Novel 3' -derivatives of antibacterial 4, 6-disubstituted 2, 5-dideoxytetracycline aminoglycoside antibiotics

文档序号:1894601 发布日期:2021-11-26 浏览:22次 中文

阅读说明:本技术 新的抗菌的4,6-二取代的2,5-二脱氧曲霉素氨基糖苷类抗生素的3″-衍生物 (Novel 3' -derivatives of antibacterial 4, 6-disubstituted 2, 5-dideoxytetracycline aminoglycoside antibiotics ) 是由 安德烈亚斯·亚力山大·巴斯蒂安 玛丽亚·巴斯蒂安 于 2019-12-05 设计创作,主要内容包括:本发明涉及具有抗微生物性质且适合例如作为用于治疗哺乳动物疾病的治疗剂的新的氨基糖苷化合物,特别是用于治疗哺乳动物中的微生物感染的新颖治疗剂。本发明还涉及包括所述剂的药物组合物在治疗哺乳动物的医学病症中,特别是在治疗微生物感染中,的用途。本发明的剂和药物组合物在治疗与抗生素耐药性微生物相关的疾病中特别相关。本发明还涉及用于治疗由于抗生素类相关细菌耐药性而难以治疗的疾病的化合物,并提供了适用于治疗多重耐药(MDR)感染的新的治疗剂。(The present invention relates to novel aminoglycoside compounds having antimicrobial properties and being suitable, for example, as therapeutic agents for the treatment of diseases in mammals, in particular for the treatment of microbial infections in mammals. The invention also relates to the use of a pharmaceutical composition comprising said agent in the treatment of a medical condition in a mammal, in particular in the treatment of a microbial infection. The agents and pharmaceutical compositions of the invention are particularly relevant in the treatment of diseases associated with antibiotic resistant microorganisms. The invention also relates to compounds useful for treating diseases that are refractory to antibiotic-associated bacterial resistance, and provides novel therapeutic agents useful for treating multidrug-resistant (MDR) infections.)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof,

wherein

(i)R1Selected from H, methyl, ethyl, straight or branched C3-6Alkyl radical, C3-6A cycloalkyl group, a,

Wherein is a group represented by the formula (I) and R in the formula (I)1The point of attachment of the attached N atom;

(ii)R2selected from the group consisting of H, methyl, -CH2F、-CF3Ethyl, n-propyl, isopropyl, cyclopropyl, halogen, hydroxy, -OCH3、-OEt、-OCH2F、-OCF3、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2、-NHCH2F、-NHCF3and-NHQ, and;

wherein when R is2Is ethyl, n-propyl, isopropyl, cyclopropyl, -OEt, -NHEt or-N (Et)2When R is in the above-mentioned range2The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OEt、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2and-NHQ, substituted with a substituent group; with the proviso that when R2is-OEt, -NHEt or-N (Et)2When the-OEt, -NHEt or-N (Et)2Et group of group linked to the-OEt, -NHEt or-N (Et)2The carbon atoms of the O or N atoms of the group may be substituted only by one or more substituents independently selected from halogen.

(iii)R3Selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -NH 2、-NHCH3、-NHCH2F、-NHCF3、-NHEt、-NHC3-8Alkyl, -N (CH)3)2、-N(Et)2、-N(C3-8Alkyl radical)2and-NHQ, and;

wherein when R is3is-OEt, -OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl radicals、-NHEt、-NHC3-8Alkyl, -N (Et)2、-N(C3-8Alkyl radical)2When R is in the above-mentioned range3The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; provided that it is directly bonded to each of said R3Each said R on O or N atom in the group3The carbon atoms in the group may be substituted only with one or more substituents independently selected from halogen; and is

Wherein when R is3is-OCH3or-NHCH3When it is in the-OCH3or-NHCH3CH of a radical3Phenyl optionally substituted in part; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and s; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S;

(iv)R4selected from the group consisting of H, methyl, ethyl, -CH2F、-CF3Straight or branched chain C 3-6Alkyl, substituted straight chain C2-6Alkyl, substituted branched C3-6Alkyl, optionally substituted C3-6Cycloalkyl, optionally substituted-CH2C3-6Cycloalkyl, formyl, optionally substituted phenyl, optionally substituted 5-or 6-membered heteroaryl,

Wherein is a group represented by the formula (I) and R in the formula (I)4A point of attachment of the N atom attached thereto, and wherein

When R is4Is a substituted straight chain C2-6Alkyl, substituted branched C3-6Alkyl, substituted C3-6Cycloalkyl, or substituted-CH2C3-6When cycloalkyl, it is substituted with one or more groups independently selected from halogen, hydroxy, -OCH3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; provided that it is directly bonded to R in the structure of formula (I)4Said R on the attached N atom4The carbon atoms of the group may be substituted only with one or more substituents independently selected from halogen;

(v)R5is selected from the group consisting of H; a methyl group; -CH2F;-CF3(ii) a An ethyl group; straight or branched C3-8An alkyl group; substituted straight chain C2-8An alkyl group; substituted branched C3-8An alkyl group; optionally substituted C3-6A cycloalkyl group; optionally substituted-CH2C3-6A cycloalkyl group; optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; -C (═ NH) NH 2;-C(=NR7)NH2;-C(=NH)NHR8;-C(=NR7)NHR8;-C(=NR7)NR8R9(ii) a And

-X-Z, wherein

X is selected from methylene, ethylene, linear or branched C3-8Alkylene, the group consisting of; each of which, in addition to being linked to Z, may optionally be further substituted by oneOne or more is/are independently selected from the group consisting of methyl, -CH2F、-CF3Ethyl, straight or branched C3-6Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR10、-NR10R11、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR12、-CONR12R13、-NHCOCH3、-NHCOC2-6Alkyl, -NR14COCH3and-NR15COC2-6Alkyl, substituted with a substituent of the group consisting of; provided that it is linked to R in the structure of formula (1)5The atoms of the x group on the attached N atom cannot be directly attached to another O or N atom;

and is

Z is selected from optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted C3-6Cycloalkyl, the group consisting of;

wherein R is7To R15Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C 3-6Cycloalkyl, the group consisting of;

(vi)R6is OH or NH2

(vii) Provided that for all compounds of formula (I) or pharmaceutically acceptable salts thereof, when in the structure of formula (I), it is linked to it and R3R to which both are attached2Is O or N, to the R of the tetrahydropyran ring3The atom(s) of (a) cannot be an O or N atom; provided that when in the structure of formula (I), it is linked to it and R2R to which both are attached3When the atom of (A) is an O or N atomR connecting it to the tetrahydropyran ring2The atom(s) of (a) cannot be an O or N atom; and is

(viii) Wherein for each group disclosed

m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

p is 0, 1, or 2;

q is 1, 2, 3, 4, or 5;

r is 1, 2, 3, or 4; and is

(ix) Wherein with respect to all of the above substituents and groups containing a Q moiety, Q isWherein x is the point of attachment to the N atom to which Q is attached.

2. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein R2Selected from the group consisting of H, methyl, -CH2F、-CF3Ethyl, n-propyl, isopropyl, cyclopropyl, halogen, hydroxy, -OCH3、-OEt、-OCH2F、-OCF3、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2、-NHCH2F、-NHCF3and-NHQ.

3. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R 2Selected from the group consisting of H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, -F, hydroxy, -OCH3、-OEt、-OCH2F and-OCF3Group (b), group (b).

4. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3Selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, and-OCH2C3-6A cycloalkyl group, a group consisting of,

wherein when R is3is-OEt, -OC3-8Alkyl, -OC3-6Cycloalkyl, or-OCH2C3-6When cycloalkyl is present, said R3The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (cH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; provided that it is directly bonded to each of said R3Each said R on the O atom in the group3The carbon atoms in the group may be substituted only with one or more substituents independently selected from halogen; and is

Wherein when R is3is-OCH3When it is in the-OCH3CH of a radical3Phenyl, part of which is optionally substituted; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and s.

5. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3Selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S), -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S), and-OCH2(optionally substituted with 1, 2 or 3 substituents selected from O, N anda 6-membered non-aromatic heterocycloalkyl group for the heteroatom of S).

6. The compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is selected from compounds of formulae (Ia) to (If)

Wherein, independently, for each of the compounds of formulae (Ia) to (If), R1、R4、R5And R6As defined in any one of the preceding claims and wherein for each of the compounds of formulae (Ie) to (If), W is independently selected from the group consisting of-OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH 2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and s); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S); and-OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

7. The compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is selected from compounds of formulae (Ig) to (Ip)

Wherein, independently, for each of the compounds of formulae (Ig) to (Ip), R1、R4And R5As defined In any one of the preceding claims and wherein for each of the compounds of formulae (In) to (Ip), W is independently selected from the group consisting of-OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and s); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S); and-OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

8. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R1Selected from H, methyl, ethyl, straight or branched C3-6Alkyl radical, C3-6A cycloalkyl group, a,The group consisting of;

preferably selected from the group consisting of H,The group consisting of;

wherein at said R1In part, Q and Q are as defined in claim 1.

9. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4Is selected from the group consisting of H,Group ofAnd wherein at said R4In which r and Q are as defined in claim 1.

10. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4Is selected from the group consisting of H,The group consisting of.

11. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R5Selected from H, methyl, ethyl, straight or branched C3-8Alkyl, substituted straight chain C2-8Alkyl, substituted branched C3-8Alkyl, optionally substituted C3-6A cycloalkyl group, a,

Wherein is a group represented by the formula (I) and R in the formula (I)5The point of attachment of the N atom to which it is attached, and

wherein A is selected from optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; and is

Wherein at said R5In the moiety, m, n, q, p and R10As defined in any one of the preceding claims.

12. A compound or according to any preceding claimA pharmaceutically acceptable salt thereof, wherein R5Selected from H, methyl, ethyl, straight or branched C3-6Alkyl, aryl, heteroaryl, and heteroaryl,

Wherein in the R5In moiety, Q and R10As defined in any one of the preceding claims.

13. A pharmaceutical composition comprising a compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

14. A composition comprising at least one compound according to any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, and at least one further antibacterial agent, wherein the at least one further antibacterial agent is different from the at least one compound according to any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof.

15. A compound according to any one of claims 1 to 12 or a pharmaceutically acceptable salt thereof or a composition according to any one of claims 13 to 14 for use as a medicament.

Technical Field

The present invention relates to novel aminoglycoside compounds having antimicrobial properties and being suitable, for example, as therapeutic agents for the treatment of diseases in mammals, in particular for the treatment of microbial infections in mammals. The invention also relates to the use of a pharmaceutical composition comprising said agent in the treatment of a medical condition in a mammal, in particular in the treatment of a microbial infection. The agents and pharmaceutical compositions of the invention are particularly relevant in the treatment of diseases associated with antibiotic resistant microorganisms. The invention also relates to compounds useful for treating diseases that are refractory to antibiotic-associated bacterial resistance, and provides novel therapeutic agents useful for treating multidrug-resistant (MDR) infections. The invention also relates to a method for producing said agents.

Background

As antibiotic resistance constitutes a serious and growing phenomenon in contemporary medicine and has emerged as a primary public health concern in the 21 st century (particularly in the case of hospital-acquired infections), society is using up life-threatening bacterial infections as a treatment option. Existing antibiotics are losing their efficacy against multiple resistant pathogens. Furthermore, although bacterial resistance continues to increase, the rate of new antibiotic approval decreases. Thus, society has entered a "post-antibiotic age" where hospitalized patients in a critical state cannot be effectively treated or cured. A particularly significant life threat is caused byESKAPEMembers of the group (enterococcus faecium (+), staphylococcus aureus (+), klebsiella pneumoniae (-), acinetobacter baumannii (-), pseudomonas aeruginosa (-), enterobacter species (-), and according to data obtained in the center for disease control and prevention (CDC) in 2008, these six bacteria caused two thirds of all health care-related infections. In addition, other details that pose a significant threat to public healthBacteria, such as E.coli, particularly strains of E.coli which are resistant to known antibacterial agents.

Infection by several drug resistant gram negative bacteria, such as MDR pseudomonas aeruginosa, acinetobacter baumannii, and carbapenem resistant klebsiella species, has attracted considerable attention. The treatment options for these pathogens are so limited that clinicians are often forced to use older drugs, such as polymyxins, which have been previously abandoned, which are associated with significant toxicity.

Aminoglycoside antibiotics, such as amikacin, are one of the last tools as antimicrobials for the treatment of life-threatening MDR gram-negative infections. They are safe, induce less bacterial resistance than other antibiotic types, and are resistant to antibiotics belonging to the above-mentioned groupESKAPEThe pathogens of the group have a broad spectrum of activity. However, due to the continued emergence of bacterial resistance, their clinical use is at risk.

One of the most clinically relevant mechanisms of bacterial resistance is based on the enzymatic inactivation of the Aminoglycoside Modifying Enzyme (AME) to drug molecules. The AME produced by pathogens changes the molecular structure of aminoglycosides, reducing their binding affinity to the target protein, 30S ribosome, rendering them inactive against drug-resistant pathogens. AME is classified into three enzyme types: the amino moiety in aminoglycosides can be acetylated by aminoglycoside-N-acetyltransferase (AAC), while the hydroxyl groups in aminoglycosides can be phosphorylated by aminoglycoside-O-phosphotransferase (APH) or modified by aminoglycoside-O-nucleotidyl transferase (ANT). In view of this, studies of the mode of action and site of AME have made site-specific modification of aminoglycosides a promising tool for the development of antibiotics aimed at overcoming the mechanisms of bacterial resistance.

In this regard, one of the most effective modifications of aminoglycosides was the introduction of the (2S) -4-amino-2-hydroxybutyrate (AHB) residue into kanamycin A, which led to the development of the semi-synthetic antibiotic amikacin. Amikacin is currently used clinically against MDR bacterial infections. However, due to the continued development of bacterial resistance, the clinical use of amikacin is at risk.

Therefore, attempts are being made in the field to develop the next generation of semisynthetic aminoglycoside antibiotics. However, research has generally focused on single structural modifications, which has resulted in limited ability of antibiotic molecules to simultaneously overcome resistance caused by multiple AME. In addition, these semisynthetic aminoglycosides exhibit a narrow spectrum of activity because their structural modifications typically increase antibacterial activity against one type of bacteria, but decrease activity against one or more other types of bacteria.

In this regard, the introduction of a guanidino group at the 3 "C position of kanamycin has been reported (Santana, A.G., Z a rate, S.G., Asensio, J.L., Revuelta, J., Bastida, A.Selective modification of the 3" -amino group of kanamyn precursors of activity in resistance bacteria strain. org.biomol.chem.14,2016, 516-525). However, this structural modification can result in an aminoglycoside derivative, which has a significantly reduced potency, particularly against certain bacterial species, such as E.coli, E.faecalis and P.aeruginosa, thereby significantly limiting the applicability and relevance of the derivative.

Another modification that has been reported is the fluorinated aminoglycoside at the 5-position (also referred to as the 5-C position) of the 2-deoxystreptavidin (2-DOS) ring. In some cases, this structural modification is taught to be associated with reduced toxicity of the resulting aminoglycoside (T.Tsuchiya, T.Shitara, S.Umezawa, T.Takeuchi, M.Hamada, N.Tomono, and E.Umemura.Synthesis of low-toxicity,5-deoxy-5-fluoro and 5-deoxy-5,5-difluoro derivative of arbekacin and identity, and determination of structure-reactivity related. Carbohydr.Res.240,1993, 307-312; T.Shitara, Y.Kobayashi, T.Tsuchiya, and S.Umezzia of 5-deoxy-5-fluoro derivative of variance, 5-fluoro derivative of carbohydrate, 273, 5-molecular derivative of carbohydrate, 273, and 273).

Despite these studies, there remains no problem in identifying new aminoglycoside compounds which are both synthetic and commercially viable and which not only overcome bacterial resistance, but also show a broad, preferably improved antibacterial activity against selected bacterial strains belonging to the ESKAPE group, in particular with respect to known aminoglycosides, such as amikacin and gentamicin. In this regard, providing compounds with improved antibacterial activity has the potential to effectively treat microbial infections at lower doses, which in turn brings the advantage of being able to treat such infections while reducing the risk of drug-class related toxicities such as nephrotoxicity and ototoxicity. Therefore, it is an object of the present invention to provide such compounds.

It is another object of the present invention to provide novel aminoglycoside compounds which exhibit excellent and preferably improved activity against at least one, preferably more than one of the following bacterial targets: coli, escherichia coli expressing one or more of AME APH (3 ') IIIa, APH (3') Ia, AAC (6 ') Ie-APH (2 ") Ia, AAC (6') Ib, AAC (3) III and AAC (3) IV (or escherichia coli expressing any one or more other AMEs expressed in the strains to which the activity data of the compounds of the invention are provided herein), enterococcus faecium, enterococcus faecalis, staphylococcus aureus, klebsiella pneumoniae, acinetobacter baumannii, pseudomonas aeruginosa, species of enterobacter, morganella morgangii, providencia stuartii and enterobacter cloacae (or strains expressing any one or more of these bacteria of any one or more of the AMEs disclosed herein), in particular strains that are multidrug or pan-drug resistant (PDR) to the bacterial target.

Another object of the present invention is to provide novel aminoglycoside compounds which are resistant to E.coli, in particular to aminoglycoside antibiotics (e.g.amikacin and/or gentamicin) in strains of E.coli, such as strains containing plasmids encoding one or more AMEs such as APH (3 ') IIIa, APH (3 ') Ia, AAC (6 ') Ie-APH (2 ') Ia, AAC (6 ') Ib, AAC (3) III and AAC (3) IV (or any other AME or AMEs expressed in the strains for which activity data for the compounds of the invention are provided herein), have excellent and preferably improved activity, and has excellent and preferably improved activity against one or more of enterococcus faecium, enterococcus faecalis, klebsiella pneumoniae, acinetobacter baumannii, pseudomonas cloacae, pseudomonas aeruginosa, morganella, providencia stuartii and staphylococcus aureus.

It is a potential object of the present invention to provide compounds that exhibit excellent and preferably improved activity against any one of the targets (or any combination of two or more targets) described herein and/or against which the experimental data disclosed herein is directed.

It is another object of the present invention to provide compounds that restore activity against certain bacterial targets that are lost following structural modification to obtain activity against one or more other bacterial targets, particularly when the over bacterial targets are resistant to one or more aminoglycoside antibiotics. It is therefore an object of the present invention to provide compounds that broaden the spectrum of bacterial targets on which certain aminoglycosides exhibit activity.

Disclosure of Invention

The present invention relates to a novel therapeutic agent, in particular a novel compound useful in medicine, in particular for the treatment of disease in a mammal, and more particularly to a novel compound useful in the treatment of microbial infections, in particular in a mammal. The compounds of the present invention are believed to overcome bacterial resistance caused by AME and simultaneously increase antibacterial activity by up to 16-fold, particularly for bacteria of the ESKAPE group, relative to amikacin antibiotics (4, 6-disubstituted 2-deoxystreptavidin (2-DOS) aminoglycoside carrying a (2S) -4-amino-2-hydroxybutyrate (AHB) group at position N1). As demonstrated herein, the NH at the 5-C position (resulting in a 5-epi-5-fluoro group, i.e., as described below in the structure of formula (I)) and at the 3 "-C position (also referred to as the 3" position) of the aminoglycoside scaffold 2The incorporation of a guanidine or functionalized guanidine moiety in the group results in a completely unexpected synergistic effect, resulting in aminoglycoside aminoglycosides having significantly improved antibacterial activity. Furthermore, the presence of substituted/functionalized guanidine moieties at the 3 "-C position is completely unknown in the art, let alone they exhibit superior levels of in vitro and in vivo efficacy against the specific targets demonstrated herein.

Thus, the compounds of the present invention are particularly useful for (1) treating bacterial resistance, (2) enhancing and broadening antibacterial activity against gram-positive and gram-negative bacteria, and (3) restoring antibacterial activity against one or more targets lost as a result of other structural modifications.

