO),

tio ) , 4.37-4.54 (2H, m, BCH-CH 2), 6.92-7.80 (2H, , a r 6.86 (1H, d , J = 5.1 , Ha ),

z). 13 6.98-7.03 (2H, m , Har), 7.31 (4H, , Har), 8.3 ( 1H , s, H a C NMR (100 MHz, MeOD): δ 30.6, 44.5 (br, C ), 52.1 , 113.5, 116.5, 117.3, 121 .3, 125.4, 126.7,

129.5, 13 1.4, 133.2, 147.5, 176.8. ESI-MS m/z 472 (MH)+. MSMS 472: m/z 354, 346, 339.

2-[4-(1 -Hydroxy-1 -methyl-ethyl)-[1 ,2,3]triazol-1 -yl]-1 -(2-thiophen-2-yl- acetylamino)-ethaneboronic acid 7d Compound 7d was obtained in 64.8% yield as white solid (mp 107-1 10°C dec),

[a] D = -68.5 (c 0.98, MeOD), de > 98%.

1 H NMR (400 MHz, MeOD): 1.64 (3H, s, CC 3) , 1.65 (3H, s, CC 3), 3.23 (1H, s,

tioph), BCH), 4.04 (2H, s, C 2CO), 4.37-4.70 (2H, m, BCH-CH2), 7.01 (2H, m, Ha r ph), 7.36 ( 1H, d, J = 5.0, Ha r ti0 8.56 ( 1H, s, Ht az). 2-(4-carboxy-[1 ,2,3]triazol-1 -yl)-1 -(2-thiophen-2-yl-acetylamino)- ethaneboronic acid 7e

Compound 7e was obtained in 88% yield as beige solid, [O ] D = -85.0 (c 0.92%; MeOD), de > 98%.

H NMR (400 MHz, MeOD): H NMR (400 MHz, MeOD): 3.22 ( 1H, dd, J = 3.9,

C ) , 10.2, CH), 4.02 (2H, s, 2CO), 4.43 (1H, dd, J = 10.2, 14.4, BCH-CH 2 4.55

( 1H , dd, = 3.9, 14.4, BCH-CH 2 ) , 6.98-7.04 (2H, m, H a r tioPh), 7.35 ( 1H, dd, J = 1. 1 ,

5.1 , Hartioph), 8.54 ( 1H , s, CHtriaz). 3C NMR (100 MHz, CDCI3): £30.6, 45.8, 52.3, 125.4, 126.7, 127.4, 128.7, 133.2, 161 .9, 177.7 ESI-MS m/z (M + Na)+, (MH)+. ESI-MS: m/z 307 [(M-18)+1] +. MSMS 307: m/z 292, 264, 212, 194, 168, 140. Fig. 2, 3 and 4 do show the synthesis of further compounds according to the invention, comprising further embodiments of the heterocylce Z.

Experimental methods for /^-lactamase Enzyme Assays

/^-Lactamase expression and purification

The a sH -i coding sequence and natural promoter were cloned into a pBC SK (-) vector and transformed into E. coli DH10B cells. 10 ml of an overnight culture grown in lysogeny broth (LB) were used to inoculate 500 ml of super optimal broth (SOB) and grown overnight. Cells were harvested by centrifugation at 4 °C and frozen at -20 °C. After thawing, β-lactamase was liberated using stringent periplasmic fractionation with 0.04 mg/mL (Sigma) and 1 mM EDTA, pH 7.8. Preparative isoelectric focusing was performed with the lysate in a Sephadex G-100 (GE Healthcare) using ampholines in the pH range 3.5-10 (Bio-Rad) and running the gel overnight at a constant power of 8 W on a Multiphor II isoelectric focusing apparatus (GE Healthcare). Purity was assessed by 5% stacking, 12% resolving sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS- PAGE). β-Lactamase concentration was determined using Bio-Rad Bradford protein assay with bovine serum albumin standards.