Specifically, the present invention relates to the following:

item (1):

1. a compound of formula (I) or a pharmaceutically acceptable salt thereof,

wherein

(i)R1Selected from H, methyl, ethyl, straight or branched C3-6Alkyl radical, C3-6A cycloalkyl group, a,

Wherein is a group represented by the formula (I) and R in the formula (I)1The point of attachment of the attached N atom;

(ii)R2selected from the group consisting of H, methyl, -CH2F、-CF3Ethyl, n-propyl, isopropyl, cyclopropyl, halogen, hydroxy, -OCH3、-OEt、-OCH2F、-OCF3、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2、-NHCH2F、-NHCF3and-NHQ, and;

wherein when R is2Is ethyl, n-propyl, isopropyl, cyclopropyl, -OEt, -NHEt or-N (Et) 2When R is in the above-mentioned range2The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OEt、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2and-NHQ, substituted with a substituent group; with the proviso that when R2is-OEt, -NHEt or-N (Et)2When the-OEt, -NHEt or-N (Et)2Et group of group linked to the-OEt, -NHEt or-N (Et)2The carbon atoms of the O or N atoms of the group may be substituted only by one or more substituents independently selected from halogen;

(iii)R3selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -NH2、-NHCH3、-NHCH2F、-NHCF3、-NHEt、-NHC3-8Alkyl, -N (CH)3)2、-N(Et)2、-N(C3-8Alkyl radical)2and-NHQ, and;

wherein when R is3is-OEt, -OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -NHEt, -NHC3-8Alkyl, -N (Et)2、-N(C3-8Alkyl radical)2When R is in the above-mentioned range3The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; provided that it is directly bonded to each of said R3Each said R on O or N atom in the group3The carbon atoms in the group may be substituted only with one or more substituents independently selected from halogen; and is

Wherein when R is3is-OCH 3or-NHCH3When it is in the-OCH3or-NHCH3CH of a radical3Phenyl optionally substituted in part; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted containing 1, 2 or 3 substituents selected from O, N and SHeteroatom 6-membered non-aromatic heterocycloalkyl;

(iv)R4selected from the group consisting of H, methyl, ethyl, -CH2F、-CF3Straight or branched chain C3-6Alkyl, substituted straight chain C2-6Alkyl, substituted branched C3-6Alkyl, optionally substituted C3-6Cycloalkyl, optionally substituted-CH2C3-6Cycloalkyl, formyl, optionally substituted phenyl, optionally substituted 5-or 6-membered heteroaryl,

Wherein is a group represented by the formula (I) and R in the formula (I)4A point of attachment of the N atom attached thereto, and wherein

When R is4Is a substituted straight chain C2-6Alkyl, substituted branched C3-6Alkyl, substituted C3-6Cycloalkyl, or substituted-CH2C3-6When cycloalkyl, it is substituted with one or more groups independently selected from halogen, hydroxy, -OCH3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; provided that it is directly bonded to R in the structure of formula (I) 4Said R on the attached N atom4The carbon atoms of the group may be substituted only with one or more substituents independently selected from halogen;

(v)R5is selected from the group consisting of H; a methyl group; -CH2F;-CF3(ii) a Second stepA group; straight or branched C3-8An alkyl group; substituted straight chain C2-8An alkyl group; substituted branched C3-8An alkyl group; optionally substituted C3-6A cycloalkyl group; optionally substituted-CH2C3-6A cycloalkyl group; optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; -C (═ NH) NH2;-C(=NR7)NH2;-C(=NH)NHR8;-C(=NR7)NHR8;-C(=NR7)NR8R9(ii) a And

-X-Z, wherein

X is selected from methylene, ethylene, linear or branched C3-8Alkylene, the group consisting of; each of which, in addition to being linked to Z, may optionally be further substituted by one or more substituents independently selected from the group consisting of methyl, -CH2F、-CF3Ethyl, straight or branched C3-6Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR10、-NR10R11、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR12、-CONR12R13、-NHCOCH3、-NHCOC2-6Alkyl, -NR14COCH3and-NR15COC2-6Alkyl, substituted with a substituent of the group consisting of; provided that it is linked to R in the structure of formula (I) 5The atoms of the X group on the attached N atom cannot be directly attached to another O or N atom;

and is

Z is selected from optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted heteroatom containing 1 heteroatom selected from O, N and S4-membered non-aromatic heterocycloalkyl of (a); optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted C3-6Cycloalkyl, the group consisting of;

wherein R is7To R15Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C3-6Cycloalkyl, the group consisting of;

(vi)R6is OH or NH2

(vii) With the proviso that for all compounds of formula (I) or pharmaceutically acceptable salts thereof,

when in the structure of formula (I), it is attached to it and R3R to which both are attached2Is O or N, to the R of the tetrahydropyran ring3The atom(s) of (a) cannot be an O or N atom; provided that when in the structure of formula (I), it is linked to it and R2R to which both are attached3R connecting it to said tetrahydropyran ring when the atom of (A) is an O or N atom 2The atom(s) of (a) cannot be an O or N atom; and is

(viii) Wherein for each group disclosed

m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

p is 0, 1, or 2;

q is 1, 2, 3, 4, or 5;

r is 1, 2, 3, or 4; and is

(ix) Wherein with respect to all of the above substituents and groups containing a Q moiety, Q isWherein x is the point of attachment to the N atom to which Q is attached.

2. A compound according to item 1, or a pharmaceutically acceptable salt thereof, wherein R2Selected from the group consisting of H, methyl, -CH2F、-CF3Ethyl, n-propyl, isopropyl, cyclopropyl, halogen, hydroxy, -OCH3、-OEt、-OCH2F、-OCF3、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2、-NHCH2F、-NHCF3and-NHQ.

3. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R2Selected from the group consisting of H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, -F, hydroxy, -OCH3、-OEt、-OCH2F and-OCF3Group (b), group (b).

4. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R2Selected from the group consisting of H, methyl, and hydroxy.

5. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R2Selected from the group consisting of H, and methyl.

6. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R 2Is H.

7. A compound according to any one of items 1 to 5, or a pharmaceutically acceptable salt thereof, wherein R2Is methyl.

8. A compound according to any one of items 1 to 4, or a pharmaceutically acceptable salt thereof, wherein R2Is a hydroxyl group.

9. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3Selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, and-OCH2C3-6A cycloalkyl group, a group consisting of,

wherein when R is3is-OEt, -OC3-8Alkyl, -OC3-6Cycloalkyl, or-OCH2C3-6When cycloalkyl is present, said R3The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; provided that it is directly bonded to each of said R3Each said R on the O atom in the group3The carbon atoms in the group may be substituted only with one or more substituents independently selected from halogen; and is

Wherein when R is3is-OCH3When it is in the-OCH3CH of a radical3Phenyl, part of which is optionally substituted; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S.

10. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when R3is-OCH3When said-OCH is3CH of a radical3Phenyl, partially optionally substituted; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S;

optionally substituted means optionally substituted with one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC3-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; wherein R is16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C3-6Cycloalkyl, the group consisting of; with the proviso that when one of said substituents is directly attached to the N atom in the respective non-aromatic heterocycloalkyl moiety, it may be selected only from the group consisting of the free methyl radicals, -CH 2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b).

11. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when R3is-NHCH3When the-NHCH is used3CH of a radical3Phenyl, partially optionally substituted; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S;

optionally substituted with one or more substituents independently selected from the group consisting of methyl, -CH2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; wherein R is16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C3-6Cycloalkyl, the group consisting of; with the proviso that when one of said substituents is directly attached to the N atom in the respective non-aromatic heterocycloalkyl moiety, it may be selected only from the group consisting of the free methyl radicals, -CH 2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b).

12. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3Selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S), -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S), and-OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

13. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R2Is H or methyl, and R3Selected from the group consisting of H, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S); -OCH 2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S); and-OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

14. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3Selected from the group consisting of H, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, and-OCH2C3-6Cycloalkyl groups, or a combination thereof.

15. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3Selected from the group consisting of H and hydroxy.

16. A compound according to any one of the preceding claims or a pharmaceutically acceptable salt thereof,wherein R is3Is H.

17. A compound according to any one of items 1 to 15, or a pharmaceutically acceptable salt thereof, wherein R3Is a hydroxyl group.

18. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is selected from compounds of formulae (Ia) to (If)

Wherein, independently, for each of the compounds of formulae (Ia) to (If), R1、R4、R5And R6As defined in any of the preceding claims, and wherein for each of the compounds of formulae (Ie) to (If), W is independently selected from the group consisting of-OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH 2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S); and-OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

19. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R6Is OH.

20. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R6Is NH2

21. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is selected from compounds of formulae (Ig) to (Ip)

Wherein, independently, for each of the compounds of formulae (Ig) to (Ip), R1、R4And R5As defined In any of the preceding claims, and wherein for each of the compounds of formulae (In) to (Ip), W is independently selected from the group consisting of-OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH 2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S); and-OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

22. A compound according to any one of claims 18 to 21, or a pharmaceutically acceptable salt thereof, wherein W may be-OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; -OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S);

optionally substituted means optionally substituted with one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR 22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; wherein R is16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl radical, C3-6Cycloalkyl, the group consisting of; with the proviso that when one of said substituents is directly attached to the N atom in the respective non-aromatic heterocycloalkyl moiety, it may be selected only from the group consisting of the free methyl radicals, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b).

23. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R1Selected from H, methyl, ethyl, straight or branched C3-6Alkyl radical, C3-6A cycloalkyl group, a,The group consisting of;

preferably selected from the group consisting of H,The group of the said components is composed of,

wherein at said R1In part, Q and Q are as defined in item 1.

24. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R1Is selected from the group consisting of H,The group consisting of; and is

Wherein Q is as defined in item 1, and for each moiety, the R1Q in moieties is independently 1, 2 or 3, most preferably wherein said R1In the section, q is 1.

25. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R 1Is H.

A compound according to any one of items 1 to 24, or a pharmaceutically acceptable salt thereof, wherein R1Is: -CH2CH2OH (i.e. the) Wherein is and R in formula (I)1The point of attachment of the attached N atom.

26. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4Is selected from the group consisting of H,And wherein in said R4In part, r and Q are as defined in item 1.

27. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4Is selected from the group consisting of H,The group consisting of.

28. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4Is H.

29. A compound according to any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof, wherein R4Is composed of

30. A compound according to any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof, wherein R4Is composed of

31. A compound according to any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof, wherein R4Is composed of

32. A compound according to any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof, wherein R4Is composed of

33. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when R5Is an optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S;

Said optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; selected from the group consisting of optionally substituted azetidinyl, optionally substituted diazetidinyl, optionally substituted oxetanyl, optionally substituted thietanyl, optionally substituted pyrrolidinyl, optionally substituted tetrahydrofuranyl, optionally substituted tetrahydrothienyl, optionally substituted piperidinyl, optionally substituted tetrahydropyranyl, optionally substituted thianyl, optionally substituted imidazolidinyl, optionally substituted pyrazolidinyl, optionally substituted oxazolidinyl, optionally substituted isoxazolidinyl, optionally substituted thiazolidinyl, optionally substituted isothiazolidinyl, optionally substituted dioxolanyl, optionally substituted dithiolanyl, optionally substituted piperazinyl, optionally substituted morpholinyl, optionally substituted thiomorpholinyl, optionally substituted 1, 3-dioxanyl, optionally substituted 1, 4-dioxanyl, Optionally substituted 1, 3-dithianyl, and optionally substituted 1, 4-dithianyl.

34. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when R5Is an optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S;

said optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; selected from the group consisting of 4-piperidinyl, 3-piperidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 4-oxazolidinyl, 2-morpholinyl, 3-morpholinyl, 4-morpholinyl, 2-tetrahydrofuranyl, and 3-tetrahydrofuranyl.

35. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when Z may be an optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S;

Said optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; selected from the group consisting of optionally substituted azetidinyl, optionally substituted diazetidinyl, optionally substituted oxetanyl, optionally substituted thietanyl, optionally substituted pyrrolidinyl, optionally substituted tetrahydrofuranyl, optionally substituted tetrahydrothienyl, optionally substituted piperidinyl, optionally substituted tetrahydropyranyl, optionally substituted thianyl, optionally substituted imidazolidinyl, optionally substituted pyrazolidinyl, optionally substituted oxazolidinyl, optionally substituted isoxazolidinyl, optionally substituted thiazolidinyl, optionally substituted isothiazolidinyl, optionally substituted dioxolanyl, optionally substituted dithiolanyl, optionally substituted piperazinyl, optionally substituted morpholinyl, optionally substituted thiomorpholinyl, optionally substituted 1, 3-dioxanyl, optionally substituted 1, 4-dioxanyl, Optionally substituted 1, 3-dithianyl, and optionally substituted 1, 4-dithianyl.

36. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when Z may be an optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S;

said optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; selected from the group consisting of 4-piperidinyl, 3-piperidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 4-oxazolidinyl, 2-morpholinyl, 3-morpholinyl, 4-morpholinyl, 2-tetrahydrofuranyl, and 3-tetrahydrofuranyl.

37. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R5Selected from H, methyl, ethyl, straight or branched C3-8Alkyl, substituted straight chain C2-8Alkyl, substituted branched C3-8Alkyl, optionally substituted C 3-6A cycloalkyl group, a,

Wherein is a group represented by the formula (I) and R in the formula (I)5The point of attachment of the N atom to which it is attached, and

wherein A is selected from optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; and is

Wherein at said R5In the moiety, m, n, q, p and R10As defined in any one of the preceding claims.

38. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when R5Is optionally substituted straight chain C2-8An alkyl group; optionally substituted branched C3-8An alkyl group; optionally substituted C3-6A cycloalkyl group; or optionally substituted-CH2C3-6When cycloalkyl, optionally substituted means optionally substituted with one or more substituents independently selected from the group consisting of methyl, -CH2F、-CF3Ethyl, straight or branched C3-6Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR10、-NR10R11、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR7、-CONR12R13、-NHCOCH3、-NHCOC2-6Alkyl, -NR14COCH3and-NR15COC2-6Alkyl, substituted with a substituent of the group consisting of; wherein R is 7To R15Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C3-6Cycloalkyl groups, or a combination thereof.

39. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when R5Is optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted means optionally substituted with one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; with the proviso that when one of said substituents is directly attached to the N atom in the corresponding non-aromatic heterocycloalkyl moiety, it may be selected only from the group consisting of the free methyl radicals, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR 19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group consisting of; wherein R is16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C3-6Cycloalkyl groups, or a combination thereof.

40. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein when Z is optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted means optionally substituted with one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; with the proviso that when one of said substituents is directly attached to the N atom in the corresponding non-aromatic heterocycloalkyl moiety, it may be selected only from the group consisting of the free methyl radicals, -CH2F、-CF3Ethyl, straight or branched C 3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group consisting of; wherein R is16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C3-6Cycloalkyl groups, or a combination thereof.

41. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R5Selected from H, methyl, ethyl, straight or branched C3-6Alkyl, aryl, heteroaryl, and heteroaryl,

Wherein in the R5In moiety, Q and R10As defined in any one of the preceding claims.

42. A compound according to any one of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R5Is selected from the group consisting of H,

The group consisting of.

43. A composition comprising at least one compound of any one of the foregoing or a pharmaceutically acceptable salt thereof.

44. A pharmaceutical composition comprising a composition, compound or pharmaceutically acceptable salt thereof according to any one of the preceding.

45. A pharmaceutical composition comprising a composition, compound or pharmaceutically acceptable salt thereof according to any one of the preceding, and a pharmaceutically acceptable carrier.

46. A pharmaceutical composition comprising a composition, compound or pharmaceutically acceptable salt thereof according to any one of the preceding, and a pharmaceutically acceptable carrier, diluent or excipient.

47. A composition comprising at least one compound according to any one of items 1 to 42 or a pharmaceutically acceptable salt thereof and at least one further antibacterial agent, wherein the at least one further antibacterial agent is different from the at least one compound according to any one of items 1 to 42 or a pharmaceutically acceptable salt thereof.

48. The composition of item 47, wherein the at least one further antibacterial agent is a regulatory approved antibiotic, in particular a regulatory approved member of the antibiotic family selected from the group consisting of penicillins, cephalosporins, carbapenems, monobactams, polymyxins, rifamycins, lipiarmycins, quinolones, sulfonamides, macrolides, lincosamides, tetracyclines, aminoglycosides, lipopeptides, glycylcyclics, glycopeptides, oxazolidinones, and lipiarmycins.

49. The composition according to item 47 or 48, wherein the at least one further antibacterial agent is a beta-lactam antibiotic, in particular a beta-lactam antibiotic selected from the group consisting of penicillin antibiotics, cephalosporin antibiotics, and carbapenem antibiotics, or any combination thereof.

50. The composition according to any one of items 47 to 49, wherein the composition further comprises at least one additional agent capable of overcoming one or more mechanisms of bacterial resistance.

51. The composition according to item 50, wherein the

(i) At least one further antibacterial agent is a β -lactam antibiotic, in particular an existing regulatory approved β -lactam antibiotic, more particularly an existing regulatory approved β -lactam antibiotic selected from the group consisting of penicillins, cephalosporins, and carbapenems, or any combination thereof; and is

(ii) The at least one additional agent capable of overcoming one or more bacterial resistance mechanisms is selected from the group consisting of clavulanic acid, sulbactam, tazobactam, avibactam, rilebabactam, fulibitan, and any combination thereof.

52. A composition according to item 51, wherein the combination of the beta-lactam antibiotic and the at least one additional agent capable of overcoming one or more bacterial resistance mechanisms is selected from the group consisting of

(i) Amoxicillin and clavulanic acid;

(ii) ticarcillin and clavulanic acid;

(iii) ampicillin and sulbactam;

(iv) cefoperazone and sulbactam;

(v) Piperacillin and tazobactam;

(vi) ceftizolidine and tazobactam;

(vii) ceftazidime and avibactam;

(viii) ceftaroline and abamectin;

(ix) carbapenems, in particular epetionam or meropenem, and releptin; and

(x) Carbapenems, in particular epeniam or meropenem, and faropenem,

the group consisting of.

52a. the composition of any one of claims 43 to 52, wherein the composition further comprises at least one adjuvant capable of inhibiting or preventing aminoglycoside-induced nephrotoxicity.

The composition of item 52b, wherein the at least one adjuvant capable of inhibiting or preventing aminoglycoside-induced nephrotoxicity is a member of a pharmacological class selected from the group consisting of antibiotics, calcium channel blockers, beta blockers, cytoprotective antianginals, iNOS inhibitors, NO precursors, hormones, antiplatelet drugs, statins, PPAR-gamma agonists, TNF-alpha synthesis inhibitors, biguanides, antioxidants, free radical scavengers, antioxidases, superoxide dismutase mimics, herbal extracts, and any combination thereof.

The composition according to item 52C, wherein the at least one adjuvant capable of inhibiting or preventing aminoglycoside-induced nephrotoxicity is selected from the group consisting of fosfomycin, fleroxacin, nifedipine, amlodipine, carvedilol, L-NIL, L-arginine, the hormones melatonin, thyroxine, trapidil, atorvastatin, rosiglitazone, pentoxifylline, metformin, probucol, aminoguanidine, L-carnitine, ebselen, N-acetylcysteine, lycopene, curcumin, thymoquinone, fish oil, vitamin E, vitamin C, sesame oil, halofuginone, resveratrol, quercetin, S-allylcysteine, diallyl sulfide, phenethyl caffeate, S-allylmercaptocysteine, superoxide dismutase mimics M40403, Rhazya stricta, Garlic, Cassia auriculata (Cassia auriculata), soybean, small return (phyllanthus amarus), Morchella esculenta (Morchella esculenta), green tea, Nigella sativa (Nigella sativa), Ligusticum wallichii (Ligusticum wallichii), mistletoe fascicularis (Viscum articula), gum arabic, Pongamia pinnata (Pongamia pinata) flowers, Nigella sativa oil, smilax indica (hemmesmus indicus), soyaphenol extract, green tea extract, purple flowers (Bauhinia purpurea), pseudo-gold flowers (Sida rhomoboidea), oleandrin, piperacillin, and any combination thereof.

53. Use of a compound according to any one of claims 1 to 42 for preventing, inhibiting or stopping bacterial growth on a surface.

54. A compound according to any one of claims 1 to 42 or a pharmaceutically acceptable salt thereof or a composition according to any one of claims 43 to 52c, for use as a medicament.

55. A compound according to any one of claims 1 to 42 or a pharmaceutically acceptable salt thereof or a composition according to any one of claims 43 to 52c, for use in the treatment of a microbial infection and/or a condition, affliction or disease caused, at least in part, by a microbial infection.

56. The compound, pharmaceutically acceptable salt thereof, or composition for use according to item 55, wherein the microbial infection is a bacterial infection.