The a pDc-3 coding sequence (minus the leader sequence) was cloned into a pET24a(+) vector and transformed into E. coli BL21(DE3) RP CodonPlus cells. 12 ml of an overnight culture grown in LB were used to inoculate 500 ml of SOB and grown at 37 °C for 2 hr. Isopropyl β-D-thiogalactopyranoside (IPTG) was added to a final concentration of 1 M, and the cells grown for an additional 2.5 hr. The cells were harvested, frozen, resuspended in 50 mM Tris (pH 7.4), and β- lactamase liberated with lysozyme. PDC-3 enzyme was purified by preparative isoelectric focusing (PIEF) followed by fast protein liquid chromatography (FPLC) with a Sephadex Hi Load 16/60 column and a HiTrap High Performance sulfopropyl strong cation exchanger (GE Healthcare). The protein was quantified by the Bradford protein assay and purity was assessed by 5% stacking, 12% resolving SDS-PAGE.

The aTE -i coding sequence (minus the leader sequence) was cloned into a pET24a(+) vector and transformed into E. coli BL21(DE3) cells. 10 ml of an overnight culture grown in LB were used to inoculate 500 ml of SOB and grown at

37 °C to OD600 = 0.6. IPTG was added to a final concentration of 0.2 mM, and the cells grown for an additional 3 hr. The cells were harvested, frozen, resuspended in 50 mM Tris (pH 7.4), and β-lactamase liberated with lysozyme. TEM-1 enzyme was purified by PIEF and FPLC with a Sephadex Hi Load 16/60 column. The protein was quantified by the Bradford protein assay and purity was assessed by 5% stacking, 12% resolving SDS-PAGE. b la - The bla -2 coding sequence (minus the leader sequence) was cloned into a pET24a(+) vector and transformed into Origami™2 DE3 pLys cells (Novagen). 10 ml of an overnight culture grown in lysogeny broth were used to inoculate 500 ml of SOB and grown at 37 °C to O.D.60o = 0.6. IPTG was added to a final concentration of 0.2 mM, and the cells grown for an additional 3 hr. The cells were harvested, frozen, resuspended in 50 mM Tris (pH 7.4), and β-lactamase liberated with lysozyme. KPC-2 enzyme was purified by PIEF followed by FPLC with a Sephadex Hi Load 16/60 column. The protein was quantified by the Bradford protein assay and purity was assessed by 5% stacking, 12% resolving SDS- PAGE.

β-lactamase inhibition The level of inhibition of β-lactamase enzymes was determined at room temperature using an Agilent 8453 diode array spectrophotometer (Agilent, Palo Alto, CA). Initially, each assay was performed using 20 µΜ solution of inhibitor in 10 mM phosphate-buffered saline (PBS) pH 7.4. The enzyme (3 nM) and the inhibitor were preincubated for 15 min. Measurements were obtained using the ∆Ε - 1 indicator substrate nitrocefin (NCF, 482 = 17,400 M cm ). Inhibition is evaluated as % residual velocities and reported in the following table:

For compounds 7d and 7e furthur kinetic experiments for the β-lactamases KPC-2 and PDC-3 were performed in order to obtain the inhibition constant ( j). In the experiments to determine K\, each inhibitor was regarded as a "competitive inhibitor". Therefore, K , values were determined in a direct competition assay with NCF. In each case, initial velocities, v, were measured using a constant concentration of enzyme [£] and increasing concentrations of inhibitor [/] against the indicator substrate NCF [S]. After the , was obtained, the data were "corrected" to account for the affinity of NCF for each enzyme used. Table 2: alues for compounds 7d and 7e against KPC-2 and PDC-3

* 5min pre-incubation

In vitro Antibacterial Assays To determine the ability of the compounds to inhibit the growth of bacterial strains when partnered with a β-lactam antibiotic, simple disc assays were performed. Ampicillin discs containing 10 pg of ampicillin and ampicillin-sulbactam discs containing 10 pg of ampicillin and 10 pg of sulbactam were purchased from Becton Dickinson. A solution containing 0 g of each inhibitor was pipetted onto ampicillin disks containing 10 pg of ampicillin and allowed to dry for 1 hr. Colonies, directly re-suspended into sterile water equivalent to a 0.5 McFarland standard, were used to inoculate Mueller Hinton (MH) agar plates. The discs were carefully placed on each plate. The bacteria were grown at 37 °C for 18 hours and zone diameters were measured.