57. The compound, pharmaceutically acceptable salt or composition thereof for use according to item 56, wherein the bacterial infection is caused at least in part by one or more gram positive bacterial species, particularly wherein the one or more gram positive bacterial species is selected from the group consisting of Staphylococcus aureus, Streptococcus pneumoniae, enterococcus faecalis, enterococcus faecium, and Mycobacterium tuberculosis.

58. The compound, pharmaceutically acceptable salt or composition thereof for use according to item 56, wherein the bacterial infection is caused at least in part by one or more gram-negative bacterial species, particularly wherein the one or more gram-negative bacterial species is selected from the group consisting of acinetobacter; enterobacteriaceae species, in particular escherichia coli species; a species of the genus Klebsiella, or a species of the genus Enterobacter; species of the genus mycobacterium; species of the genus Morganella; species of the genus providencia; and a species of Pseudomonas, to a subject; in particular selected from the group consisting of Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, Klebsiella oxytoca, Enterobacter cloacae, Enterobacter aerogenes, Mycobacterium tuberculosis, Morganella morganii, providencia stuartii, and Pseudomonas aeruginosa.

59. The compound, pharmaceutically acceptable salt or composition thereof for use according to item 56, wherein the bacterial infection is caused at least in part by one or more gram positive bacterial species and at least in part by one or more gram negative bacterial species, particularly wherein the one or more gram positive bacterial species is selected from the group consisting of staphylococcus aureus, streptococcus pneumoniae, enterococcus faecalis, enterococcus faecium and mycobacterium tuberculosis; and wherein the one or more gram-negative bacterial species are selected from the group consisting of acinetobacter; enterobacteriaceae species, in particular escherichia coli species; a species of the genus Klebsiella, or a species of the genus Enterobacter; species of the genus mycobacterium; species of the genus Morganella; species of the genus providencia; and a species of Pseudomonas, to a subject; in particular selected from the group consisting of Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, Klebsiella oxytoca, Enterobacter cloacae, Enterobacter aerogenes, Mycobacterium tuberculosis, Morganella morganii, providencia stuartii, and Pseudomonas aeruginosa.

60. The compound, pharmaceutically acceptable salt or composition thereof for use according to item 56, wherein the bacterial infection is at least partially by a species selected from the group consisting of Escherichia species, enterococcus species, Staphylococcus species, Klebsiella species, Acinetobacter species. Pseudomonas species, Enterobacter species, Morganella species, providencia species, Mycobacterium species, and any combination thereof.

61. The compound, pharmaceutically acceptable salt or composition thereof for use according to item 60, wherein the Escherichia coli species is Escherichia coli, preferably wherein the Escherichia coli species is selected from the group consisting of wild-type Escherichia coli and a strain of Escherichia coli expressing one or more aminoglycoside modifying enzymes.

62. The compound, pharmaceutically acceptable salt or composition thereof for use according to item 61, wherein the at least one aminoglycoside modifying enzyme is selected from the group consisting of aminoglycoside phosphotransferases, in particular APH (3 ') IIIa, APH (3') IIa, APH (3 ') Ia, APH (3 ") Ib, APH (3') VI, APH (6) Id, APH (3 ') VIa, APH (3') IIb and APH (6) Ic; aminoglycoside acetyltransferases, in particular AAC (6') Ib, AAC (3) III, AAC (3) IV, AAC (3) Ia, AAC (3) IId and AAC (3) Ic, more in particular AAC (3) III and AAC (3) IV; aminoglycoside nucleotidyl transferases, in particular ANT (3 ') Ia, ANT (2 ') Ia and ANT (3 '); and bifunctional aminoglycoside modifying enzymes, particularly AAC (6 ') Ie-APH (2') Ia; and any combination thereof.

63. The compound, pharmaceutically acceptable salt or composition thereof for use according to item 60, wherein the species of enterococcus is enterococcus faecium or enterococcus faecalis, particularly enterococcus faecalis; said species of Staphylococcus is Staphylococcus aureus; the species of the genus Klebsiella is Klebsiella pneumoniae; the species of Acinetobacter is Acinetobacter baumannii; the species of Pseudomonas is Pseudomonas aeruginosa; the species of the genus Enterobacter is Enterobacter cloacae; the Morganella species is Morganella morganii; said providencia species is providencia stuartii; and the species of Mycobacterium is Mycobacterium tuberculosis.

A compound, pharmaceutically acceptable salt thereof, or composition for use according to item 63, wherein the enterococcus faecium, enterococcus faecalis, staphylococcus aureus, klebsiella pneumoniae, acinetobacter baumannii, pseudomonas aeruginosa, enterobacter cloacae, morganella, and/or providencia stuartii expresses at least one aminoglycoside-modifying enzyme.

The compound, pharmaceutically acceptable salt or composition thereof for use according to item 63a, wherein the at least one aminoglycoside modifying enzyme is selected from the group consisting of aminoglycoside phosphotransferases, in particular, APH (3 ') IIIa, APH (3') IIa, APH (3 ') Ia, APH (3 ") Ib, APH (3') VI, APH (6) Id, APH (3 ') VIa, APH (3') IIb and APH (6) Ic; aminoglycoside acetyltransferases, in particular AAC (6') Ib, AAC (3) III, AAC (3) IV, AAC (3) Ia, AAC (3) IId and AAC (3) Ic, more in particular AAC (3) III and AAC (3) IV; aminoglycoside nucleotidyl transferases, in particular ANT (3 ') Ia, ANT (2 ') Ia and ANT (3 '); and bifunctional aminoglycoside modifying enzymes, particularly AAC (6 ') Ie-APH (2') Ia; and any combination thereof.

64. A compound, pharmaceutically acceptable salt or composition thereof for use according to any one of items 56 to 63b, wherein the bacterial infection is caused at least in part by a bacterial species that exhibits resistance to at least one member of an existing regulatory-approved family of antibiotics, in particular against bacterial species showing resistance to at least one member of the antibiotic family selected from the group consisting of penicillins, cephalosporins, carbapenems, monobactams, polymyxins, rifamycins, lipiarins, quinolones, sulfonamides, macrolides, lincosamides, tetracyclines, aminoglycosides, lipopeptides, glycylcyclines, glycopeptides, oxazolidinones, and lipiarines.

65. A compound, pharmaceutically acceptable salt or composition thereof for use according to any one of items 56 to 64, wherein the bacterial infection is selected from the list consisting of respiratory tract infections, concurrent skin and soft tissue infections, concurrent abdominal cavity infections, community-acquired pneumonia, hospital-acquired pneumonia, ventilator-associated pneumonia, urinary tract infections, bacterial meningitis, infectious endocarditis, septicemia, osteomyelitis, septic arthritis, septicemia, anthrax, osteomyelitis, tuberculosis, leprosy, necrotizing fasciitis, scarlet fever, rheumatic fever, postpartum fever, and streptococcal toxic shock syndrome, and one or more of infections and infectious diseases, and further nosocomial infections, such as infections caused by the use of intravascular catheters.

In the remainder of the application, all general radicals R, unless otherwise stated, are1、R2、R3、R4、R5、R6X, W, Y, Z, m, n, p, q, r, etc. are as defined for the moiety of formula (I), and whether or not these groups are further defined and/or limited with respect to their broadest, most general definition in the structural moiety of formula (I)The conditions specified for the structural part of formula (I) apply to all structures, formulae and embodiments herein.

Hereinafter, when it is mentioned that the group/substituent is "as defined for formula (I)," as defined for a compound of formula (I) "etc., for example" R1、R4、R5、R6As defined for compounds of formula (I)', this is to be understood as meaning all possible definitions of the corresponding groups/substituents as described herein for formula (I), including, for each individual group/substituent, the broadest definition of said group as disclosed herein as well as any narrower definition and any variation of the particular group/substituent "preferred", "more preferred", "most preferred", "in particular", etc. Furthermore, for compounds of formula (I), any particular example or list of groups or substituents stated herein as "preferred", "more preferred", etc., is also to be understood as "preferred", "more preferred", etc., for any compound and formula listed herein that falls within the broadest scope of formula (I).

In the following, when a formula or a particular compound is depicted in such a way that the full valency of an atom, e.g. a heteroatom, does not appear to be satisfied, it is understood that the valency of the atom is satisfied by the presence of an undepicted hydrogen atom, i.e. the depicted species is not a group, cation, anion, carbene, etc., but is a neutral species. By way of example, the following structure/scenario is presented:

Structural part described asShould be understood as respectively representing

Further, in some instances herein, illustrations of specific structural portions given, which are also understood by the skilled artisan, are used, but are distinct from one another. These should be understood to represent moieties that one of skill in the art would understand when analyzing the moieties alone, rather than by analogy with the textOther descriptions of the same parts presented herein are compared. For example, a methyl group can be represented by the formula "CH" as explicitly written3"or" Me "or can be expressed using standard" linear "formulas as are well understood by those skilled in the art. Thus, both of the following structures include methyl groups:

in the form of "linear

The compounds of the invention (compounds of formula (I)) are described herein in the form of a 3-membered ring of the aminoglycoside core drawn in a chair conformation. Thus, the various substituents attached to the ring are described as being either equatorial or orthosteric, the description showing the respective stereochemistry (if any) of a given stereocenter. To avoid any confusion in this regard, for example where the bond angles of the flat or upright substituents are not repeated with 100% precision (e.g., due to constraints within the drawing components used), the following explanation of the structures set forth herein is provided:

The compounds of the present invention are represented by the structure of formula (I). For clarity, the conventional numbering of the various atoms of the 3 six-membered rings of the aminoglycoside core is shown above (with this numbering being used here). As shown in the above structures and as shown in all other structures herein, NHR of 1 bit regardless of the fidelity of the reproduction of true key angle4The groups are equatorial. As shown in the above structure and as shown in all other structures herein, the NH of 3 bits regardless of the reproduction accuracy of the true key angle2The groups are equatorial. As shown in the above structures and as shown in all other structures herein, the O atom at the 4-position is equatorial regardless of the fidelity of the true bond angle. As shown in the above structures and as shown in all other structures herein, the F atom at the 5-position is upright regardless of the fidelity of the reproduction of the true bond angle. As shown in the above structures and as shown in all other structures herein, whether or notThe reproduction accuracy of the true bond angle is how well the 6-bit O atom is flat. As shown in the above structures and as shown in all other structures herein, the O atom at the 1' position is straight regardless of the accuracy of the reproduction of the true bond angle. As shown in the above structure and as shown in all other structures herein, R of 2' bit regardless of the reproduction accuracy of the true key angle 6The groups are equatorial. As shown in the above structure and as shown in all other structures herein, R of the 3' bit regardless of the reproduction accuracy of the true key angle2The groups are equatorial. As shown in the above structure and as shown in all other structures herein, R of the 3' bit regardless of the reproduction accuracy of the true key angle3The radicals are upright. As shown in the above structures and as shown in all other structures herein, the OH group at the 4' position is equatorial regardless of the fidelity of the true bond angle. As shown in the above structure and as shown in all other structures herein, the CH of the 5' bit regardless of the reproduction accuracy of the true key angle2NHR1The groups are equatorial. As shown in the above structures and as shown in all other structures herein, the O atom at the 1 "position is straight regardless of the fidelity of the reproduction of the true bond angle. As shown in the above structures and as shown in all other structures herein, the OH group at the 2 "position is equatorial regardless of the fidelity of the true bond angle. As shown in the above structures and as shown in all other structures herein, the guanidino group at the 3 "position is equatorial, regardless of the fidelity of reproduction of the true bond angle. As shown in the above structures and as shown in all other structures herein, the OH group at the 4 "position is equatorial regardless of the fidelity of the true bond angle. As shown in the above structure and as shown in all other structures herein, the CH at the 5 "position regardless of the reproduction accuracy of the true key angle 2The OH groups are equatorial.

Thus, the above description of the compounds of formula (I), as is customary in the art of organic chemistry, is to be understood as meaning the following structures (two conventional, slightly different ways of representing the same structure and stereochemistry) which exhibit the following stereochemistry:

Detailed Description

In its most general form, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof,

wherein the radical R1、R2、R3、R4、R5And R6Each as defined herein above.

Thus, in the compounds of the invention, R1Selected from H, methyl, ethyl, straight or branched C3-6Alkyl radical, C3-6A cycloalkyl group, a,

Wherein is a group represented by the formula (I) and R in the formula (I)1The point of attachment of the attached N atom, and wherein m, N, p, Q, r and Q are as defined above for the compound of formula (I). R1Preferably selected from the group consisting of H, methyl, ethyl, straight or branched C3-6Alkyl radical, C3-6A cycloalkyl group, a, The group consisting of.

R1More preferably selected from the group consisting of H,The group consisting of;

wherein independently for each moiety, at said R1In moieties, q is 1, 2 or 3, most preferably 1.

In the compounds of the invention, R1Most preferably H.

In the compounds of the invention, R2Selected from the group consisting of H, methyl, -CH 2F、-CF3Ethyl, n-propyl, isopropyl, cyclopropyl, halogen, hydroxy, -OCH3、-OEt、-OCH2F、-OCF3、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2、-NHCH2F、-NHCF3and-NHQ, and;

wherein when R is2Is ethyl, n-propyl, isopropyl, cyclopropyl, -OEt, -NHEt or-N (Et)2When R is in the above-mentioned range2The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OEt、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2and-NHQ, substituted with a substituent group; with the proviso that when R2is-OEt, -NHEt or-N (Et)2When the-OEt, -NHEt or-N (Et)2Et group of group linked to the-OEt, -NHEt or-N (Et)2The carbon atoms of the O or N atoms of the group may be substituted only by one or more substituents independently selected from halogen.

In said R2In which Q is as defined above for the compounds of formula (I).

The expression "wherein R is2Is ethyl, n-propyl, isopropyl, cyclopropyl, -OEt, -NHEt, or-N (Et)2When R is in the above-mentioned range2The alkyl and cycloalkyl moieties in the radical may optionally be substituted by … … "is understood to mean ethyl per se, n-propyl per se, cyclopropyl per se, the ethyl moiety of the-OEt group, the ethyl moiety of the-NHEt group, and-N (Et)2The ethyl portion of the group may be optionally substituted with the substituents listed below.

The expression "provided that when R is2is-OEt, -NHEt or-N (Et)2When the-OEt, -NHEt or-N (Et)2Et group of group linked to the-OEt, -NHEt or-N (Et)2The carbon atoms of the O or N atoms of the group may be substituted only by one or more substituents independently selected from halogen "is understood to mean that with R2As an example, when R is ═ OEt2when-OEt, the underlined carbon atom-OCCH2CH3(converting the-OEt, -NHEt or-N (Et))2Et group of group linked to the-OEt, -NHEt or-N (Et)2Carbon atoms of the O or N atoms of the group) may be substituted only with one or more substituents independently selected from halogen, and the non-underlined carbon atoms (-CH for the OEt substituent)3Carbon atom of a group) may be substituted by any of the foregoing permissible substituents, i.e. it may optionally be substituted by one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OEt、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2and-NHQ, or a substituent of the group consisting of.

In the compounds of the invention, R2Preferably selected from the group consisting of H, methyl, -CH2F、-CF3Ethyl, n-propyl, isopropyl, cyclopropyl, halogen, hydroxy, -OCH3、-OEt、-OCH2F、-OCF3、-NH2、-NHCH3、-NHEt、-N(CH3)2、-N(Et)2、-NHCH2F、-NHCF3and-NHQ. In the compounds of the invention, R2More preferably selected from the group consisting of H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, -F, hydroxy, -OCH 3、-OEt、-OCH2F. and-OCF3Group (b), group (b). In the compounds of the invention, R2More preferably selected from the group consisting of H, methyl, and hydroxy; most preferred is H or methyl. R2May be a hydroxyl group. R2May be H. R2May be a methyl group.

In the compounds of the invention, R3Selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -NH2、-NHCH3、-NHCH2F、-NHCF3、-NHEt、-NHC3-8Alkyl, -N (CH)3)2、-N(Et)2、-N(C3-8Alkyl radical)2and-NHQ, and;

wherein when R is3is-OEt, -OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -NHEt, -NHC3-8Alkyl, -N (Et)2or-N (C)3-8Alkyl radical)2When R is in the above-mentioned range3The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; with the proviso that each of said R3The carbon atoms of the group directly bonded to the O or N atoms may be substituted only with one or more substituents independently selected from halogen; and is

Wherein when R is3is-OCH3or-NHCH3When it is in the-OCH3or-NHCH3CH of a radical3Phenyl optionally substituted in part; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S.

In said R3In which Q is as defined above for the compounds of formula (I).

The expression "wherein R is3is-OEt, -OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -NHEt, -NHC3-8Alkyl, -N (Et)2or-N (C)3-8Alkyl radical)2When R is in the above-mentioned range3The alkyl and cycloalkyl moieties in a group may optionally be substituted by … … "is understood to mean, only the ethyl moiety of the-OEt group, the OC3-8C of alkyl3-8Alkyl moieties and the like may be optionally substituted with substituents listed subsequently. In other words, the O atom of the-OEt group may for example not be substituted by the subsequently listed groups (such substitution is not technically meaningful, since when O has only a valence of 2, the O atom will be bonded to 3 different groups), but the Et moiety (alkyl moiety) of the-OEt group may be further substituted as defined.

Furthermore, the expression "provided that each of said R3Directly bonded to each of said R in the radical3The carbon atoms of the O or N atoms in the radical may be substituted only by one or more substituents independently selected from halogen "is understood to mean that R is3By way of example, -OEt, where R3is-OEt, underlined carbon atom-OCH2CH3(i.e., the carbon atom linking the respective Et group to the O or N atom) may be substituted only with one or more substituents independently selected from halogen, and the non-underlined carbon atom (-CH for the OEt substituent) 3Carbon atoms of the group) may be substituted with any of the foregoing permissible substituents, i.e., it may be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, or a substituent of the group consisting of. Mention of "each of said R3Directly bonded to each of said R in the radical3The carbon atom of the O or N atom in the radical "is understood to mean that in the list R3Carbon atoms explicitly listed in the radical, i.e. at R3Within the radical itself (e.g. -underlined carbon atoms of the OEt substituent-OCH2CH3) Instead of R in the structure of formula (I)3To an aminoglycoside ring carbon atom (not in said R)3Carbon atoms in the group).

As mentioned above, when in the compounds of the present inventionR3is-OCH3or-NHCH3When it is in the-OCH3or-NHCH3CH of a radical3Phenyl optionally substituted in part; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S. This is understood to mean, for example, when R 3is-OCH3When said-OCH is3CH of a radical3Moieties may be substituted with any of the listed substituents, thereby yielding compounds wherein R is3is-OCH2-a group of substituents. at-OCH3In the case where the radical is substituted, for example, by optionally substituted phenyl, R is obtained3The group will be-OCH2(optionally substituted phenyl). An example thereof is-OCH2Ph and-OCH2- (4-fluorophenyl).

In the compounds of the invention, R3Preferably selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, and-OCH2C3-6A cycloalkyl group, a group consisting of,

wherein when R is3is-OEt, -OC3-8Alkyl, -OC3-6Cycloalkyl, or-OCH2C3-6When cycloalkyl is present, said R3The alkyl and cycloalkyl moieties in the group may optionally be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; provided that it is directly bonded to each of said R3Each said R on the O atom in the moiety3The carbon atoms in the group may be substituted only with one or more substituents independently selected from halogen; and is

Wherein when R is3is-OCH3When it is in the-OCH3CH of a radical3Phenyl, part of which is optionally substituted; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S. Q is as defined above for the compound of formula (I).

In the compounds of the invention, R3More preferably selected from the group consisting of H, halogen, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; and-OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

In the compounds of the invention, R3More preferably selected from the group consisting of H, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, in particular selected from the group consisting of H and hydroxy. Thus, in particularly preferred compounds, R3May be H. Thus, in a further particularly preferred compound, R3May be a hydroxyl group.

In a particularly preferred series of compounds of the invention, or pharmaceutically acceptable salts thereof, R2Is H or methyl, R3Selected from the group consisting of H, hydroxy, -OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH 2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S); -OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S); and R is1、R4、R5And R6As defined herein for compounds of formula (I) (which is the broadest definition disclosed herein for the group recited or any narrower definition disclosed for the group recited, including the definition "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

In the compounds of the invention, R4Selected from the group consisting of H, methyl, ethyl, -CH2F、-CF3Straight or branched chain C3-6Alkyl, substituted straight chain C2-6Alkyl, substituted branched C3-6Alkyl, optionally substituted C3-6Cycloalkyl, optionally substituted-CH2C3-6Cycloalkyl, formyl, optionally substituted phenyl, optionally substituted 5-or 6-membered heteroaryl,

Wherein is a group represented by the formula (I) and R in the formula (I)4A point of attachment of the N atom attached thereto, and wherein

When R is 4Is a substituted straight chain C2-6Alkyl, substituted branched C3-6Alkyl, substituted C3-6Cycloalkyl radicalsOr substituted-CH2C3-6When cycloalkyl, it is substituted with one or more groups independently selected from halogen, hydroxy, -OCH3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, substituted with a substituent group; provided that in the structure of formula (I) it is directly bonded to R4Said R on the attached N atom4The carbon atoms of the group may be substituted with only one or more substituents independently selected from halogen.

In said R4In which r and Q are as defined above for the compounds of formula (I).

In this connection, the expression "provided that the bond to R is direct in the structure of the formula (I)4Said R on the attached N atom4The carbon atoms of the group may be substituted only by one or more substituents independently selected from halogen "is understood to mean that with R4Substituted ethyl, wherein R is4Is a substituted ethyl group, the underlined carbon atom-CH2CH3(i.e., the attachment of the-Et group to R in formula (I))4Carbon atom of the N atom) may be substituted only with one or more substituents independently selected from halogen, rather than the underlined carbon atoms (-CH in the Et substituent)3Of a group) may be substituted with any of the foregoing permissible substituents, i.e., it may be substituted with one or more substituents independently selected from the group consisting of halogen, hydroxy, -OCH 3、-OC2-4Alkyl, -NH2、-NHCH3、-NHC2-4Alkyl, -N (CH)3)2、-N(C2-4Alkyl radical)2and-NHQ, or a substituent of the group consisting of.

In the compounds of the invention, R4Preferably selected from the group consisting of H, Wherein in the R4In part, r and Q are as defined above for compounds of formula (I).

In the compounds of the invention, R4More preferably selected from the group consisting of H,The group consisting of.

Thus, R4May be H.

Thus, in particularly preferred compounds of the invention, R4(may be)

Thus, in a further particularly preferred compound of the invention, R4(may be)

Thus, in a further particularly preferred compound of the invention, R4(may be)

Thus, in a further particularly preferred compound of the invention, R4(may be)

In the compounds of the invention, R5Is selected from the group consisting of H; a methyl group; -CH2F;-CF3(ii) a An ethyl group; straight or branched C3-8An alkyl group; substituted straight chain C2-8An alkyl group; substituted branched C3-8An alkyl group; optionally substituted C3-6A cycloalkyl group; optionally substituted-CH2C3-6A cycloalkyl group; optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; -C (═ NH) NH 2;-C(=NR7)NH2;-C(=NH)NHR8;-C(=NR7)NHR8;-C(=NR7)NR8R9(ii) a And

-X-Z, wherein

X is selected from methylene, ethylene, linear or branched C3-8Alkylene, the group consisting of; each of which, in addition to being linked to Z, may optionally be further substituted by one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3Ethyl, straight or branched C3-6Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR10、-NR10R11、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR12、-CONR12R13、-NHCOCH3、-NHCOC2-6Alkyl, -NR14COCH3and-NR15COC2-6Alkyl, substituted with a substituent of the group consisting of; provided that it is linked to R in the structure of formula (I)5The atoms of the X group on the attached N atom cannot be directly attached to another O or N atom;

and is

Z is selected from optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted C3-6Cycloalkyl, the group consisting of;

wherein R is7To R15Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C 3-6Cycloalkyl groups, or a combination thereof.

In this respect, the expression "provided that it is linked to R in the structure of formula (I)5Is connected withThe atoms of said X group on the attached N atom not being able to be directly linked to another O or N atom "is understood to mean, for example, that X ═ ethylene is taken as an example (the X group may be methylene, ethylene, or a linear or branched C group3-8Alkylene) in which X is a substituted ethylene group, the underlined carbon atoms of the ethylene group-CH2CH2-Z (i.e. linking it to R in the structure of formula (I))5The atom of the X group on the attached N atom) cannot be substituted by a group that would yield the underlined carbon atom directly attached to the O or N atom of the substituent. In other words, the underlined carbon atoms may be linked to only N or O atoms which are linked to R in the structure of formula (I)5The attached N atom. This condition is not suitable for-CH2CH2The carbon atoms of the-Z group which are not underlined.

In the compounds of the invention, R5Is a substituted straight chain C2-8Alkyl, substituted branched C3-8Alkyl, substituted C3-6Cycloalkyl, or substituted-CH2C3-6Cycloalkyl, which may be substituted with any substituent. However, in this context, R5Preferably by one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3Ethyl, straight or branched C3-6Alkyl, halogen, -OH, -OCH 3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR10、-NR10R11、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR7、-CONR12R13、-NHCOCH3、-NHCOC2-6Alkyl, -NR14COCH3and-NR15COC2-6Alkyl, substituted with a substituent of the group consisting of; wherein R is7To R15Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl, and C3-6Cycloalkyl groups, or a combination thereof. In this context, provided that said C2-8Alkyl, substituted branched C3-8Alkyl, substituted C3-6Cycloalkyl, or substituted-CH2C3-6In cycloalkyl radicals with R in the structure of formula (I)5The atom to which the attached N atom is attached is preferably not directly attached to another O or N atom.

In the compounds of the invention, when R is5Optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; the heterocycloalkyl group may preferably be selected from the group consisting of optionally substituted azetidinyl, optionally substituted diazetidinyl, optionally substituted oxetanyl, optionally substituted thietanyl, optionally substituted pyrrolidinyl, optionally substituted tetrahydrofuranyl, optionally substituted tetrahydrothienyl, optionally substituted piperidinyl, optionally substituted tetrahydropyranyl, optionally substituted thiaalkyl, optionally substituted imidazolidinyl, optionally substituted pyrazolidinyl, optionally substituted oxazolidinyl, optionally substituted isoxazolidinyl, optionally substituted thiazolidinyl, optionally substituted isothiazolidinyl, optionally substituted dioxolanyl, optionally substituted dithialanyl, optionally substituted piperazinyl, optionally substituted morpholinyl, optionally substituted thiomorpholinyl, optionally substituted 1, 3-dioxanyl, optionally substituted 1, 4-dioxanyl, optionally substituted 1, 3-dithianyl, and optionally substituted 1, 4-dithianyl; wherein optional substituents are as defined elsewhere herein for said heterocycloalkyl. In this context, the optionally substituted 4-membered non-heteroaromatic alkyl group containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; more preferably, the compound is selected from the group consisting of 4-piperidyl, 3-piperidyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 4-oxazolidinyl, 2-morpholinyl, 3-morpholinyl, 4-morpholinyl, 2-tetrahydrofuryl, and 3-tetrahydrofuryl.

In the compounds of the present invention, wherein R5Or Z is optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted means optionally substituted with one or more of any substituent. In this context, optionally substituted preferably means optionally substituted by one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; wherein R is16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl radical, C3-6Cycloalkyl groups, or a combination thereof. In this context, with the proviso that when one of said substituents is directly attached to the N atom in the corresponding non-aromatic heterocycloalkyl moiety, it preferably applies that it may be selected from the group consisting of only methyl, -CH 2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b). In this context, with the proviso that when one of the substituents is directly attached to the N atom in the corresponding heteroaryl moiety, it preferably applies that it may be selected from the group consisting of only methyl, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b).

In the compounds of the invention, R5Preferably selected from the group consisting of H, methyl, ethyl, straight or branched C3-8Alkyl, substituted straight chain C2-8Alkyl, substituted branched C3-8An alkyl group; optionally substituted C3-6A cycloalkyl group, a,

Wherein is a group represented by the formula (I) and R in the formula (I)5The point of attachment of the N atom to which it is attached, and

wherein A is selected from the group consisting of optionally substituted phenyl, optionally substituted 5-membered heteroaryl, and optionally substituted 6-membered heteroaryl, optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; and is

Wherein at said R 5In the moiety, m, n, q, p and R10As defined herein for compounds of formula (I) (which is the broadest definition disclosed herein for the group recited or any narrower definition disclosed for the group recited, including the definition "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

In this context, wherein a is optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted means optionally substituted with one or more of any substituent. In this context, optionally substituted preferably means optionally substituted by one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; wherein R is 16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl radical, C3-6Cycloalkyl groups, or a combination thereof.

In this context, with the proviso that when one of said substituents is directly attached to the N atom in the corresponding non-aromatic heterocycloalkyl moiety, it preferably applies that it may be selected from the group consisting of only methyl, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b). In this context, with the proviso that when one of the substituents is directly attached to the N atom in the corresponding heteroaryl moiety, it preferably applies that it may be selected from the group consisting of only methyl, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b).

In the compounds of the invention, R5More preferably selected from the group consisting of H, methyl,Ethyl, straight or branched C3-6Alkyl, aryl, heteroaryl, and heteroaryl,

Wherein in the R5In moiety, Q and R10As defined herein for compounds of formula (I) (which is the broadest definition disclosed herein for the group recited or any narrower definition disclosed for the group recited, including the definition "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

In the compounds of the invention, R5Most preferably selected from the group consisting of H, The group consisting of.

In the present invention, R6Selected from the group consisting of OH and NH2The group consisting of. In preferred compounds, R6Is OH. In other preferred compounds, R6Is NH2

The compound of formula (I) or a pharmaceutically acceptable salt thereof may preferably be selected from compounds of formulae (Ia) to (If)

Wherein, independently of each otherFor each of the compounds of formulae (Ia) to (If), R1、R4、R5And R6As defined for compounds of formula (I) (which is the broadest definition disclosed herein for that group or any narrower definition disclosed for that group, including the terms "preferred", "more preferred", "most preferred", etc. or words of effect defined) and wherein for each of the compounds of formulae (Ie) to (If), W is independently selected from the group consisting of-OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S); and-OCH 2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

Particularly preferred in this context are compounds of formulae (Ia) to (If), wherein R1Is H. Further preferred compounds in this context are compounds of formulae (Ia) to (If) R6Is OH. Thus, particularly preferred compounds of the formulae (Ia) to (If) are those in which R is1Is H and R6Is OH. Further preferred compounds in this context are compounds of formulae (Ia) to (If) R6Is NH2In (1). Thus, particularly preferred compounds of the formulae (Ia) to (If) are those in which R is1Is H and R6Is NH2

Further preferred in this context are compounds of formulae (Ia) to (If), wherein R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Ia) to (If) are those in which R is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Ia) to (If) are those in which R is1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references relating to compounds of formulae (Ia) to (If), particular preference is given to compounds of formula (Ic). Thus, even more particularly preferred are compounds of formula (Ic) wherein R is 1Is H, especially wherein R1Is H, R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of formula (Ic) wherein R is1Is H, especially wherein R1Is H, R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In the above-mentioned context relating to compounds of formulae (Ia) to (If), further particular preference is given to compounds of formulae (Ia) to (If) in which R is1Is: -CH2CH2OH (wherein R represents in each of formulae (Ia) to (If)5The point of attachment of the attached N atom). Further preferred compounds in this context are compounds of formulae (Ia) to (If) R6Is OH. Thus, particularly preferred compounds of the formulae (Ia) to (If) are those in which R is1Is: -CH2CH2OH and R6Is OH. Further preferred compounds in this context are compounds of formulae (Ia) to (If) R6Is NH2In (1). Thus, particularly preferred compounds of the formulae (Ia) to (If) are those in which R is1Is: -CH2CH2OH and R6Is NH2

Further preferred in this context are compounds of formulae (Ia) to (If), wherein R is 4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Ia) to (If) are those in which R is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Ia) to (If) are those in which R is1Is: -CH2CH2OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references relating to compounds of formulae (Ia) to (If), particular preference is given to compounds of formula (Ic). Thus, even more particularly preferred are compounds of formula (Ic) wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of formula (Ic) wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H, The group consisting of.

The compound of formula (I) or a pharmaceutically acceptable salt thereof may even more preferably be selected from the group consisting of compounds of formulae (Ig) to (Ip)

Wherein, independently, for each of the compounds of formulae (Ig) to (Ip), R1、R4And R5Such as for compounds of formula (I)(iii) as defined (which is the broadest definition disclosed herein for that group or any narrower definition disclosed for that group, including the terms "preferred", "more preferred", "most preferred", etc. or words which achieve that effect, as defined), and wherein for each of the compounds of formulae (In) to (Ip), W is independently selected from the group consisting of-OCH3、-OCH2F、-OCF3、-OEt、-OC3-8Alkyl, -OC3-6Cycloalkyl, -OCH2C3-6Cycloalkyl, -OCH2(optionally substituted phenyl), -OCH2(optionally substituted 5-membered heteroaryl), -OCH2(optionally substituted 6-membered heteroaryl), -OCH2(optionally substituted non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S); -OCH2(optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S); and-OCH2(optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S).

Particularly preferred in this context are compounds of formulae (Ig) to (Ip) wherein R is 1Is H.

Further preferred in this context are compounds of formulae (Ig) to (Ip), wherein R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Ip) to (Ig) are those in which R is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Ig) to (Ip) are those in which R is1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Of all the above mentioned references to compounds of formulae (Ig) to (Ip), particular preference is given to compounds of formula (Ii). Thus, even more particularly preferred are compounds of formula (Ii), wherein R is1Is H, especially wherein R1Is H, and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In the above-mentioned context relating to compounds of formulae (Ig) to (Ip), further especially preferred are compounds of formulae (Ig) to (Ip), wherein R1 is-CH2CH2OH (wherein R represents in each of formulae (Ig) to (Ip)5The point of attachment of the attached N atom).

Further preferred in this context are compounds of formulae (Ig) to (Ip), wherein R is 4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Ip) to (Ig) are those in which R is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Ig) to (Ip) are those in which R is1Is: -CH2CH2OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Of all the above mentioned references to compounds of formulae (Ig) to (Ip), particular preference is given to compounds of formula (Ii). Thus, even more particularly preferred are compounds of formula (Ii), wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH, and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In all the above statements relating to compounds of the formulae (Ia) to (If) and (Ig) to (Ip), the preferred compounds and R listed for the compounds of the formulae (Ia) to (If) and (Ig) to (Ip) are included1、R4And/or R6Preferred combination of (1), R5Preferably selected from the group consisting of H, methyl, ethyl, straight or branched C3-8Alkyl, substituted straight chain C2-8Alkyl, substituted branched C 2-8An alkyl group; optionally substituted C3-6A cycloalkyl group, a, Wherein is a group represented by the formula (I) and R in the formula (I)5Linked N atomA point of attachment of the son, and

wherein A is selected from the group consisting of optionally substituted phenyl, optionally substituted 5-membered heteroaryl, and optionally substituted 6-membered heteroaryl, optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; and is

Wherein at said R5In the moiety, m, n, q, p and R10As defined herein for compounds of formula (I) (which is the broadest definition disclosed herein for the group recited or any narrower definition disclosed for the group recited, including the definition "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

In this context, wherein a is optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted means optionally substituted with one or more of any substituent. In this context, optionally substituted preferably means optionally substituted by one or more groups independently selected from the group consisting of methyl, -CH 2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; wherein R is16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl radical, C3-6Cycloalkyl groups, or a combination thereof. In this context, with the proviso that when one of said substituents is directly attached to the N atom in the corresponding non-aromatic heterocycloalkyl moiety, it preferably applies that it may be selected from the group consisting of only methyl, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b). In this context, with the proviso that when one of the substituents is directly attached to the N atom in the corresponding heteroaryl moiety, it preferably applies that it may be selected from the group consisting of only methyl, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b).

All of the above mentioned are related to the formulae (Ia) to (If) and (Ig) to (I)p) of the compounds of the formulae (Ia) to (If) and (Ig) to (Ip), preferably the compounds listed and R1、R4And/or R6Preferred combination of (1), R 5More preferably selected from the group consisting of H, methyl, ethyl, straight or branched C3-6Alkyl, aryl, heteroaryl, and heteroaryl, Wherein in the R5In moiety, Q and R10As defined herein for compounds of formula (I) (which is the broadest definition disclosed herein for the group recited or any narrower definition disclosed for the group recited, including the definition "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

In all the above mentioned cases involving compounds of formulae (Ia) to (If) and (Ig) to (Ip), R is included for compounds of formulae (Ia) to (If) and (Ig) to (Ip)1、R4And/or R6Preferred combination of (1), R5Most preferably selected from the group consisting of H, The group consisting of.

Other preferred compounds of the invention are compounds of formula (Iq)

Wherein R is1、R2、R3、R4And R6As defined above for compounds of formula (I) (which is the broadest definition as disclosed herein for such group or any narrower definition as disclosed for such group, including the definitions "preferred", "more preferred", "most preferred", etc. or words which achieve that effect). Thus, particularly preferred compounds in this context include compounds of formulae (Iq-a) to (Iq-f)

Wherein R is1、R4And R6As defined above for compounds of formula (I) (which is the broadest definition as disclosed herein for such group or any narrower definition as disclosed for such group, including the definitions "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

Particularly preferred in this context are compounds of formulae (Iq-a) to (Iq-f), wherein R1Is H. Further preferred compounds in this context are those of formulae (Iq-a) to (Iq-f) wherein R6Is OH. Thus, particularly preferred compounds of the formulae (Iq-a) to (Iq-f) are those in which R is1Is H and R6Is OH. Further preferred compounds in this context are those of formulae (Iq-a) to (Iq-f) wherein R6Is NH2In (1). Thus, particularly preferred compounds of the formulae (Iq-a) to (Iq-f) are those in which R is1Is H and R6Is NH2

Further preferred in this context are compounds of formulae (Iq-a) to (Iq-f), wherein R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Iq-a) to (Iq-f) are those in which R is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Iq-a) to (Iq-f) are those in which R is1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references relating to compounds of formulae (Iq-a) to (Iq-f), particular preference is given to compounds of formula (Iq-c). Thus, even more particularly preferred are compounds of formula (Iq-c), wherein R is 1Is H, especially wherein R1Is H, R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of formula (Iq-c), wherein R is1Is H, especially wherein R1Is H, R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In the above-mentioned context relating to compounds of formulae (Iq-a) to (Iq-f), further particular preference is given to compounds of formulae (Iq-a) to (Iq-f), wherein R is1Is: -CH2CH2OH (wherein R represents in each of formulae (Iq-a) to (Iq-f)5The point of attachment of the attached N atom). Further preferred compounds in this context are those of formulae (Iq-a) to (Iq-f) wherein R6Is OH. Thus, particularly preferred compounds of the formulae (Iq-a) to (Iq-f) are those in which R is1Is: -CH2CH2OH and R6Is OH. Further preferred compounds in this context are those of formulae (Iq-a) to (Iq-f) wherein R6Is NH2In (1). Thus, particularly preferred compounds of the formulae (Iq-a) to (Iq-f) are those in which R is1Is: -CH2CH2OH and R6Is NH2

Further preferred in this context are compounds of formulae (Iq-a) to (Iq-f), wherein R 4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Iq-a) to (Iq-f) are those in which R is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Iq-a) to (Iq-f) are those in which R is1Is: -CH2CH2OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references relating to compounds of formulae (Iq-a) to (Iq-f), particular preference is given to compounds of formula (Iq-c). Thus, even more particularly preferred are compounds of formula (Iq-c), wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of formula (Iq-c), wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H, The group consisting of.