Table 3: Disc Assay (10 g Ampicillin, 10 g inhibitor) Zone size (mm) Claims

A compound, particularly a boronic acid containing at least one heterocyle, having the general formula (I)

wherein

L is a bond or a C1-C8 saturated or unsaturated linear or branched aliphatic chain, optionally substituted with one or more groups chosen from hydroxyl, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, amino, aminocarbonyl, carbonyl, aminosulfonyl, alkylaryl, aryl, aryloxy, carboxyl, cyano, guanidine, halogen, heteroaryl, heterocyclyl, sulfide, sulfonyl, sulfoxido, sulfonic acid, sulfate and thiol;

R is chosen from hydrogen, linear or branched C1-C12 alkyl, linear or branched C1-C12 alkenyl, or C3-C8 cycloalkyl; said linear or branched C1-C12 alkyl, linear or branched C1-C12 alkenyl, or C3- C8 cycloalkyl being optionally substituted with one or more groups independently chosen from the group consisting of hydroxyl,

halogen, carboxyl, cyano, C(0)R , C(0)NR R5 thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido; is 0, 1, 2 , 3, 4, 5, or 6; and X 2 are independently hydroxy!, halogen, NR1R2, C1-C6 alkoxy, or acyloxy; or X and X 2 together form a cyclic boron ester, a cyclic boron amide, or a cyclic boron amide-ester, said cyclic boron ester, boron amide or boron amide-ester having a chain or a ring containing from 2 to 12 carbon atoms and, optionally, 1-3 heteroatoms which can be O, N, or S;

is chosen from hydrogen, optionally substituted linear or branched C1-C12 alkyl, optionally substituted linear or branched C1-C12 alkenyl, NR1R2, OR3, SR3, or from the group consisting of aryl group substituted with from 0 to 3 substituents, heteroaryl group substituted with from 0 to 3 substituents and heterocyclic group substituted with from 0 to 3 substituents, said substituents being independently selected from the group consisting of hydroxyl,

halogen, carboxyl, cyano, C(O)R , C(0)NR R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido;

, R2, R3, R4, R 5 are independently selected from the group consisting of: (a) hydrogen; (b) C1-C10 alkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents; (c) C3-C10 cycloalkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents; (d) heteroaryl group substituted with 0 , 1, 2 or 3 substituents; (e) heterocyclic group substituted with 0, 1, 2 or 3 substituents; (f) C1-C12 alkyl, C1-C12 cycloalkyl, C1-C12 alkenyl; (g) aryl group substituted with 0, 1, 2 , 3 , 4 or 5 substituents;

(h) C(0)R 6; -C(0)NR 6R7; -C(0)OR 6, C(=NR6); NR6R7, -C(=NR6)R7; (i) C1-C6 alkyl any carbon of which can be substituted with 0 , 1, 2 or 3 substituents; (j) C3-C7 cycloalkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents;

said substituents of Ri, R2, R3, R and R 5 being independently selected from the group consisting of hydroxyl, halogen, carboxyl,

cyano, C(O)R6, C(0)NR R7, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C10 carbons of (b), any of the carbons of the cycloalkyl group of (c) other than the one attached to the rest of the molecule, any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule, any of the C1-C6 carbons of (i) or any of the carbons of the cycloalkyl group of (j) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, sulfoxido; and R and R7 being independently selected from hydrogen, or from the group consisting of (a) C1-C6 alkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents, (b) C3-C7 cycloalkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents, (c) aryl group substituted with 0, 1, 2 or 3 substituents (d) heteroaryl group substituted with 0, , 2 or 3 substituents and (e) heterocyclic group substituted with 0, 1, 2 or 3 substituents, the substituents of and R7 being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C6 carbons of (a), any of the carbons of the cycloalkyl group of (b) other than the one attached to the rest of the molecule, or any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, and sulfoxido or an oxyimino group as indicated above; and

is an heterocyclic or heteroaromatic ring system optionally substituted with 1 or more substituents selected from the group consisting of hydroxyl, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, amino, aminocarbonyl, carbonyl, aminosulfonyl, alkylaryl, aryl, aryloxy, carboxyl, cyano, guanidine, halogen, heteroaryl, heterocyclyl, sulfide, sulfonyl, sulfoxido, sulfonic acid, sulfate and thiol.

The compound according to claim 1, wherein the heterocyclic or heteroaromatic ring system Z is a stable 3- to 8-membered monocyclic or stable 7- to 14-membered bicyclic heterocyclic ring which is either saturated or unsaturated, and which consists of carbon atoms and from 1 to 4 heteroatoms selected from the group consisting of N, O, and S, preferably N .

The compound according to claim 1 or claim 2, wherein the heterocyclic or heteroaromatic ring system Z is a bicyclic group in which a heterocyclic ring is fused to a benzene ring. 4. The compound according to any of claims 1 to 3, wherein the heterocyclic or heteroaromatic ring system Z is selected from the group comprising azetidinyl, oxetanyl, oxazolyl, pirazolyl, thiazolyl, triazolyl, tetrazolyl, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, 1,3-dioxolanyl, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, 2- oxopiperazinyl, 2-oxopiperdinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, 2-pyridinonyl, pyrazinyl, pyrazolidinyl.pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, thienyl and triazolyl.

5. The compound according to any of claims 1 to 4, wherein the heterocyclic or heteroaromatic ring system Z is a heteroaryl group, preferably a group having 5 to 14 ring atoms, more preferably a group having 5, 6, 9, or 10 ring atoms; said heteroaryl group preferably having 6 , 0, or 14 π electrons shared in a cyclic array; and having 1, 2 or 3 heteroatoms independently selected from the group consisting of N, O, and S, even more preferably the heterocyclic or heteroaromatic ring system of Z is a heteroaryl group selected from the group comprising thienyl, benzothienyl, furyl, benzofuryl, dibenzofuryl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, indolyl, quinolyl, isoquinolyl, quinoxalinyl, tetrazolyl, oxazolyl, thiazolyl, isoxazolyl and triazolyl.

6. The compound according to any of claims 1 to 5, wherein the heterocyclic or heteroaromatic ring system Z is fused to an aryl or heteroaryl group, preferably the heterocyclic or heteroaromatic ring system Z is selected from the group comprising tetrahydroquinolinyl, dihydrobenzofuranyl, benzoxazinyl, 1,2,3,4-tetrahydro-quinoxalinyl, benzoimidazolyl, benzothiazolyl, benzotriazolyl.

7. The compound according to any of claims 1 to 6, wherein the heterocyclic or heteroaromatic ring system Z is substituted with 0, 1, 2 or 3 substituents selected from the group consisting of hydroxyl, halogen, carboxyl, cyano,

R7, C(0)R 6, C(0)NR 6 thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido, wherein

Re and R7 are independently selected from hydrogen, or from the group consisting of (a) C1-C6 alkyl any carbon of which can be substituted with 0, 1, 2 or 3 substituents, (b) C3-C7 cycloalkyl any carbon of which can be substituted with 0 , 1, 2 or 3 substituents, (c) aryl group substituted with 0, , 2 or 3 substituents (d) heteroaryl group substituted with 0, 1, 2 or 3 substituents and (e) heterocyclic group substituted with 0, 1, 2 or 3 substituents,

the substituents of R 6 and R7 being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C6 carbons of (a), any of the carbons of the cycloalkyl group of (b) other than the one attached to the rest of the molecule, or any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, and sulfoxido.

The compound according to any of claims 1 to 7, wherein L is a bond or a C1-C8 saturated or unsaturated linear or branched aliphatic chain,

preferably a bond, (C ) or (CH2 )2, more preferably (C ).

The compound according to any of claims 1 to 8 wherein Z is an triazol optionally substituted with 1 or more substituents selected from the group consisting of hydroxyl, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, amino, aminocarbonyl, carbonyl, aminosulfonyl, alkylaryl, aryl, aryloxy, carboxyl, cyano, guanidine, halogen, heteroaryl, heterocyclyl, sulfide, sulfonyl, sulfoxido, sulfonic acid, sulfate and thiol, preferably from

The compound according to any of claims 1 to 9, wherein Y is a heteroaryl group substituted with from 0 to 3 substituents, said substituents being independently selected from the group consisting of hydroxyl, halogen,

carboxyl, cyano, C(0)R , C(0)NR 4R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido; preferably Y is selected from the group comprising thienyl, benzothienyl, furyl, benzofuryl, dibenzofuryl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, indolyl, quinolyl, isoquinolyl, quinoxalinyl, tetrazolyl, oxazolyl, thiazolyl, isoxazolyl and triazolyl.