Other preferred compounds of the invention are compounds of formula (Ir)

Wherein R is1、R2、R3、R4And R6As hereinbefore defined for compounds of formula (I) (which is the broadest definition as disclosed herein for the group or for the group)Any narrower definition disclosed, including "preferred," "more preferred," "most preferred," etc., or words to achieve the same, as defined). Thus, particularly preferred compounds in this context include compounds of formulae (Ir-a) to (Ir-f)

Wherein R is1、R4And R6As defined above for compounds of formula (I) (which is the broadest definition as disclosed herein for such group or any narrower definition as disclosed for such group, including the definitions "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

Particularly preferred in this context are compounds of formulae (Ir-a) to (Ir-f), wherein R1Is H. Further preferred compounds in this context are compounds of formulae (Ir-a) to (Ir-f) R6Is OH. Thus, particularly preferred compounds of the formulae (Ir-a) to (Ir-f) are those in which R is1Is H and R6Is OH. Further preferred compounds in this context are compounds of formulae (Ir-a) to (Ir-f) R 6Is NH2In (1). Thus, particularly preferred compounds of the formulae (Ir-a) to (Ir-f) are those in which R is1Is H and R6Is NH2

Further preferred in this context are compounds of formulae (Ir-a) to (Ir-f), wherein R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Ir-a) to (Ir-f) are those in which R4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Ir-a) to (Ir-f) are those in which R is1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references to compounds of the formulae (Ir-a) to (Ir-f), particular preference is given to compounds of the formula (Ir-c). Thus, even more particularly preferred are compounds of formula (Ir-c), wherein R is1Is H, especially wherein R1Is H, R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of the formula (Ir-c), wherein R 1Is H, especially wherein R1Is H, R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In the above-mentioned context relating to compounds of the formulae (Ir-a) to (Ir-f), further particular preference is given to compounds of the formulae (Ir-a) to (Ir-f), in which R is1Is: -CH2CH2OH (wherein R represents in each of formulae (Ir-a) to (Ir-f)5The point of attachment of the attached N atom). Further preferred compounds in this context are compounds of formulae (Ir-a) to (Ir-f) R6Is OH. Thus, particularly preferred compounds of the formulae (Ir-a) to (Ir-f) are those in which R is1Is: -CH2CH2OH and R6Is OH. Further preferred compounds in this context are compounds of formulae (Ir-a) to (Ir-f) R6Is NH2In (1). Thus, particularly preferred compounds of the formulae (Ir-a) to (Ir-f) are those in which R is1Is: -CH2CH2OH and R6Is NH2

Further preferred in this context are compounds of formulae (Ir-a) to (Ir-f), wherein R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Ir-a) to (Ir-f) are those in which R4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Ir-a) to (Ir-f) are those in which R is 1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references to compounds of the formulae (Ir-a) to (Ir-f), particular preference is given to compounds of the formula (Ir-c). Thus, even more particularly preferred are compounds of formula (Ir-c), wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of the formula (Ir-c), wherein R1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Other preferred compounds of the invention are compounds of formula (Is)

Wherein R is1、R2、R3、R4And R6As defined above for compounds of formula (I) (which is the broadest definition as disclosed herein for the group or any narrower definition as disclosed for the group, including "preferred", "more preferred" as defined Or the like, or the word achieving the effect). Thus, particularly preferred compounds in this context include compounds of formulae (Is-a) to (Is-f)

Wherein R is1、R4And R6As defined above for compounds of formula (I) (which is the broadest definition as disclosed herein for such group or any narrower definition as disclosed for such group, including the definitions "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

Particularly preferred in this context are compounds of formulae (Is-a) to (Is-f), wherein R Is1Is H. Further preferred compounds in this context are compounds of formulae (Is-a) to (Is-f) wherein R Is6Is OH. Thus, particularly preferred compounds of the formulae (Is-a) to (Is-f) are those in which R Is1Is H and R6Is OH. Further preferred compounds in this context are compounds of formulae (Is-a) to (Is-f) wherein R Is6Is NH2In (1). Thus, particularly preferred compounds of the formulae (Is-a) to (Is-f) are those in which R Is1Is H and R6Is NH2

Further preferred in this context are compounds of formulae (Is-a) to (Is-f), wherein R Is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Is-a) to (Is-f) are those in which R Is 4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Is-a) to (Is-f) are those in which R Is1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references to compounds of formulae (Is-a) to (Is-f), particular preference Is given to compounds of formula (Is-c). Thus, even more particularly preferred are compounds of formula (Is-c), wherein R Is1Is H, especially wherein R1Is H, R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of the formula (Is-c), wherein R Is1Is H, especially wherein R1Is H, R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In the above-mentioned context relating to compounds of the formulae (Is-a) to (Is-f), further particular preference Is given to compounds of the formulae (Is-a) to (Is-f), in which R Is 1Is: -CH2CH2OH (wherein: (Is-a) to (Is-f)Each of R5The point of attachment of the attached N atom). Further preferred compounds in this context are compounds of formulae (Is-a) to (Is-f) wherein R Is6Is OH. Thus, particularly preferred compounds of the formulae (Is-a) to (Is-f) are those in which R Is1Is: -CH2CH2OH and R6Is OH. Further preferred compounds in this context are compounds of formulae (Is-a) to (Is-f) wherein R Is6Is NH2In (1). Thus, particularly preferred compounds of the formulae (Is-a) to (Is-f) are those in which R Is1Is: -CH2CH2OH and R6Is NH2

Further preferred in this context are compounds of formulae (Is-a) to (Is-f), wherein R Is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Is-a) to (Is-f) are those in which R Is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (Is-a) to (Is-f) are those in which R Is1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references to compounds of formulae (Is-a) to (Is-f), particular preference Is given to compounds of formula (Is-c). Thus, even more particularly preferred are compounds of formula (Is-c), wherein R Is 1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of the formula (Is-c), wherein R Is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is NH2And R is4Is selected from H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Other preferred compounds of the invention are compounds of formula (It)

Wherein R is1、R2、R3、R4And R6As defined above for compounds of formula (I) (which is the broadest definition as disclosed herein for such group or any narrower definition as disclosed for such group, including the definitions "preferred", "more preferred", "most preferred", etc. or words which achieve that effect). Thus, particularly preferred compounds in this context include compounds of formulae (It-a) to (It-f)

Wherein R is1、R4And R6As defined above for compounds of formula (I) (which is the broadest definition as disclosed herein for such group or any narrower definition as disclosed for such group, including the definitions "preferred", "more preferred", "most preferred", etc. or words which achieve that effect).

Particularly preferred in this context are compounds of formulae (It-a) to (It-f), wherein R1Is H. Further preferred compounds in this context are compounds of formulae (It-a) to (It-f) wherein R is6Is OH. Thus, particularly preferred compounds of the formulae (It-a) to (It-f) are those in which R is1Is H and R6Is OH. Further preferred compounds in this context are compounds of formulae (It-a) to (It-f) wherein R is6Is NH2In (1). Thus, particularly preferred compounds of the formulae (It-a) to (It-f) are those in which R is1Is H and R6Is NH2

Further preferred in this context are compounds of formulae (It-a) to (It-f), wherein R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (It-a) to (It-f) are those in which R4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (It-a) to (It-f) are those in which R is1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references relating to compounds of formulae (It-a) to (It-f), particular preference is given to compounds of formula (It-c). Thus, even more particularly preferred are compounds of formula (It-c), wherein R is 1Is H, especially wherein R1Is H, R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of formula (It-c), wherein R is1Is H, especially wherein R1Is H, R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In the above-mentioned context relating to compounds of formulae (It-a) to (It-f), further especially preferred are compounds of formulae (It-a) to (It-f), wherein R1Is: -CH2CH2OH (wherein R represents in each of formulae (It-a) to (It-f)5The point of attachment of the attached N atom). Further preferred compounds in this context are compounds of formulae (It-a) to (It-f) wherein R is6Is OH. Thus, particularly preferred compounds of the formulae (It-a) to (It-f) are those in which R is1Is: -CH2CH2OH andR6is OH. Further preferred compounds in this context are compounds of formulae (It-a) to (It-f) wherein R is6Is NH2In (1). Thus, particularly preferred compounds of the formulae (It-a) to (It-f) are those in which R is1Is: -CH2CH2OH and R6Is NH2

Further preferred in this context are compounds of formulae (It-a) to (It-f), wherein R 4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (It-a) to (It-f) are those in which R4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of the formulae (It-a) to (It-f) are those in which R is1Is: -CH2CH2OH and R4Is selected from H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references relating to compounds of formulae (It-a) to (It-f), particular preference is given to compounds of formula (It-c). Thus, even more particularly preferred are compounds of formula (It-c), wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of formula (It-c), wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is NH2And R is4Is selected from H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H, The group consisting of.

Other preferred compounds of the invention are compounds of formula (Iu)

Wherein R is1、R2、R3、R4And R6As defined above for compounds of formula (I) (which is the broadest definition as disclosed herein for such group or any narrower definition as disclosed for such group, including the definitions "preferred", "more preferred", "most preferred", etc. or words which achieve that effect). Thus, particularly preferred compounds in this context include compounds of formulae (Iu-a) to (Iu-f)

Wherein R is1、R4And R6As hereinbefore defined for compounds of formula (I) (said definitions are for groups as defined herein)The broadest definition disclosed for a group or any narrower definition disclosed for that group, including the definitions "preferred," "more preferred," "most preferred," etc., or words which achieve that effect).

Particularly preferred in this context are compounds of formulae (Iu-a) to (Iu-f), wherein R1Is H. Further preferred compounds in this context are compounds of formulae (Iu-a) to (Iu-f) R6Is OH. Thus, particularly preferred compounds of formulae (Iu-a) to (Iu-f) are those in which R is1Is H and R6Is OH. Further preferred compounds in this context are compounds of formulae (Iu-a) to (Iu-f) R 6Is NH2In (1). Thus, particularly preferred compounds of formulae (Iu-a) to (Iu-f) are those in which R is1Is H and R6Is NH2

Further preferred in this context are compounds of formulae (Iu-a) to (Iu-f), wherein R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Iu-a) to (Iu-f) are those in which R is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of formulae (Iu-a) to (Iu-f) are those in which R is1Is H and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references to compounds of formulae (Iu-a) to (Iu-f), particularly preferred are compounds of formula (Iu-c). Thus, even more particularly preferred are compounds of formula (Iu-c), wherein R is1Is H, especially wherein R1Is H, R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of formula (Iu-c), wherein R is 1Is H, especially wherein R1Is H, R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In the above-mentioned context relating to compounds of formulae (Iu-a) to (Iu-f), further especially preferred are compounds of formulae (Iu-a) to (Iu-f), wherein R is1Is: -CH2CH2OH (wherein R represents in each of formulae (Iu-a) to (Iu-f)5The point of attachment of the attached N atom). Further preferred compounds in this context are compounds of formulae (Iu-a) to (Iu-f) R6Is OH. Thus, particularly preferred compounds of formulae (Iu-a) to (Iu-f) are those in which R is1Is: -CH2CH2OH and R6Is OH. Further preferred compounds in this context are compounds of formulae (Iu-a) to (Iu-f) R6Is NH2In (1). Thus, particularly preferred compounds of formulae (Iu-a) to (Iu-f) are those in which R is1Is: -CH2CH2OH and R6Is NH2

Further preferred in this context are compounds of formulae (Iu-a) to (Iu-f), wherein R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I). Particularly preferred compounds of the formulae (Iu-a) to (Iu-f) are those in which R is4Is selected from the group consisting of H,The group consisting of.

Thus, particularly preferred compounds of formulae (Iu-a) to (Iu-f) are those in which R is 1Is: -CH2CH2OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

In this context, R6May be OH. In this context, R6May be NH2

Of all the above mentioned references to compounds of formulae (Iu-a) to (Iu-f), particularly preferred are compounds of formula (Iu-c). Thus, even more particularly preferred are compounds of formula (Iu-c), wherein R is1Is: -CH3CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is OH and R4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

Thus, further even more particularly preferred are compounds of formula (Iu-c), wherein R is1Is: -CH2CH2OH, especially wherein R1Is: -CH2CH2OH、R6Is NH2And R is4Is selected from the group consisting of H,Wherein in the R4In part, r and Q are as defined for compounds of formula (I), most preferably selected from the group consisting of H,The group consisting of.

The compounds of the invention may be selected from the group consisting of:

the group consisting of.

In the compounds of the present invention, one of the moieties or groups may be or contain (or be referred to herein as) optionally substituted phenyl; optionally substituted 5-membered heteroaryl; optionally substituted 6-membered heteroaryl; optionally substituted 4-membered non-aromatic heterocycloalkyl containing 1 heteroatom selected from O, N and S; optionally substituted 5-membered non-aromatic heterocycloalkyl containing 1 or 2 heteroatoms selected from O, N and S; or optionally substituted 6-membered non-aromatic heterocycloalkyl containing 1, 2 or 3 heteroatoms selected from O, N and S; optionally substituted, unless otherwise specified, means optionally substituted with any substituent. Preferably, the optional substitution is Means optionally substituted with one or more groups independently selected from the group consisting of methyl, -CH2F、-CF3O, ═ NH, ═ S, ethyl, straight or branched C3-8Alkyl, halogen, -OH, -OCH3、-OCH2F、-OCF3、-OC2-6Alkyl, -NH2、-NHQ、-NHR16、-NR17R18、-CO2H、-CO2CH3、-CO2C2-6Alkyl, -OCOCH3、-OCOC2-6Alkyl, -CN, -CONHR19、-CONR20R21、-NHCOCH3、-NHCOC2-6Alkyl, -NR22COCH3、-NR23COC2-6Alkyl, -C (═ NH) R24、-C(=NR25)R26、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of; wherein R is16To R38Each of which is independently selected from the group consisting of H, methyl, ethyl, C3-6Alkyl radical, C3-6Cycloalkyl, the group consisting of; with the proviso that when one of said substituents is directly attached to the N atom in the respective non-aromatic heterocycloalkyl moiety, it may be selected only from the group consisting of the free methyl radicals, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Group (b), group (b). Furthermore, phenyl, 5-membered heteroaryl and substituted 6-membered heteroaryl cannot be substituted by ═ O, ═ NH, ═ S, or if desired with said phenyl, 5-membered heteroarylAny other substituent where the atom of a group or substituted 6-membered heteroaryl forms a double bond will result in the ring no longer being aromatic.

The expression "provided that when one of said substituents is directly linked to the N atom in the corresponding non-aromatic heterocycloalkyl moiety, it can be selected only from the group consisting of the radicals methyl, -CH2F、-CF3Ethyl, straight or branched C 3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38The meaning of the group "consisting of, will now be indicated by R3=-OCH2(4-piperidine) is explained as an example. The piperidine ring is optionally substituted, and in this context the N atom of the piperidine ring may be substituted only by methyl, -CH2F、-CF3Ethyl, straight or branched C3-8Alkyl, -CO2CH3、-CO2C2-6Alkyl, -CN, -CONHR19、-CONR20R21、-C(=NH)R24、-C(=N-OH)R27、-C(=N-OR28)R29、-C(=NH)NH2、-C(=NR30)NH2、-C(=NH)NHR31、-C(=NH)NR32R33、-C(=NR34)NR35R36and-C (═ NR)37)NHR38Substituted with a substituent of the group consisting of. Thus, substitution on the N atom of the piperidine does not result in a heteroatom-heteroatom bond. Conversely, this condition does not apply to the carbon atom of the piperidine ring, which may be substituted by any of the substituents listed.

When a "straight" bond (a stereochemical "wedge" (defined as a single stereochemical) or a "tortuous" (racemic) bond, relative to a bond between atoms in which at least one represents a stereocenter, is used in this disclosure as a standard in the art to describe a single isomer or racemate, respectively, it is to be understood in the context of this application that it is a direct and unambiguous disclosure of all stereoisomers falling within, and such description is also standard in the art to represent all possible isomers and is used herein only for the purpose of imparting the greatest conciseness to the disclosure.

Therefore, the temperature of the molten metal is controlled,(examples of moieties having two stereocenters from which a "straight" bond connects an atom of a stereocenter to an adjacent atom) is to be understood as each being directly and explicitly disclosedThat is to say that the first and second electrodes,

therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,is to be understood as directly and unequivocally disclosingEach one of

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,should be understood as direct andeach one is explicitly disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,is to be understood as disclosing each and every one of the

Therefore, the temperature of the molten metal is controlled,is to be understood as disclosing each and every one of the

Therefore, the temperature of the molten metal is controlled,is to be understood as disclosing each and every one of the

Therefore, the temperature of the molten metal is controlled,is to be understood as disclosing each and every one of the

Therefore, the temperature of the molten metal is controlled,is to be understood as disclosing each and every one of the

Therefore, the temperature of the molten metal is controlled,is to be understood as disclosing each and every one of the

Therefore, the temperature of the molten metal is controlled,is to be understood as disclosing each and every one of the

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

Therefore, the temperature of the molten metal is controlled,it is to be understood that each of the foregoing is directly and specifically disclosed

If any structure is described in this application in such a way as to use a "straight" bond at the stereocenter, but not explicitly listed above, it is to be understood that the explicit rules and teachings outlined above with respect to the direct and explicit disclosure of each of the corresponding stereoisomers falling within the structure also apply to such structures.

In addition, the first and second substrates are,it is to be understood that each of the foregoing is directly and specifically disclosed

In addition, the first and second substrates are,it is to be understood that each of the foregoing is directly and specifically disclosed

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,are to be understood as being directly and unequivocally disclosedEach one is provided with

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,it is to be understood that each of the foregoing is directly and specifically disclosed

In addition, the first and second substrates are,it is to be understood that each of the foregoing is directly and specifically disclosed

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

In addition, the first and second substrates are,is to be understood as disclosing each and every one of the

As used throughout this document, unless otherwise indicated, "alkyl" refers to a hydrocarbon chain that may be linear (straight chain) or branched. As used herein, C2-4Alkyl means a list of groups including or consisting of ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl and tert-butyl. As used herein, C2-6Alkyl means including or consisting of ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butylA list of groups consisting of yl, t-butyl, n-pentyl, 1-dimethylpropyl, 1-methylbutyl, 1-ethylpropyl, 2-dimethylpropyl, 3-methylbutyl, 1, 2-dimethylpropyl, 2-methylbutyl, n-hexyl, 2-methylpentyl, 3-methylpentyl, 2-dimethylbutyl, and 2, 3-dimethylbutyl. As used herein, C 1-6Alkyl means including or consisting of methyl and above in C2-6List of groups consisting of each of those listed in the definition of alkyl. As used herein, straight chain C2-6Alkyl means the list of groups consisting of ethyl, n-propyl, n-butyl, n-pentyl, and n-hexyl. As used herein, straight chain C3-6Alkyl refers to the list of groups consisting of n-propyl, n-butyl, n-pentyl, and n-hexyl. As used herein, straight chain C3-8Alkyl refers to the list of groups consisting of n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, and n-octyl. As used herein, straight chain C2-8Alkyl means the list of groups consisting of ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, and n-octyl. As used herein, straight chain C3-8Alkylene means a list of groups consisting of n-propylene, n-butylene, n-pentylene, n-hexylene, n-heptylene, and n-octylene.

As used herein, branched C3-6Alkyl means a list of groups including or consisting of isopropyl, isobutyl, sec-butyl, tert-butyl, 1-dimethylpropyl, 1-methylbutyl, 1-ethylpropyl, 2-dimethylpropyl, 3-methylbutyl, 1, 2-dimethylpropyl, 2-methylbutyl, 2-methylpentyl, 3-methylpentyl, 2-dimethylbutyl, and 2, 3-dimethylbutyl.

As used herein, branched C3-8Alkyl means including or consisting of isopropyl, isobutyl, sec-butyl, tert-butyl, 1-dimethylpropyl, 1-methylbutyl, 1-ethylpropyl, 2-dimethylpropyl, 3-methylbutyl, 1, 2-dimethylpropyl, 2-methylbutyl, 2-methylpentyl, 3-methylpentyl, 2-dimethylbutyl, 2, 3-dimethylbutyl, 2-methylhexyl, 3-methylhexyl, 2-dimethylpentyl, 2, 3-dimethylpentyl, 2, 4-dimethylpropylA list of groups consisting of pentyl, 3, 3-dimethylpentyl, 3-ethylpentyl, 2, 3-trimethylbutyl, 2-methylheptyl, 3-methylheptyl, 4-methylheptyl, 3-ethylhexyl, 2-dimethylhexyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 3, 3-dimethylhexyl, 3, 4-dimethylhexyl, 3-ethyl-2-methylpentyl, 3-trimethylpentyl, 2, 3-trimethylpentyl, and 2,3, 3-trimethylpentyl.

As used herein, C3-8Alkyl means including or consisting of the above in straight chain C3-8The definitions of alkyl groups being given above for the branched chain C3-8List of groups consisting of both those listed in the definition of alkyl.

As used herein, branched C3-8Alkylene means including or consisting of isopropylene, sec-butylene, tert-butylene, 1-dimethylpropylene, 1-methylbutylene, 1-ethylpropylene, 2-dimethylpropylene, 3-methylbutylene, 1, 2-dimethylpropylene, 2-methylbutylene, 2-methylpentylene, 3-methylpentylene, 2-dimethylbutylene, 2, 3-dimethylbutylene, 2-methylhexylene, 3-methylhexylene, 2-dimethylpentylene, 2, 3-dimethylpentylene, 2, 4-dimethylpentylene, 3-dimethylpentylene, 3-ethylpentylene, 2, 3-trimethylbutylene, 2-methylheptylene, 2-methylpentylene, 1-dimethylpropylene, 1-methylbutylene, 1-ethylpentylene, 2-methylpentylene, 3-methylpentylene, 2-methylheptylylene, 2-methylpentylene, 3-methylpentylene, 2-methylpentylene, or mixtures of any of the same type, A list of groups consisting of 3-methylheptylene, 4-ethylhexyl, 2-dimethylhexylene, 2, 3-dimethylhexylene, 2, 4-dimethylhexylene, 2, 5-dimethylhexylene, 3-dimethylhexylene, 3, 4-dimethylhexylene, 3-dimethylpentylene, 3, 4-dimethylpentylene, 3-trimethylpentylene, 2, 3-ethylpentylene, 2, 3-trimethylpentylene, and 2, 3-trimethylpentylene.