1.The Compound according to any of claims 1 to 10 characterized by having the general formula (II), (V), (VI) or (VII)

wherein Y, m, R, X and X 2 are defined as above and

Re, R9, Rio, 11 and R12 are independently selected from the group consisting of (a) hydrogen; (b) C1-C6 alkyl any carbon of which can be substituted with from 0 to 3 substituents, (c) C3-C7 cycloalkyl any carbon of which can be substituted with from 0 to 3 substituents, (d) aryl group substituted with from 0 to 3 substituents (e) heteroaryl group substituted with from 0 to 3 substituents and (f) heterocyclic group substituted with from 0 to 3 substituents,

the substituents of s and R g being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, oxyimino group wherein any of the C1-C6 carbons of (a), any of the carbons of the cycloalkyl group of (b) other than the one attached to the rest of the molecule, or any of the carbons of the heterocyclic group of (e) other than the one attached to the rest of the molecule comprise part of said oxyimino group, sulfido, and sulfoxido;

most preferably Rs, Rg, Rio, Rn and R12 are selected independently from either hydrogen or a compound according to one of the following formulas a., b., c , d., or e.:

12. Compound according to any of claims 1 to 11, wherein the compound is

, or

, or a salt thereof. 13. Compound according to any of claims 1 to 12, characterized by the fact

that it is an inhibitor for β-lactamase, preferably for a class A , C or D β- lactamase, even more preferably for a β-lactamase selected from SHV-1 , TEM-1 , PDC-3 or KPC-2.

14. Compound according to any of claims 1 to 13 for use in the prevention or treatment of bacterial infections, preferably for the treatment of an bacterial infection caused by bacteria of the genus Staphylococcus, preferably Staphylococcus aureus or Staphylococcus epidermidis, Streptococcus, preferably Streptococcus agalactiae, Streptococcus pneumoniae or Streptococcus faecalis, Micrococcus, preferably Micrococcus luteus, Bacillus, preferably Bacillus subtilis, Listerella, preferably Listerella monocytogene, Escherichia, preferably Escherichia coli, Klebsiella, preferably Klebsiella pneumoniae, Proteus, preferably Proteus mirabilis or Proteus vulgaris, Salmonella, preferably Salmonella typhosa, Shigella, preferably Shigella sonnef, Enterobacter, preferably Enterobacter aerogenes or Enterobacter cloacae, Serratia, preferably Serratia marcescens, Pseudomonas, preferably Pseudomonas aeruginosa, Acinetobacter, preferably Acinetobacter baumanii, Nocardia, preferably Nocardia autotrophica, or Mycobacterium, preferably Mycobacterium fortuitum.

15. Method for the preparation of a compound according to any of claims 1 to 13 wherein the heterocyclic or heteroaromatic ring system Z is synthesized by click chemistry, preferably by cycloaddition, more preferably by 1,3- dipolar cycloaddition, 2,4 cycloaddition or 2+2-cycloaddition, even more preferably by Huisgen 1,3-dipolar cycloaddition or strained-cycloaddtion. 16. Method according to claim 13, characterized by comprising the steps a. providing an alkylboronate, preferably providing an azido- alkylboronate, more preferably a compound of the formula (III)

(III)

wherein Y, R, m, L, Χι, X 2 are defined as above, wherein Χ , X 2 are preferably a protectiongroup, more preferably X 1, X2 are (+)- pinanediol;

b. sythesis of Z by cycloaddtion, preferably by Cu/C-catalyzed click reaction, even more preferably by Huisgen 1,3-dipolar cycloaddtion, providing a compound of the formula IV

wherein Y , m, R, L, Xi, X2, R8, R9 are defined in step a. optionally purification of the product; optionally deprotection of the protected boronate-group.

17. Method according to any of claims 14 or 15, characterized by the fact that

the click reaction is a Cu/C-catalyzed click reaction comprising at least the steps

a. providing a solution of an azido-alkylboronoate and an alkyne in dioxane;

b. adding Cu(0)/C to the provided solution of step a);

c. optionally providing inert gas-influx, preferably Argon-influx, into sealed reaction vessel; . optionally exposure of the reaction mixture to irradiation, preferably microwave irradiation under stirring at 150°C.

18. Compound according to one of the formulas (III), (IV), (VIII), (IX),or (X)

or a salt thereof, preferably according to one of the formulas (III) or (IV), wherein Y is chosen from hydrogen or from the group consisting of aryl group substituted with from 0 to 3 substituents, heteroaryl group substituted with from 0 to 3 substituents and heterocyclic group substituted with from 0 to 3 substituents, said substituents being independently selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(0)R ,

C(0)NR R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxid; is a heteroaryl group substituted with from 0 to 3 substituents, said substituents being independently selected from the group consisting of

hydroxyl, halogen, carboxyl, cyano, C(0)R , C(O)NR4R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido; preferably Y is selected from the group comprising thienyl, benzothienyl, furyl, benzofuryl, dibenzofuryl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, indolyl, quinolyl, isoquinolyl, quinoxalinyl, tetrazolyl, oxazolyl, thiazolyl,

isoxazolyl and triazolyl; wherein R and R5 are as defined above; m is 0, 1, ,2, 3, 4, 5 or 6 , preferably is 0, 1, 2, more preferably m is 1;

X and X2 are independently hydroxyl, halogen, NR^, C1-C6 alkoxy, or

acyloxy; or X and X2 together form a cyclic boron ester, a cyclic boron amide, or a cyclic boron amide-ester, said cyclic boron ester, boron amide or boron amide-ester having a chain or a ring containing from 2 to 12 carbon atoms and, optionally,

1-3 heteroatoms which can be O, N , or S; wherein Ri, R2 are

as defined above; preferably X and X2 are protection groups or do form together one protection group, wherein preferably the protection group is pinanediol;

L is a bond or a C1-C8 saturated or unsaturated linear or branched aliphatic chain, optionally substituted with one or more groups chosen from hydroxyl, alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, amino, aminocarbonyl, carbonyl, aminosulfonyl, alkylaryl, aryl, aryloxy, carboxyl, cyano, guanidine, halogen, heteroaryl, heterocyclyl, sulfide, sulfonyl,

sulfoxido, sulfonic acid, sulfate and thiol;

R is chosen from hydrogen, linear or branched C1-C12 alkyl, linear or branched C1-C12 alkenyl, or C3-C8 cycloalkyi; said linear or branched C1-C12 alkyl, linear or branched C1-C12 alkenyl, or C3-C8 cycloalkyi being optionally substituted with one or more groups independently chosen from the group consisting of hydroxyl, halogen, carboxyl, cyano, C(O)R ,

C(0)NR R5, thiol, sulfonic acid, sulfate, alkyl, cycloalkyi, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl,

guanidino, sulfido, and sulfoxido; wherein R4 and R5 are defined as above; and wherein

Re, R9 are independently selected from the group consisting of (a) hydrogen; (b) C1-C6 alkyl any carbon of which can be substituted with from 0 to 3 substituents, wherein optionally at least one

substituent is an oxyminio group and wherein any of the C1-C6 carbons comprise part of said oxyimino group, (c) C3-C7 cycloalkyi any carbon of which can be substituted with from 0 to 3 substituents, wherein optionally at least

one substituent is an oxyminio group and wherein any of the carbons of the cycloalkyi group other than the one attached to the rest of the molecule comprise part of said oxyimino group, (d) aryl group substituted with from 0 to 3 substituents (e) heteroaryl group substituted with from 0 to 3 substituents and

(f) heterocyclic group substituted with from 0 to 3 substituents, wherein optionally at least one substituent is an oxyminio group and wherein any of the carbons of the cycloalkyl group other than the one attached to the rest of the molecule comprise part of said oxyimino group, the substituents being selected from the group consisting of hydroxyl, halogen, carboxyl, cyano, thiol, sulfonic acid, sulfate, optionally substituted: alkyl, cycloalkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, alkylaryl, heteroarylalkyl, alkylheteroaryl, cycloalkoxy, heterocyclyloxy, aryloxy, heteroaryloxy, amino, carbonyl, aminocarbonyl, oxycarbonyl, aminosulfonyl, sulfonyl, guanidino, sulfido, and sulfoxido; most preferably R8 and R9 are selected independently from either hydrogen or a compound according to one of the formulas a., b., c , d., or e.:

19. Compound according to claim 18, characterized by the fact that it is an inhibitor for β-lactamase, preferably for a class A , C or D β-lactamase, even more preferably for a β-lactamase selected from SHV-1 , TEM-1 , PDC-3 or KPC-2.

20. Compound according to any of claims 18 or 19 for use in preparation of a compound according to any of claims 1 to 14. 2 1.Compound according to any of claims 18 to 20 for use in a method according to any of claims 15 to 17.