As used throughout this document, unless otherwise specified, "cycloalkyl" refers to a ring whose ring-forming atoms are themselves only carbon atoms. As used herein, C3-6Cycloalkyl refers to a list of groups consisting of cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.

As used throughout this document, the term "halogen" refers to F, Cl, Br, or I. As used herein, the term "halogen" preferably refers to F, Cl or Br, most preferably F.

As used herein throughout, the term "heteroaryl" or "heteroaromatic" refers to an aryl ring system having one to four heteroatoms (e.g., O, S, N, or a combination thereof) as ring atoms in a heteroaromatic ring system, wherein the remaining atoms forming the ring system are carbon atoms. Heteroaryl rings or groups may consist of a single ring or fused ring system. Typical fused heteroaromatic ring systems are 9 or 10 membered ring systems containing one to four heteroatoms selected from oxygen, sulphur and nitrogen. In this sense, a typical mono-heteroaromatic ring is a 5 to 6 membered ring containing one to four, preferably one to three heteroatoms selected from oxygen, sulphur and nitrogen. Examples of 5-membered heteroaryl groups include pyrrolyl, furanyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, furazanyl, oxadiazolyl, thiadiazolyl, dithiazolyl, and tetrazolyl. Examples of 6-membered heteroaryl groups include pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, and tetrazinyl. As used throughout, a 5-membered heteroaryl group can include pyrrolyl (e.g., 1-, 2-, 3-, 4-, or 5-pyrrolyl), furyl (e.g., 2-, 3-, 4-, or 5-furyl), thienyl (e.g., 2-, 3-, 4-, or 5-thienyl), imidazolyl (e.g., 1-, 2-, 4-, or 5-imidazolyl), pyrazolyl (e.g., 1-, 3-, 4-, or 5-pyrazolyl), oxazolyl (e.g., 2-, 4-, or 5-oxazolyl), isoxazolyl (e.g., 3-, 4-, or 5-isoxazolyl), thiazolyl (e.g., 2-, 4-, or 5-thiazolyl), isothiazolyl (e.g., 3-,), 4-, or 5-isothiazolyl), triazolyl (e.g., 4-, or 5- (1H-1,2, 3-triazolyl) -yl; 4-, or 5- (2H-1,2, 3-triazole) -yl; 3-, or 5- (1H-1,2, 4-triazole) -yl; 3-, or 5- (4H-1,2, 4-triazole) -yl, furazanyl (e.g., 3-, or 4-furazanyl), oxadiazolyl (e.g., 3-, or 4- (1,2, 5-oxadiazolyl) -yl; 3-, or 5- (1,2, 4-oxadiazolyl) -yl; 4-, or 5- (1,2, 3-oxadiazolyl) -yl; 2-, or 5- (1,3, 4-oxadiazolyl) -yl), thiadiazolyl (e.g., 3-, or 4- (1,2, 5-thiadiazolyl) -yl; 3-, or 5- (1,2, 4-thiadiazolyl) -yl; 4-, or 5- (1,2, 3-thiadiazolyl) -yl; 2- Or 5- (1,3, 4-thiadiazole) group), and tetrazolyl (e.g., 1-, or 5- (1H-tetrazole) -group; 1-, or 4- (2H-tetrazol) -yl). As used throughout, a 6-membered heteroaryl group can include a pyridyl group (e.g., 2-, 3-, 4-, 5-, or 6-pyridyl), a pyrazinyl group (e.g., 2-, 3-, 5-, or 6-pyrazinyl), a pyrimidinyl group (e.g., 2-, 4-, 5-, or 6-pyrimidinyl), a pyridazinyl group (e.g., 3-, 4-, 5-, or 6-pyridazinyl), a triazinyl group (e.g., 4-, or 5-, or 6- (1,2, 3-triazine) -yl; 3-, or 5-or 6- (1,2, 4-triazine) -yl; 2-, or 4-, or 6- (1,3, 5-triazine) -yl).

As used throughout this document, a 4, 5 or 6 membered non-aromatic heterocycloalkyl or a partial or non-aromatic heterocyclic ring is understood to mean a 4, 5 or 6 membered cycloaliphatic ring in which 1, 2 or 3 ring carbon atoms have been independently replaced by O, N or S atoms. The resulting ring is non-aromatic. For example, the carbon atom in the cyclohexane of the cycloaliphatic compound is replaced with a nitrogen atom to produce the piperidine, a non-aromatic heterocyclic compound, or the carbon atom in the cyclopentyl of the cycloaliphatic group is replaced with a nitrogen atom to produce a 5-membered non-aromatic heterocycloalkyl (containing 1 heteroatom) pyrrolidinyl group.

As used throughout, optionally substituted 4-membered non-aromatic heterocycloalkyl may include optionally substituted azetidinyl (e.g., optionally substituted 1-azetidinyl, 2-azetidinyl, 3-azetidinyl, or 4-azetidinyl), optionally substituted 1, 3-diazetinyl (e.g., optionally substituted 1-diazetinyl, 2-diazetinyl, 3-diazetinyl, or 4-diazetinyl), optionally substituted oxetanyl (e.g., optionally substituted 2-oxetanyl, 3-oxetanyl, or 4-oxetanyl), and optionally substituted thietanyl (e.g., optionally substituted 2-thietanyl, 3-thietanyl, 4-oxetanyl), and optionally substituted thietanyl (e.g., optionally substituted 2-thietanyl, 2-azetidinyl, 4-azetidinyl, Or 4-thietanyl).

As used throughout, optionally substituted 5-membered non-heteroaromatic alkyl groups may include optionally substituted pyrrolidinyl (e.g., optionally substituted 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 4-pyrrolidinyl, or 5-pyrrolidinyl), optionally substituted pyrazolidinyl (e.g., optionally substituted 1-pyrazolidinyl, 2-pyrazolidinyl, 3-pyrazolidinyl, 4-pyrazolidinyl, or 5-pyrazolidinyl), optionally substituted imidazolidinyl (e.g., optionally substituted 1-imidazolidinyl, 2-imidazolidinyl, 3-imidazolidinyl, 4-imidazolidinyl, or 5-imidazolidinyl), optionally substituted tetrahydrofuranyl (e.g., optionally substituted 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 4-tetrahydrofuranyl, pyrazolidinyl, and pyrazolidinyl groups, Or 5-tetrahydrofuryl), optionally substituted 1, 3-dioxolanyl (e.g. optionally substituted 1, 3-dioxolan-2-yl, 1, 3-dioxolan-4-yl, or 1, 3-dioxolan-5-yl), optionally substituted tetrahydrothienyl (e.g. optionally substituted 2-tetrahydrothienyl, 3-tetrahydrothienyl, 4-tetrahydrothienyl, or 5-tetrahydrothienyl), optionally substituted 1, 3-oxazolidinyl (e.g. optionally substituted 2-oxazolidinyl, 4-oxazolidinyl, or 5-oxazolidinyl), optionally substituted 1, 5-isoxazolidinyl (e.g. optionally substituted 3-isoxazolidinyl, 4-isoxazolidinyl, 5-isoxazolidinyl, or 5-isoxazolidinyl), Or 5-isoxazolidinyl), optionally substituted 1, 2-oxathiolanyl (e.g., optionally substituted 1, 2-oxathiolan-3-yl, 1, 2-oxathiolan-4-yl, or 1, 2-oxathiolan-5-yl), optionally substituted 1, 3-oxathiolanyl (e.g., optionally substituted 1, 3-oxathiolan-2-yl, 1, 3-oxathiolan-4-yl, or 1, 3-oxathiolan-5-yl), optionally substituted thiazolidinyl (e.g., optionally substituted 2-thiazolidinyl, 3-thiazolidinyl, 4-thiazolidinyl, or 5-thiazolidinyl), and optionally substituted isothiazolinyl (e.g., optionally substituted 2-isothiazolidinyl, 5-thiazolidinyl), 3-isothiazolidinyl, 4-isothiazolidinyl, or 5-isothiazolidinyl), optionally substituted 1, 3-dithiolanyl (e.g., optionally substituted 3-dithiolanyl, 4-dithiolanyl, or 5-dithiolanyl).

As used throughout, an optionally substituted 6-membered non-heteroaromatic alkyl group can include an optionally substituted piperidinyl group (e.g., an optionally substituted 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 5-piperidinyl, or 6-piperidinyl group), an optionally substituted tetrahydropyranyl group (e.g., an optionally substituted 2-tetrahydropyranyl, 3-tetrahydropyranyl, 4-tetrahydropyranyl, 5-tetrahydropyranyl, or 6-tetrahydropyranyl group), an optionally substituted thiaalkyl group (e.g., an optionally substituted 2-thiaalkyl, 3-thiaalkyl, 4-thiaalkyl, 5-thiaalkyl, or 6-thiaalkyl group), an optionally substituted piperazinyl group (e.g., an optionally substituted 1-piperazinyl, 2-piperazinyl, 3-piperazinyl, 4-piperazinyl, 5-piperazinyl, or 6-piperazinyl), optionally substituted morpholinyl (e.g., optionally substituted 2-morpholinyl, 3-morpholinyl, 4-morpholinyl, 5-morpholinyl, or 6-morpholinyl), optionally substituted thiomorpholinyl (e.g., optionally substituted 2-thiomorpholinyl, 3-thiomorpholinyl, 4-thiomorpholinyl, 5-thiomorpholinyl, or 6-thiomorpholinyl), optionally substituted 1, 3-dioxanyl (e.g., 1, 3-dioxan-2 yl, 1, 3-dioxan-4 yl, 1, 3-dioxan-5-yl, or 1, 3-dioxan-6-yl), optionally substituted 1, 4-dioxane (e.g., optionally substituted 1, 4-dioxan-2-yl, or 1, 2-dioxan-3-yl), optionally substituted 1, 3-dithianyl (e.g., optionally substituted 1, 3-dithian-2-yl, 1, 3-dithian-4-yl, 1, 3-dithian-5-yl, or 1, 3-dithian-6-yl), optionally substituted 1, 4-dithianyl (e.g., optionally substituted 1, 4-dithian-2-yl, or 1, 2-dithian-3-yl).

As used throughout this document, "aryl" preferably refers to an unsaturated aromatic carbocyclic group having 6 to 14 carbon atoms, having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthracenyl). Specific aryl groups include phenyl, biphenyl, naphthyl, and the like.

Examples of bicyclic fused heteroaryl ring systems include indolyl (e.g., 1-, 2-, 3-, 4-, 5-, 6-, or 7-indolyl), benzofuranyl (e.g., 2-, 3-, 4-, 5-, 6-, or 7-benzofuranyl), indazolyl (e.g., 1-, 3-, 4-, 5-, 6-, or 7-indazolyl), oxindolyl (e.g., 1-, 3-, 4-, 5-, 6-, or 7-oxindolyl), benzimidazolyl (e.g., 1-, 2-, 4-, 5-, 6-, or 7-benzimidazolyl), benzothienyl (e.g., 2-, 3-, 4-, or, 5-, 6-, or 7-benzothienyl), benzoxazolyl (e.g., 2-, 4-, 5-, 6-, or 7-benzoxazolyl), benzo [ d ] thiazolyl (e.g., 2-, 4-, 5-, 6-, or 7-benzo [ d ] thiazolyl), quinolinyl (e.g., 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolinyl), isoquinolinyl (e.g., 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolinyl), coumarinyl (e.g., 3-, 4-, 5-, 6-, 7-, or 8-coumarinyl), purinyl (e.g., 2-, 6-, or 7-), 8-, or 9-purinyl), 1, 2-naphthyridinyl (e.g., 3-, 4-, 5-, 6-, 7-, 8- (1, 2-naphthyridine) -yl), 1, 3-naphthyridinyl (e.g., 2-, 4-, 5-, 6-, 7-, 8- (1, 3-naphthyridine) -yl), 1, 4-naphthyridinyl (e.g., 2-, 3-, 5-, 6-, 7-, 8- (1, 4-naphthyridine) -yl), 1, 5-naphthyridinyl (e.g., 2-, 3-, 4-, 6-, 7-, 8- (1, 5-naphthyridine) -yl), or a pharmaceutically acceptable salt thereof, 1, 6-naphthyridinyl (e.g., 2-, 3-, 4-, 5-, 7-, 8- (1, 6-naphthyridine) -yl), 1, 7-naphthyridinyl (e.g., 2-, 3-, 4-, 5-, 7-, 8- (1, 7-naphthyridine) -yl), 1, 8-naphthyridinyl (e.g., 2,3-, 4-, 5-, 6-, 7- (1, 8-naphthyridine) -yl), 2, 3-naphthyridinyl (e.g., 1-, 4-, 5-, 6-, 7-, 8- (2, 3-naphthyridine) -yl), 2, 6-naphthyridinyl (e.g., 1-, 3-, 4-, 5-, 7-, 8- (2, 6-naphthyridine) -yl), and 2, 7-naphthyridine (e.g., 1-, 3-, 4-, 5-, 6-, 8- (2, 7-naphthyridine) -yl).

When reference is made herein to a group or moiety that may be "substituted or unsubstituted", "optionally substituted", "substituted with any substituent(s)", when substituted, they may be substituted with one or more any substituent(s), unless otherwise specified. When reference is made herein to a group or moiety that may be "substituted or unsubstituted", "optionally substituted", "substituted with" or "substituted" by any substituent, it is preferred that when substituted, the introduction of said substituent does not directly result in a carbon atom bearing more than one O, N, or S, atom. It is also preferred that the introduction of the substituent does not directly result in a heteroatom-heteroatom bond (e.g., N-N, N-O, S-N, O-S, O-O, or S-S bond).

Examples of such substituents are those found in the exemplary compounds and embodiments disclosed herein, as well as, for example, halogen (e.g., chlorine, iodine, bromine, or fluorine); c1-Or2-6An alkyl group; is one or more independently selected from OH and NH2、NHCH3、N(CH3)2、NH(C2-6Alkyl group), N (C)2-6Alkyl radical)2、SH、SCH3、SC2-6Alkyl, CN, CONH2、CO2H、NO2、SO2H、SO2CH3And SO2Aryl substituted by substituents C1-Or2-8An alkyl group; c2-8An alkenyl group; c2-8An alkynyl group; a hydroxyl group; c1-Or2-8An alkoxy group; -NH2;-NHCH3;-NHC2-6An alkyl group;-N(CH3)2;-N(C2-6alkyl radical) 2(ii) a Amino (primary, secondary, or tertiary); -NO2;-SH;-SCH3;-SC2-6An alkyl group; -C ═ NH; -C ═ NCH3;-C=NC2-6An alkyl group; -CN; -CONH2;-CONHCH3;-CONHC2-6An alkyl group; -CON (CH)3)2;-CON(C2-6Alkyl radical)2(ii) a A phosphonate ion group; -P (O) (OH)2;-P(O)(OH)(OCH3);-P(O)(OCH3)2;-P(O)(OH)(OC2-6Alkyl groups); -P (O) (OC)2-6Alkyl radical)2;-OP(O)(OH)2;-OP(O)(OH)(OCH3);-OP(O)(OCH3)2;-OP(O)(OH)(OC2-6Alkyl groups); -OP (O) (OC)2-6Alkyl radical)2(ii) a A phosphine; -P (CH)3)2;-P(C2-6Alkyl radical)2;-P(C3-6Cycloalkyl radicals2(ii) a -P (aryl)2(ii) a -P (phenyl)2(ii) a -P (heteroaryl)2(ii) a A carboxyl group; -CO2H; an aminocarbonyl group; -OCONH2;-OCONHCH3;-OCON(CH3)C2-6An alkyl group; -OCON (CH)3)C3-6A cycloalkyl group; -OCON (CH)3) An aryl group; -OCON (CH)3) A phenyl group; -OCON (CH)3) A heteroaryl group; -OCON (CH)3)2;-OCONHC2-6An alkyl group; -OCON (C)2-6Alkyl) C3-6A cycloalkyl group; -OCON (C)2-6Alkyl) aryl; -OCON (C)2-6Alkyl) phenyl; -OCON (C)2-6Alkyl) heteroaryl; -OCON (C)2-6Alkyl radical)2;-OCONHC3-6A cycloalkyl group; -OCON (C)3-6Cycloalkyl) aryl; -OCON (C)3-6Cycloalkyl) phenyl; -OCON (C)3-6Cycloalkyl) heteroaryl; -OCON (C)3-6Cycloalkyl radicals2(ii) a -OCONH aryl; -OCON (aryl) phenyl; -OCON (aryl) heteroaryl; -OCON (aryl)2(ii) a -OCONH phenyl; -OCON (phenyl) heteroaryl; -OCON (phenyl)2(ii) a -an OCONH heteroaryl group; -OCON (heteroaryl)2;-OCON(CH3)2;-OCON(C2-6Alkyl radical)2;-OCONCH3(C2-6Alkyl groups); -NHCOOCH3;-NHCOOC2-6An alkyl group; -NHCOOC3-6A cycloalkyl group; -NHCOO aryl; -NHCOO phenyl; -NHCOO (heteroaryl); -N (CH)3)COOCH3;-N(CH3)COOC2-6An alkyl group; -N (CH)3)COOC3-6A cycloalkyl group; -N (CH)3) COO aryl group; -N (CH)3) COO phenyl; -N (CH)3) COO (heteroaryl); -N (C) 2-6Alkyl) COOCH3;-N(C2-6Alkyl) COOC2-6An alkyl group; -N (C)2-6Alkyl) COOC3-6A cycloalkyl group; -N (C)2-6Alkyl) COO aryl; -N (C)2-6Alkyl) COO phenyl; -N (C)2-6Alkyl) COO (heteroaryl); -N (COOC)3-6Cycloalkyl) COOCH3;-N(COOC3-6Cycloalkyl) COOC2-6An alkyl group; -N (COOC)3-6Cycloalkyl) COOC3-6A cycloalkyl group; -N (COOC)3-6Cycloalkyl) COO aryl; -N (COOC)3-6Cycloalkyl) COO phenyl; -N (COOC)3-6Cycloalkyl) COO (heteroaryl); -N (aryl) COOCH3(ii) a -N (aryl) COOC2-6An alkyl group; -N (aryl) COOC3-6A cycloalkyl group; -N (aryl) cooaryl; -N (aryl) coophenyl; -N (aryl) COO (heteroaryl); -N (phenyl) COOCH3(ii) a -N (phenyl) COOC2-6An alkyl group; -N (phenyl) COOC3-6A cycloalkyl group; -N (phenyl) cooaryl; -N (phenyl) coophenyl; -N (phenyl) COO (heteroaryl); -N (heteroaryl) COOCH3(ii) a -N (heteroaryl) COOC2-6An alkyl group; -N (heteroaryl) COOC3-6A cycloalkyl group; -N (heteroaryl) cooaryl; -N (heteroaryl) coophenyl; -N (heteroaryl) COO (heteroaryl); -NCOOCH3;-N(CH3)COOC2-6An alkyl group; -N (C)2-6Alkyl) COOCH3;-N(C2-6Alkyl) COOC2-6An alkyl group; a carbamate; an acetal; urea; -NHCONH2;-NHCONH(CH3);-NHCON(CH3)2;-NHCONH(C2-6Alkyl groups); -NHCON (C)2-6Alkyl radical)2;-NHCONHC3-6A cycloalkyl group; -NHCON (C)3-6Cycloalkyl radicals2(ii) a -NHCONH aryl; -NHCON (aryl)2(ii) a -NHCONH phenyl; -NHCON (phenyl)2(ii) a -NHCONH heteroaryl; -NHCON (heteroaryl) 2;-NHCON(CH3)(C2-6Alkyl groups); -NHCON (CH)3)(C3-6Cycloalkyl groups); -NHCON (CH)3) (aryl); -NHCON (CH)3) (phenyl); -NHCON (CH)3) (heteroaryl); -NHCON (C)2-6Alkyl) (C3-6Cycloalkyl groups); -NHCON (C)2-6Alkyl) (aryl); -NHCON (C)2-6Alkyl) (phenyl); -NHCON (C)2-6Alkyl) (heteroaryl); -NHCON (C)3-6Cycloalkyl) (aryl); -NHCON (C)3-6Cycloalkyl) (phenyl); -NHCON (C)3-6Cycloalkyl) (heteroaryl); -NHCON (aryl) (phenyl); -NHCON (aryl) (heteroaryl); -NHCON (phenyl) (heteroaryl); -N (CH)3)CONH2;-N(CH3)CONH(CH3);-N(CH3)CON(CH3)2;-N(CH3)CONH(C2-6Alkyl groups); -N (CH)3)CON(C2-6Alkyl radical)2;-N(CH3)CONHC3-6A cycloalkyl group; -N (CH)3)CON(C3-6Cycloalkyl radicals2;-N(CH3) CONH aryl; -N (CH)3) CON (aryl)2;-N(CH3) CONH phenyl; -N (CH)3) CON (phenyl)2;-N(CH3) CONH heteroaryl; -N (CH)3) CON (heteroaryl)2;-N(CH3)CON(CH3)(C2-6Alkyl groups); -N (CH)3)CON(CH3)(C3-6Cycloalkyl groups); -N (CH)3)CON(CH3) (aryl); -N (CH)3)CON(CH3) (phenyl); -N (CH)3)CON(CH3) (heteroaryl); -N (CH)3)CON(C2-6Alkyl) (C3-6Cycloalkyl groups); -N (CH)3)CON(C2-6Alkyl) (aryl); -N (CH)3)CON(C2-6Alkyl) (phenyl); -N (CH)3)CON(C2-6Alkyl) (heteroaryl); -N (CH)3)CON(C3-6Cycloalkyl) (aryl); -N (CH)3)CON(C3-6Cycloalkyl) (phenyl); -N (CH)3)CON(C3-6Cycloalkyl) (heteroaryl); -N (CH)3) CON (aryl) (phenyl); -N (CH)3) CON (aryl) (heteroaryl); -N (CH)3) CON (phenyl) (heteroaryl); -N (C)2-6Alkyl) CONH2;-N(C2-6Alkyl) CONH (CH)3);-N(C2-6Alkyl) CON (CH)3)2;-N(C2-6Alkyl) CONH (C)2-6Alkyl groups); -N (C)2-6Alkyl) CON (C)2-6Alkyl radical)2;-N(C2-6Alkyl) CONHC 3-6A cycloalkyl group; -N (C)2-6Alkyl) CON (C)3-6Cycloalkyl radicals2;-N(C2-6Alkyl) CONH aryl; -N (C)2-6Alkyl) CON (aryl)2;-N(C2-6Alkyl) CONH phenyl; -N (C)2-6Alkyl) CON (phenyl)2;-N(C2-6Alkyl) CONH heteroaryl; -N (C)2-6Alkyl) CON (heteroaryl)2;-N(C2-6Alkyl) CON (CH)3)(C2-6Alkyl groups); -N (C)2-6Alkyl) CON (CH)3)(C3-6Cycloalkyl groups); -N (C)2-6Alkyl) CON (CH)3) (aryl); -N (C)2-6Alkyl) CON (CH)3) (phenyl); -N (C)2-6Alkyl) CON (CH)3) (heteroaryl); -N (C)2-6Alkyl) CON (C)2-6Alkyl) (C3-6Cycloalkyl groups); -N (C)2-6Alkyl) CON (C)2-6Alkyl) (aryl); -N (C)2-6Alkyl) CON (C)2-6Alkyl) (phenyl); -N (C)2-6Alkyl) CON (C)2-6Alkyl) (heteroaryl); -N (C)2-6Alkyl) CON (C)3-6Cycloalkyl) (aryl); -N (C)2-6Alkyl) CON (C)3-6Cycloalkyl) (phenyl); -N (C)2-6Alkyl) CON (C)3-6Cycloalkyl) (heteroaryl); -N (C)2-6Alkyl) CON (aryl) (phenyl); -N (C)2-6Alkyl) CON (aryl) (heteroaryl); -N (C)2-6Alkyl) CON (phenyl) (heteroaryl); -N (C)3-6Cycloalkyl) CONH2;-N(C3-6Cycloalkyl) CONH (CH)3);-N(C3-6Cycloalkyl) CON (CH)3)2;-N(C3-6Cycloalkyl) CONH (C)2-6Alkyl groups); -N (C)3-6Cycloalkyl) CON (C)2-6Alkyl radical)2;-N(C3-6Cycloalkyl) CONHC3-6A cycloalkyl group; -N (C)3-6Cycloalkyl) CON (C)3-6Cycloalkyl radicals2;-N(C3-6Cycloalkyl) CONH aryl; -N (C)3-6Cycloalkyl) CON (aryl)Base)2;-N(C3-6Cycloalkyl) CONH phenyl; -N (C)3-6Cycloalkyl) CON (phenyl)2;-N(C3-6Cycloalkyl) CONH heteroaryl; -N (C)3-6Cycloalkyl) CON (heteroaryl) 2;-N(C3-6Cycloalkyl) CON (CH)3)(C2-6Alkyl groups); -N (C)3-6Cycloalkyl) CON (CH)3)(C3-6Cycloalkyl groups); -N (C)3-6Cycloalkyl) CON (CH)3) (aryl); -N (C)3-6Cycloalkyl) CON (CH)3) (phenyl); -N (C)3-6Cycloalkyl) CON (CH)3) (heteroaryl); -N (C)3-6Cycloalkyl) CON (C)2-6Alkyl) (C3-6Cycloalkyl groups); -N (C)3-6Cycloalkyl) CON (C)2-6Alkyl) (aryl); -N (C)3-6Cycloalkyl) CON (C)2-6Alkyl) (phenyl); -N (C)3-6Cycloalkyl) CON (C)2-6Alkyl) (heteroaryl); -N (C)3-6Cycloalkyl) CON (C)3-6Cycloalkyl) (aryl); -N (C)3-6Cycloalkyl) CON (C)3-6Cycloalkyl) (phenyl); -N (C)3-6Cycloalkyl) CON (C)3-6Cycloalkyl) (heteroaryl); -N (C)3-6Cycloalkyl) CON (aryl) (phenyl); -N (C)3-6Cycloalkyl) CON (aryl) (heteroaryl); -N (C)3-6Cycloalkyl) CON (phenyl) (heteroaryl); -N (aryl) CONH2(ii) a -N (aryl) CONH (CH)3) (ii) a -N (aryl) CON (CH)3)2(ii) a -N (aryl) CONH (C)2-6Alkyl groups); -N (aryl) CON (C)2-6Alkyl radical)2(ii) a -N (aryl) CONHC3-6A cycloalkyl group; -N (aryl) CON (C)3-6Cycloalkyl radicals2(ii) a -N (aryl) CONH aryl; -N (aryl) CON (aryl)2(ii) a -N (aryl) CONH phenyl; -N (aryl) CON (phenyl)2(ii) a -N (aryl) CONH heteroaryl; -N (aryl) CON (heteroaryl)2(ii) a -N (aryl) CON (CH)3)(C2-6Alkyl groups); -N (aryl) CON (CH)3)(C3-6Cycloalkyl groups); -N (aryl) CON (CH)3) (aryl); -N (aryl) CON (CH)3) (phenyl); -N (aryl) CON (CH) 3) (heteroaryl); -N (aryl) CON (C)2-6Alkyl) (C3-6Cycloalkyl groups); -N (aryl) CON (C)2-6Alkyl) (aryl); -N (aryl) CON (C)2-6Alkyl) (benzene)Radical); -N (aryl) CON (C)2-6Alkyl) (heteroaryl); -N (aryl) CON (C)3-6Cycloalkyl) (aryl); -N (aryl) CON (C)3-6Cycloalkyl) (phenyl); -N (aryl) CON (C)3-6Cycloalkyl) (heteroaryl); -N (aryl) CON (aryl) (phenyl); -N (aryl) CON (aryl) (heteroaryl); -N (aryl) CON (phenyl) (heteroaryl); -N (phenyl) CONH2(ii) a -N (phenyl) CONH (CH)3) (ii) a -N (phenyl) CON (CH)3)2(ii) a -N (phenyl) CONH (C)2-6Alkyl groups); -N (phenyl) CON (C)2-6Alkyl radical)2(ii) a -N (phenyl) CONHC3-6A cycloalkyl group; -N (phenyl) CON (C)3-3Cycloalkyl radicals2(ii) a -N (phenyl) CONH aryl; -N (phenyl) CON (aryl)2(ii) a -N (phenyl) CONH phenyl; -N (phenyl) CON (phenyl)2(ii) a -N (phenyl) CONH heteroaryl; -N (phenyl) CON (heteroaryl)2(ii) a -N (phenyl) CON (CH)3)(C2-6Alkyl groups); -N (phenyl) CON (CH)3)(C3-6Cycloalkyl groups); -N (phenyl) CON (CH)3) (aryl); -N (phenyl) CON (CH)3) (phenyl); -N (phenyl) CON (CH)3) (heteroaryl); -N (phenyl) CON (C)2-6Alkyl) (C3-6Cycloalkyl groups); -N (phenyl) CON (C)2-6Alkyl) (aryl); -N (phenyl) CON (C)2-6Alkyl) (phenyl); -N (phenyl) CON (C)2-6Alkyl) (heteroaryl); -N (phenyl) CON (C)3-6Cycloalkyl) (aryl); -N (phenyl) CON (C) 3-6Cycloalkyl) (phenyl); -N (phenyl) CON (C)3-6Cycloalkyl) (heteroaryl); -N (phenyl) CON (aryl) (phenyl); -N (phenyl) CON (aryl) (heteroaryl); -N (phenyl) CON (phenyl) (heteroaryl); -N (heteroaryl) CONH2(ii) a -N (heteroaryl) CONH (CH)3) (ii) a -N (heteroaryl) CON (CH)3)2(ii) a -N (heteroaryl) CONH (C)2-6Alkyl groups); -N (heteroaryl) CON (C)2-6Alkyl radical)2(ii) a -N (heteroaryl) CONHC3-6A cycloalkyl group; -N (heteroaryl) CON (C)3-6Cycloalkyl radicals2(ii) a -N (heteroaryl) CONH aryl; -N (heteroaryl) CON (aryl)2(ii) a -N (heteroaryl) CONH phenyl; -N (heteroaryl) CON (phenyl)2(ii) a -N (heteroaryl) CONH heteroaryl; -N (heteroaryl) CON (heteroaryl)2(ii) a -N (heteroaryl) CON (CH)3)(C2-6Alkyl groups); -N (heteroaryl) CON (CH)3)(C3-6Cycloalkyl groups); -N (heteroaryl) CON (CH)3) (aryl); -N (heteroaryl) CON (CH)3) (phenyl); -N (heteroaryl) CON (CH)3) (heteroaryl); -N (heteroaryl) CON (C)2-6Alkyl) (C3-6Cycloalkyl groups); -N (heteroaryl) CON (C)2-6Alkyl) (aryl); -N (heteroaryl) CON (C)2-6Alkyl) (phenyl); -N (heteroaryl) CON (C)2-6Alkyl) (heteroaryl); -N (heteroaryl) CON (C)3-6Cycloalkyl) (aryl); -N (heteroaryl) CON (C)3-6Cycloalkyl) (phenyl); -N (heteroaryl) CON (C)3-6Cycloalkyl) (heteroaryl); -N (heteroaryl) CON (aryl) (phenyl); -N (heteroaryl) CON (aryl) (heteroaryl); -N (heteroaryl) CON (phenyl) (heteroaryl); -NHCONH (CH) 3);-NHCON(CH3)2;-NHCONH(C2-6Alkyl groups); -NHCON (C)2-6Alkyl radical)2;-NHCON(CH3)(C2-6Alkyl groups); -N (C)2-6Alkyl) CONH2;-N(C2-6Alkyl) CONH (CH)3);-N(C2-6Alkyl) CON (CH)3)2;-N(C2-6Alkyl) CONH (C)2-6Alkyl groups); -N (C)2-6Alkyl) CON (C)2-6Alkyl radical)2;-N(C2-6Alkyl) CON (CH)3)(C2-6Alkyl groups); a thiocarbonyl group; -C (S) CH3;-C(S)C2-6An alkyl group; -C (S) C3-6A cycloalkyl group; -C (S) aryl; -c(s) phenyl; -c(s) heteroaryl; a sulfonyl group; -SO2CH3;-SO2C2-6An alkyl group; -SO2C3-6A cycloalkyl group; -SO2An aryl group; -SO2Ph;-SO2A heteroaryl group; a sulfinyl group; -SOCH3;-SOC2-6An alkyl group; -SOC3-6A cycloalkyl group; -SO aryl; -SOPh; -SO heteroaryl; sulfate radical; -OSO2CH3;-OSO2C2-6An alkyl group; -OSO2C3-6A cycloalkyl group; -OSO2An aryl group; -OSO2A phenyl group; -OSO2A heteroaryl group; a sulfonamide; -SO2NH2;-SO2NHCH3;-SO2NHC2-6An alkyl group; -SO2NHC3-6A cycloalkyl group; -SO2NH aryl; -SO2NHPh;-SO2NH heteroaryl; -SO2N(CH3)2;-SO2N(C2-6Alkyl radical)2;-SO2N(C3-6Cycloalkyl radicals2;-SO2N (aryl)2;-SO2N(Ph)2;-SO2N (heteroaryl)2;-SO2N(CH3)(C2-6Alkyl groups); -SO2N(CH3)(C3-6Cycloalkyl groups); -SO2N(CH3) (aryl); -SO2N(CH3) (phenyl); -SO2N(CH3) (heteroaryl); -SO2N(C2-6Alkyl) (C3-6Cycloalkyl groups); -SO2N(C2-6Alkyl) (aryl); -SO2N(C2-6Alkyl) (phenyl); -SO2N(C2-6Alkyl) (heteroaryl); -SO2N(C3-6Cycloalkyl) (aryl); -SO2N(C3-6Cycloalkyl) (phenyl); -SO2N(C3-6Cycloalkyl) (heteroaryl); -SO2N (aryl) (phenyl); -SO2N (aryl) (heteroaryl); -SO2N (phenyl) (heteroaryl); -NHSO2CH3;-NHSO2C2-6An alkyl group; -NHSO2C3-6A cycloalkyl group; -NHSO2An aryl group; -NHSO2A phenyl group; -NHSO2A heteroaryl group; -N (CH) 3)SO2CH3;-N(CH3)SO2C2-6An alkyl group; -N (CH)3)SO2C3-6A cycloalkyl group; -N (CH)3)SO2An aryl group; -N (CH)3)SO2A phenyl group; -N (CH)3)SO2A heteroaryl group; -N (C)2-6Alkyl) SO2CH3;-N(C2-6Alkyl) SO2C2-6An alkyl group; -N (C)2-6Alkyl) SO2C3-6A cycloalkyl group; -N (C)2-6Alkyl) SO2An aryl group; -N (C)2-6Alkyl) SO2A phenyl group; -N (C)2-6Alkyl) SO2A heteroaryl group; -N (C)2-6Cycloalkyl) SO2CH3;-N(C3-6Cycloalkyl) SO2C2-6An alkyl group; -N (C)3-6Cycloalkyl) SO2C3-6A cycloalkyl group; -N (C)3-6Cycloalkyl) SO2An aryl group; -N (C)3-6Cycloalkyl) SO2A phenyl group; -N (C)3-6Cycloalkyl) SO2A heteroaryl group; -N (aryl) SO2CH3(ii) a -N (aryl) SO2C2-6An alkyl group; -N (aryl) SO2C3-6A cycloalkyl group; -N (aryl) SO2An aryl group; -N (aryl) SO2A phenyl group; -N (aryl) SO2A heteroaryl group; -N (phenyl) SO2CH3(ii) a -N (phenyl) SO2C2-6An alkyl group; -N (phenyl) SO2C3-6A cycloalkyl group; -N (phenyl) SO2An aryl group; -N (phenyl) SO2A phenyl group; -N (phenyl) SO2A heteroaryl group; -N (heteroaryl) SO2CH3(ii) a -N (heteroaryl) SO2C2-6An alkyl group; -N (heteroaryl) SO2C3-6A cycloalkyl group; -N (heteroaryl) SO2An aryl group; -N (heteroaryl) SO2A phenyl group; -N (heteroaryl) SO2A heteroaryl group; an oxime; NOH; NOCH (NOCH)3;=NOC2-6An alkyl group; NOC ═ NOC3-6A cycloalkyl group; NO aryl; NO phenyl; a ═ NO heteroaryl group; -CH ═ NOH; -CH ═ NOCH3;-CH=NOC2-6An alkyl group; -CH ═ NOC3-6A cycloalkyl group; -CH ═ NO aryl; -CH ═ NO phenyl; -CH ═ NO heteroaryl; -C (CH)3)=NOH;-C(CH3)=NOCH3;-C(CH3)=NOC2-6An alkyl group; -C (CH)3)=NOC3-6A