22. Compound according to any of claims 18 to 2 1, for use in manufacturing a medicament for the prevention or treatment of bacterial infections, preferably for the treatment of an bacterial infection caused by bacteria of the genus Staphylococcus, preferably Staphylococcus aureus or Staphylococcus epidermidis, Streptococcus, preferably Streptococcus agalactiae, Streptococcus pneumoniae or Streptococcus faecalis, Micrococcus, preferably Micrococcus luteus, Bacillus, preferably Bacillus subtilis, Listerella, preferably Listerella monocytogene, Escherichia, preferably Escherichia coli, Klebsiella, preferably Klebsiella pneumoniae, Proteus, preferably Proteus mirabilis or Proteus vulgaris, Salmonella, preferably Salmonella typhosa, Shigella, preferably Shigella sonnef, Enterobacter, preferably Enterobacter aerogenes or Enterobacter cloacae, Serratia, preferably Serratia marcescens, Pseudomonas, preferably Pseudomonas aeruginosa, Acinetobacter, preferably Acinetobacter baumanii, Nocardia, preferably Nocardia autotrophica, or Mycobacterium, preferably Mycobacterium fortuitum.

23. Pharmaceutical composition comprising a compound according to any of claims 1 to 14 or a salt thereof.

24. Pharmaceutical composition comprising a compound according to any of claims 18 to 22 or a salt thereof.

25. Pharmaceutical composition according to any of claims 23 or 24, characterized by further comprising a β-lactam antibiotic, preferably a β- lactam antibiotic selected from the group comprising penicillins, cephalosporins, penems, carbapenems, and monobactams. 26. Pharmaceutical composition according to any of claims 23 to 25, characterized by further comprising a pharmaceutically acceptable excipient.

27. Pharmaceutical composition according to any of claims 23 to 26 for use in the prevention or treatment of bacterial infection caused by gram-positive or gram-negative bacteria, preferably for the treatment of an infection caused by bacteria of the genus Staphylococcus, preferably Staphylococcus aureus or Staphylococcus epidermidis, Streptococcus, preferably Streptococcus agalactiae, Streptococcus pneumoniae or Streptococcus faecalis, Micrococcus, preferably Micrococcus luteus, Bacillus, preferably Bacillus subtilis, Listerella, preferably Listerella monocytogene, Escherichia, preferably Escherichia coli, Klebsiella, preferably Klebsiella pneumoniae, Proteus, preferably Proteus mirabilis or Proteus vulgaris, Salmonella, preferably Salmonella typhosa, Shigella, preferably Shigella sonnef, Enterobacter, preferably Enterobacter aerogenes or Enterobacter cloacae, Serratia, preferably Serratia marcescens, Pseudomonas, preferably Pseudomonas aeruginosa, Acinetobacter, preferably Acinetobacter baumanii, Nocardia, preferably Nocardia autotrophica, or Mycobacterium, preferably Mycobacterium fortuitum.

28. Method for determining β-lactamase activity in a bacterial strain comprising the steps a. cultivation of a first colonie of the bacterial strain in a medium containing an β-lactam antibiotic; b. cultivation of a second colony of the bacterial strain in a medium containing an β-lactam antibiotic and a compound according to one of claims 1 to 14 or a compound according to one of claims 18 to 22; c. comparing the growth rate of the first and the second colony.

A . CLASSIFICATION O F SUBJECT MATTER INV. C07F5/02 A61K31/69 A61P31/04 ADD.

According to International Patent Classification (IPC) o r t o both national classification and IPC

B . FIELDS SEARCHED Minimum documentation searched (classification system followed by classification symbols) C07F A61K A61P

Documentation searched other than minimum documentation to the extent that such documents are included in the fields searched

Electronic data base consulted during the international search (name of data base and, where practicable, search terms used)

EPO-Internal , CHEM ABS Data, WPI Data

C . DOCUMENTS CONSIDERED T O B E RELEVANT

Category* Citation of document, with indication, where appropriate, of the relevant passages Relevant to claim No.

US 7 271 186 Bl (SHOICHET BRIAN K [US] ET 1-28 AL) 18 September 2007 (2007-09-18) ci ted i n the appl i cati on col umn 3 , l i ne 20 - col umn 3 , l i ne 25 ; tabl es 1, 3

FR 2 758 329 Al (SYNTHELABO [FR] ) 1, 2 ,4,5 , 17 July 1998 (1998-07-17) 7,8, 10, 26 page 27 , l i ne 1 - page 28, l i ne 1; compounds 4,6-8

US 5 731 439 A (CARINI DAVID JOHN [US] ET 1, 2 ,4,7 , AL) 24 March 1998 (1998-03-24) 8,26 col umn 16, compond 18; col umn 1, l i ne 5 - col umn 1, l i ne 10

□ Further documents are listed in the continuation of Box C . See patent family annex.

* Special categories of cited documents : "T" later document published after the international filing date o r priority date and not in conflict with the application but cited to understand "A" document defining the general state of the art which is not considered the principle o r theory underlying the invention to be of particular relevance "E" earlier application o r patent but published o n o r after the international "X" document of particular relevance; the claimed invention cannot be filing date considered novel o r cannot b e considered to involve a n inventive "L" documentwhich may throw doubts o n priority claim(s) orwhich is step when the document is taken alone cited to establish the publication date of another citation o r other "Y" document of particular relevance; the claimed invention cannot be special reason (as specified) considered to involve a n inventive step when the document is "O" document referring to a n oral disclosure, use, exhibition o r other combined with one o r more other such documents, such combination means being obvious to a person skilled in the art "P" document published prior to the international filing date but later than the priority date claimed "&" document member of the same patent family

Date of the actual completion of the international search Date of mailing of the international search report

29 May 2012 23/01/2013

Name and mailing address of the ISA/ Authorized officer European Patent Office, P.B. 5818 Patentlaan 2 N L - 2280 HV Rijswijk Tel. (+31-70) 340-2040, Fax: (+31-70) 340-3016 Schmi d , Arnol d INTERNATIONAL SEARCH REPORT

Box No. II Observations where certain claims were found unsearchable (Continuation of item 2 of first sheet)

This international search report has not been established in respect of certain claims under Article 17(2)(a) for the following reasons:

□ Claims Nos.: because they relate to subject matter not required to be searched by this Authority, namely:

□ Claims Nos.: because they relate to parts of the international application that do not comply with the prescribed requirements to such an extent that no meaningful international search can be carried out, specifically:

3 . □I I Claims Nos.: because they are dependent claims and are not drafted in accordance with the second and third sentences of Rule 6.4(a).

Box No. Ill Observations where unity of invention is lacking (Continuation of item 3 of first sheet)

This International Searching Authority found multiple inventions in this international application, as follows:

see addi t i onal sheet

□ As all required additional search fees were timely paid by the applicant, this international search report covers all searchable claims.

□ As all searchable claims could be searched without effort justifying an additional fees, this Authority did not invite payment of additional fees.

As only some of the required additional search fees were timely paid by the applicant, this international search report covers ' ' only those claims for which fees were paid, specifically claims Nos. :

4 . I I No required additional search fees were timely paid by the applicant. Consequently, this international search report is restricted to the invention first mentioned in the claims; it is covered by claims Nos. :

1-17 , 23 (compl etely) ; 18-22 , 24-28(parti al ly)

Remark on Protest The additional search fees were accompanied by the applicant's protest and, where applicable, the ' ' payment of a protest fee. The additional search fees were accompanied by the applicant's protest but the applicable protest ' ' fee was not paid within the time limit specified in the invitation.

I INo protest accompanied the payment of additional search fees.

Form PCT/ISA/21 0 (continuation of first sheet (2)) (April 2005) International Application No. PCTV EP2011/ 005142

FURTHER INFORMATION CONTINUED FROM PCT/ISA/ 210

Thi s Internati onal Searchi ng Authori t y found mul t i pl e (groups i nventi ons i n thi s i nternati onal appl i cati on , as fol l ows :

1. cl aims : 1-17 , 23 (compl etely) ; 18-22 , 24-28(parti al ly)

compounds ( I ) , composi t i ons , use and preparati on

2. cl aims : 18-22 , 24-28(al l parti al ly)

compounds ( I I I ) and i t s use Patent document Publication Patent family Publication cited in search report date member(s) date

US 7271186 B l 18-09-2007 US 7271186 B l 18-09-2007 US 7928129 B l 19-04-2011

FR 2758329 A l 17-07-1998 NONE

US 5731439 A 24-03-1998 NONE