cycloalkyl group; -C (CH)3) NO aryl; -C (CH) 3) NO phenyl; -C (CH)3) A ═ NO heteroaryl group; -C (C)2-6Alkyl) ═ NOH; -C (C)2-6Alkyl) ═ NOCH3;-C(C2-6Alkyl) ═ NOC2-6An alkyl group; -C (C)2-6Alkyl) ═ NOC3-6A cycloalkyl group; -C (C)2-6Alkyl) ═ NO aryl; -C (C)2-6Alkyl) ═ NO phenyl; -C (C)2-6Alkyl) ═ NO heteroaryl; -C (C)3-6Cycloalkyl) ═ NOH; -C (C)3-6Cycloalkyl) ═ NOCH3;-C(C3-6Cycloalkyl) ═ NOC2-6An alkyl group; -C (C)3-6Cycloalkyl) ═ NOC3-6A cycloalkyl group; -C (C)3-6Cycloalkyl) ═ NO aryl; -C (C)3-6Cycloalkyl) ═ NO phenyl; -C (C)3-6Cycloalkyl) ═ NO heteroaryl; -C (aryl) ═ NOH; -C (aryl) ═ NOCH3(ii) a -C (aryl) ═ NOC2-6An alkyl group; -C (aryl) ═ NOC3-6A cycloalkyl group; -C (aryl) ═ NO aryl; -C (aryl) ═ NO phenyl; -C (aryl) ═ NO heteroaryl; -C (phenyl) ═ NOH; -C (phenyl) ═ NOCH3(ii) a -C (phenyl) ═ NOC2-6An alkyl group; -C (phenyl) ═ NOC3-6A cycloalkyl group; -C (phenyl) ═ NO aryl; -C (phenyl) ═ NO phenyl; -C (phenyl) ═ NO heteroaryl; -C (heteroaryl) ═ NOH; -C (heteroaryl) ═ NOCH3(ii) a -C (heteroaryl) ═ NOC2-6An alkyl group; -C (heteroaryl) ═ NOC3-6A cycloalkyl group; -C (heteroaryl) ═ NO aryl; -C (heteroaryl) ═ NO phenyl; -C (heteroaryl) ═ NO heteroaryl; -ON ═ CH (CH)3);-ON=CH(C2-6Alkyl groups); -ON ═ CH (C)3-6Cycloalkyl groups); -ON ═ CH (aryl); -ON ═ CH (phenyl); -ON ═ CH (heteroaryl); -ON ═ C (CH) 3)2;-ON=C(CH3)(C2-6Alkyl groups); -ON ═ C (CH)3)(C3-6Cycloalkyl groups); -ON ═ C (CH)3) (aryl); -ON ═ C (CH)3) (phenyl); -ON ═ C (CH)3) (heteroaryl); -ON ═ C (C)2-6Alkyl radical)2;-ON=C(C2-6Alkyl) (C3-6Cycloalkyl groups); -ON ═ C (C)2-6Alkyl) (aryl); -ON ═ C (C)2-6Alkyl) (phenyl); -ON ═ C (C)2-6Alkyl) (heteroaryl); -ON ═ C (C)3-6Cycloalkyl radicals2;-ON=C(C3-6Cycloalkyl) (aryl); -ON ═ C (C)3-6Cycloalkyl) (phenyl); -ON ═ C (C)3-6Cycloalkyl) (heteroaryl); -ON ═ C (aryl)2(ii) a -ON ═ C (aryl) (phenyl); -ON ═ C (aryl) (heteroaryl); -ON ═ C (phenyl)2(ii) a -ON ═ C (phenyl) (heteroaryl); -ON ═ C (heteroaryl)2(ii) a An imine; NH; as NCH3;=NC2-6An alkyl group; NC ═ NC3-6A cycloalkyl group; -N-aryl; -N-phenyl; -N-heteroaryl; -CH ═ NH; -CH ═ NCH3;-CH=NC2-6An alkyl group; -CH ═ NC3-6A cycloalkyl group; -CH ═ N aryl; -CH ═ N phenyl;-CH ═ N heteroaryl; -C (CH)3)=NH;-C(CH3)=NCH3;-C(CH3)=NC2-6An alkyl group; -C (CH)3)=NC3-6A cycloalkyl group; -C (CH)3) -N-aryl; -C (CH)3) -N-phenyl; -C (CH)3) -N-heteroaryl; -C (C)2-6Alkyl) ═ NH; -C (C)2-6Alkyl) ═ NCH3;-C(C2-6Alkyl) ═ NC2-6An alkyl group; -C (C)2-6Alkyl) ═ NC3-6A cycloalkyl group; -C (C)2-6Alkyl) ═ N aryl; -C (C)2-6Alkyl) ═ N phenyl; -C (C)2-6Alkyl) ═ N heteroaryl; -C (C)3-6Cycloalkyl) ═ NH; -C (C)3-6Cycloalkyl) ═ NCH3;-C(C3-6Cycloalkyl) ═ NC2-6An alkyl group; -C (C)3-6Cycloalkyl) ═ NC 3-6A cycloalkyl group; -C (C)3-6Cycloalkyl) ═ N aryl; -C (C)3-6Cycloalkyl) ═ N phenyl; -C (C)3-6Cycloalkyl) ═ N heteroaryl; -C (aryl) ═ NH; -C (aryl) ═ NCH3(ii) a -C (aryl) ═ NC2-6An alkyl group; -C (aryl) ═ NC3-6A cycloalkyl group; -C (aryl) ═ N aryl; -C (aryl) ═ N phenyl; -C (aryl) ═ N heteroaryl; -C (phenyl) ═ NH; -C (phenyl) ═ NCH3(ii) a -C (phenyl) ═ NC2-6An alkyl group; -C (phenyl) ═ NC3-6A cycloalkyl group; -C (phenyl) ═ N aryl; -C (phenyl) ═ N phenyl; -C (phenyl) ═ N heteroaryl; -C (heteroaryl) ═ NH; -C (heteroaryl) ═ NCH3(ii) a -C (heteroaryl) ═ NC2-6An alkyl group; -C (heteroaryl) ═ NC3-6A cycloalkyl group; -C (heteroaryl) ═ N aryl; -C (heteroaryl) ═ N phenyl; -C (heteroaryl) ═ N heteroaryl; -N ═ CH (CH)3);-N=CH(C2-6Alkyl groups); -N ═ CH (C)3-6Cycloalkyl groups); -N ═ CH (aryl); -N ═ CH (phenyl); -N ═ CH (heteroaryl); -N ═ C (CH)3)2;-N=C(CH3)(C2-6Alkyl groups); -N ═ C (CH)3)(C3-6Cycloalkyl groups); -N ═ C (CH)3) (aryl); -N ═ C (CH)3) (phenyl); -N ═ C (CH)3) (heteroaryl); -N ═ C (C)2-6Alkyl radical)2;-N=C(C2-6Alkyl) (C3-6Cycloalkyl groups); -N ═ C (C)2-6Alkyl radical)(aryl); -N ═ C (C)2-6Alkyl) (phenyl); -N ═ C (C)2-6Alkyl) (heteroaryl); -N ═ C (C)3-6Cycloalkyl radicals2;-N=C(C3-6Cycloalkyl) (aryl); -N ═ C (C)3-6Cycloalkyl) (phenyl); -N ═ C (C) 3-6Cycloalkyl) (heteroaryl); -N ═ C (aryl)2(ii) a -N ═ C (aryl) (phenyl); -N ═ C (aryl) (heteroaryl); -N ═ C (phenyl)2(ii) a -N ═ C (phenyl) (heteroaryl); -N ═ C (heteroaryl)2(ii) a A ketone; acetyl; -C (O) CH3;-C(O)C2-6An alkyl group; -C (O) C3-6A cycloalkyl group; -C (O) aryl; -c (o) phenyl; -c (o) heteroaryl; -CH2C(O)CH3;-CH2C(O)C2-6An alkyl group; -CH2C(O)C3-6A cycloalkyl group; -CH2C (O) aryl; -CH2C (O) phenyl; -CH2C (O) heteroaryl; -C2-6Alkyl radical C (O) C2-6An alkyl group; -C2-6Alkyl radical C (O) C3-6A cycloalkyl group; -C2-6Alkyl C (O) aryl; -C2-6Alkyl C (O) phenyl; -C2-6Alkyl C (O) heteroaryl; -C (O) CH2CH3;-C(O)CH2C2-6An alkyl group; -C (O) CH2C3-6A cycloalkyl group; -C (O) CH2An aryl group; -C (O) CH2A phenyl group; -C (O) CH2A heteroaryl group; -C (O) C2-6Alkyl group CH3;-C(O)C2-6Alkyl radical C2-6An alkyl group; -C (O) C2-6Alkyl radical C3-6A cycloalkyl group; -C (O) C2-6An alkylaryl group; -C (O) C2-6An alkyl phenyl group; -C (O) C2-6An alkyl heteroaryl group; an aldehyde; -CHO; -CH2CHO;-C2-6Alkyl CHO; an ester; -CO2CH3;-CO2C2-6An alkyl group; -CO2C3-6A cycloalkyl group; -CO2An aryl group; -CO2A phenyl group; -CO2A heteroaryl group; reverse ester; an acetoxy group; -OCOCH3;-OCOC2-6An alkyl group; -OCOC3-6A cycloalkyl group; -an OCO aryl group; -ocophenyl; -an OCO heteroaryl group; oxygen (═ O); a hydrazine group; -NHNH2;-N(CH3)NH2;-N(C2-6Alkyl) NH2;-N(C3-6Cycloalkyl) NH2;-N (aryl) NH2(ii) a -N (phenyl) NH2(ii) a -N (heteroaryl) NH2;-N(COCH3)NH2;-N(COC2-6Alkyl) NH2;-N(COC3-6Cycloalkyl) NH2(ii) a -N (COaryl) NH 2(ii) a -N (CO phenyl) NH2(ii) a -N (CO heteroaryl) NH2;-NHNH(CH3);-NHNH(C2-6Alkyl groups); -NHNH (C)3-6Cycloalkyl groups); -NHNH (aryl); -NHNH (phenyl); -NHNH (heteroaryl); -NHNH (COCH)3);-NHNH(COC2-6Alkyl groups); -NHNH (COC)3-6Cycloalkyl groups); -NHNH (coeryl); -NHNH (CO phenyl); -NHNH (CO heteroaryl); -N (CH)3)NH(CH3);-N(CH3)NH(C2-6Alkyl groups); -N (CH)3)NH(C3-6Cycloalkyl groups); -N (CH)3) NH (aryl); -N (CH)3) NH (phenyl); -N (CH)3) NH (heteroaryl); -N (CH)3)NH(COCH3);-N(CH3)NH(COC2-6Alkyl groups); -N (CH)3)NH(COC3-6Cycloalkyl groups); -N (CH)3) NH (CO aryl); -N (CH)3) NH (CO phenyl); -N (CH)3) NH (CO heteroaryl); -N (C)2-6Alkyl) NH (CH)3);-N(C2-6Alkyl) NH (C)2-6Alkyl groups); -N (C)2-6Alkyl) NH (C)3-6Cycloalkyl groups); -N (C)2-6Alkyl) NH (aryl); -N (C)2-6Alkyl) NH (phenyl); -N (C)2-6Alkyl) NH (heteroaryl); -N (C)2-6Alkyl) NH (COCH)3);-N(C2-6Alkyl) NH (COC)2-6Alkyl groups); -N (C)2-6Alkyl) NH (COC)3-6Cycloalkyl groups); -N (C)2-6Alkyl) NH (coaryl); -N (C)2-6Alkyl) NH (CO phenyl); -N (C)2-6Alkyl) NH (CO heteroaryl); -N (C)3-6Cycloalkyl) NH (CH)3);-N(C3-6Cycloalkyl) NH (C)2-6Alkyl groups); -N (C)3-6Cycloalkyl) NH (C)3-6Cycloalkyl groups); -N (C)3-6Cycloalkyl) NH (aryl); -N (C)3-6Cycloalkyl) NH (phenyl); -N (C)3-6Cycloalkyl) NH (heteroaryl); -N (C)3-6Cycloalkyl) NH (COCH)3);-N(C3-6Cycloalkyl) NH (COC2-6Alkyl groups); -N (C)3-6Cycloalkyl radicalsNH(COC3-6Cycloalkyl groups); -N (C)3-6Cycloalkyl) NH (coaryl); -N (C)3-6Cycloalkyl) NH (cofhenyl); -N (C) 3-6Cycloalkyl) NH (CO heteroaryl); -N (aryl) NH (CH)3) (ii) a -N (aryl) NH (C)2-6Alkyl groups); -N (aryl) NH (C)3-6Cycloalkyl groups); -N (aryl) NH (aryl); -N (aryl) NH (phenyl); -N (aryl) NH (heteroaryl); -N (aryl) NH (COCH)3) (ii) a -N (aryl) NH (COC)2-6Alkyl groups); -N (aryl) NH (COC)3-6Cycloalkyl groups); -N (aryl) NH (fluoroaryl); -N (aryl) NH (cophenyl); -N (aryl) NH (CO heteroaryl); -N (phenyl) NH (CH)3) (ii) a -N (phenyl) NH (C)2-6Alkyl groups); -N (phenyl) NH (C)3-6Cycloalkyl groups); -N (phenyl) NH (aryl); -N (phenyl) NH (phenyl); -N (phenyl) NH (heteroaryl); -N (phenyl) NH (COCH)3) (ii) a -N (phenyl) NH (COC)2-6Alkyl groups); -N (phenyl) NH (COC)3-6Cycloalkyl groups); -N (phenyl) NH (fluoroaryl); -N (phenyl) NH (cofhenyl); -N (phenyl) NH (CO heteroaryl); -N (heteroaryl) NH (CH)3) (ii) a -N (heteroaryl) NH (C)2-6Alkyl groups); -N (heteroaryl) NH (C)3-6Cycloalkyl groups); -N (heteroaryl) NH (aryl); -N (heteroaryl) NH (phenyl); -N (heteroaryl) NH (heteroaryl); -N (heteroaryl) NH (COCH)3) (ii) a -N (heteroaryl) NH (COC)2-6Alkyl groups); -N (heteroaryl) NH (COC)3-6Cycloalkyl groups); -N (heteroaryl) NH (fluoroaryl); -N (heteroaryl) NH (cophenyl); -N (heteroaryl) NH (CO heteroaryl); -N (COCH)3)NH(CH3);-N(COCH3)NH(C2-6Alkyl groups); -N (COCH)3)NH(C3-6Cycloalkyl groups); -N (COCH)3) NH (aryl); -N (COCH) 3) NH (phenyl); -N (COCH)3) NH (heteroaryl); -N (COCH)3)NH(COCH3);-N(COCH3)NH(COC2-6Alkyl groups); -N (COCH)3)NH(COC3-6Cycloalkyl groups); -N (COCH)3) NH (CO aryl); -N (COCH)3) NH (CO phenyl); -N (COCH)3) NH (CO heteroaryl); -N (COC)2-6Alkyl) NH (CH)3);-N(COC2-6Alkyl) NH (C)2-6Alkyl groups); -N (COC)2-6Alkyl) NH (C)3-6Cycloalkyl groups); -N (COC)2-6Alkyl) NH (aryl); -N (COC)2-6Alkyl) NH (phenyl); -N (COC)2-6Alkyl) NH (heteroaryl); -N (COC)2-6Alkyl) NH (COCH)3);-N(COC2-6Alkyl) NH (COC)2-6Alkyl groups); -N (COC)2-6Alkyl) NH (COC)3-6Cycloalkyl groups); -N (COC)2-6Alkyl) NH (coaryl); -N (COC)2-6Alkyl) NH (CO phenyl); -N (COC)2-6Alkyl) NH (CO heteroaryl); -N (COC)3-6Cycloalkyl) NH (CH)3);-N(COC3-6Cycloalkyl) NH (C)2-6Alkyl groups); -N (COC)3-6Cycloalkyl) NH (C)3-6Cycloalkyl groups); -N (COC)3-6Cycloalkyl) NH (aryl); -N (COC)3-6Cycloalkyl) NH (phenyl); -N (COC)3-6Cycloalkyl) NH (heteroaryl); -N (COC)3-6Cycloalkyl) NH (COCH)3);-N(COC3-6Cycloalkyl) NH (COC2-6Alkyl groups); -N (COC)3-6Cycloalkyl) NH (COC3-6Cycloalkyl groups); -N (COC)3-6Cycloalkyl) NH (coaryl); -N (COC)3-6Cycloalkyl) NH (cofhenyl); -N (COC)3-6Cycloalkyl) NH (CO heteroaryl); -N (COaryl) NH (CH)3) (ii) a -N (COaryl) NH (C)2-6Alkyl groups); -N (COaryl) NH (C)3-6Cycloalkyl groups); -N (coeryl) NH (aryl); -N (coeryl) NH (phenyl); -N (coeryl) NH (heteroaryl); -N (COaryl) NH (COCH)3) (ii) a -N (COaryl) NH (COC) 2-6Alkyl groups); -N (COaryl) NH (COC)3-6Cycloalkyl groups); -N (coaryl) NH (coaryl); -N (coaryl) NH (coaph); -N (coaryl) NH (coaheteroaryl); -N (CO phenyl) NH (CH)3) (ii) a -N (CO phenyl) NH (C)2-6Alkyl groups); -N (CO phenyl) NH (C)3-6Cycloalkyl groups); -N (cophenyl) NH (aryl); -N (cophenyl) NH (phenyl); -N (cophenyl) NH (heteroaryl); -N (Cophenyl) NH (COCH)3) (ii) a -N (CO phenyl) NH (COC)2-6Alkyl groups); -N (CO phenyl) NH (COC)3-6Cycloalkyl groups); -N (cophenyl) NH (coparyl); -N (cophenyl) NH (cophenyl); -N (cophenyl) NH (CO heteroaryl); -N (CO heteroaryl) NH (CH)3) (ii) a -N (CO heteroaryl) NH (C)2-6Alkyl groups); -N (CO heteroaryl) NH (C)3-6Cycloalkyl groups); -N (CO heteroaryl) NH (aryl); -N (CO heteroaryl) NH (phenyl); -N (CO heteroaryl) NH (heteroaryl); -N (CO heteroaryl) NH (COCH)3) (ii) a -N (CO heteroaryl) NH (COC)2-6Alkyl groups); -N (CO heteroaryl) NH (COC)3-6Cycloalkyl groups); -N (CO heteroaryl) NH (cooaryl); -N (CO heteroaryl) NH (cophenyl); -N (CO heteroaryl) NH (CO heteroaryl); -NHN (CH)3)2;-NHN(CH3)(C2-6Alkyl groups); -NHN (CH)3)(C3-6Cycloalkyl groups); -NHN (CH)3) (aryl); -NHN (CH)3) (phenyl); -NHN (CH)3) (heteroaryl); -NHN (CH)3)(COCH3);-NHN(CH3)(COC2-6Alkyl groups); -NHN (CH)3)(COC3-6Cycloalkyl groups); -NHN (CH)3) (CO aryl); -NHN (CH)3) (CO phenyl); -NHN (CH)3) (CO heteroaryl); -NHN (C) 2-6Alkyl radical)2;-NHN(C2-6Alkyl) (C3-6Cycloalkyl groups); -NHN (C)2-6Alkyl) (aryl); -NHN (C)2-6Alkyl) (phenyl); -NHN (C)2-6Alkyl) (heteroaryl); -NHN (C)2-6Alkyl) (COCH3);-NHN(C2-6Alkyl) (COC2-6Alkyl groups); -NHN (C)2-6Alkyl) (COC2-6Cycloalkyl groups); -NHN (C)2-6Alkyl) (coaryl); -NHN (C)2-6Alkyl) (CO phenyl); -NHN (C)2-6Alkyl) (CO heteroaryl); -NHN (C)3-6Cycloalkyl radicals2;-NHN(C3-6Cycloalkyl) (aryl); -NHN (C)3-6Cycloalkyl) (phenyl); -NHN (C)3-6Cycloalkyl) (heteroaryl); -NHN (C)3-6Cycloalkyl) (COCH3);-NHN(C3-6Cycloalkyl) (COC2-6Alkyl groups); -NHN (C)3-6Cycloalkyl) (COC3-6Cycloalkyl groups); -NHN (C)3-6Cycloalkyl) (coaryl); -NHN (C)3-6Cycloalkyl) (CO phenyl); -NHN (C)3-6Cycloalkyl) (CO heteroaryl); -NHN (aryl)2(ii) a -NHN (aryl) (phenyl); -NHN (aryl) (heteroaryl); -NHN (aryl) (COCH)3) (ii) a -NHN (aryl) (COC)2-6Alkyl groups); -NHN (aryl) (COC)3-6Cycloalkyl groups); -NHN (aryl) (coeryl); -NHN (aryl) (cofhenyl); -NHN (aryl) (CO heteroaryl); -NHN (phenyl)2(ii) a -NHN (phenyl) (heteroaryl); -NHN (phenyl) (COCH3) (ii) a -NHN (phenyl) (COC)2-6Alkyl groups); -NHN (phenyl) (COC)3-6Cycloalkyl groups); -NHN (Phenyl) (CO aryl); -NHN (phenyl) (cofhenyl); -NHN (phenyl) (CO heteroaryl); -NHN (heteroaryl)2(ii) a -NHN (heteroaryl) (COCH)3) (ii) a -NHN (heteroaryl) (COC) 2-6Alkyl groups); -NHN (heteroaryl) (COC)3-6Cycloalkyl groups); -NHN (heteroaryl) (coeryl); -NHN (heteroaryl) (cofhenyl); -NHN (heteroaryl) (CO heteroaryl); -NHN (COCH)3)2;-NHN(COCH3)(COC2-6Alkyl groups); -NHN (COCH)3)(COC3-6Cycloalkyl groups); -NHN (COCH)3) (CO aryl); -NHN (COCH)3) (CO phenyl); -NHN (COCH)3) (CO heteroaryl); -NHN (COC)2-6Alkyl radical)2;-NHN(COC2-6Alkyl) (COC3-6Cycloalkyl groups); -NHN (COC)2-6Alkyl) (coaryl); -NHN (COC)2-6Alkyl) (CO phenyl); -NHN (COC)3-6Alkyl) (CO heteroaryl); -NHN (COC)3-6Cycloalkyl radicals2;-NHN(COC3-6Cycloalkyl) (coaryl); -NHN (COC)3-6Cycloalkyl) (CO phenyl); -NHN (COC)3-6Cycloalkyl) (CO heteroaryl); -NHN (CO aryl)2(ii) a -NHN (coaryl) (cophenyl); -NHN (coeryl) (CO heteroaryl); -NHN (CO phenyl)2(ii) a -NHN (CO phenyl) (CO heteroaryl); -NHN (CO heteroaryl)2;-N(CH3)N(CH3)2;-N(CH3)N(CH3)(C2-6Alkyl groups); -N (CH)3)N(CH3)(C3-6Cycloalkyl groups); -N (CH)3)N(CH3) (aryl); -N (CH)3)N(CH3) (phenyl); -N (CH)3)N(CH3) (heteroaryl); -N (CH)3)N(CH3)(COCH3);-N(CH3)N(CH3)(COC2-6Alkyl groups); -N (CH)3)N(CH3)(COC3-6Cycloalkyl groups); -N (CH)3)N(CH3) (CO aryl); -N (CH)3)N(CH3) (CO phenyl); -N (CH)3)N(CH3) (CO heteroaryl); -N (CH)3)N(C2-6Alkyl radical)2;-N(CH3)N(C2-6Alkyl) (C3-6Cycloalkyl groups); -N (CH)3)N(C2-6Alkyl) (aryl); -N (CH)3)N(C2-6Alkyl) (phenyl); -N (CH)3)N(C2-6Alkyl) (heteroaryl); -N (CH)3)N(C2-6Alkyl) (COCH3);-N(CH3)N(C2-6Alkyl) (COC2-6Alkyl groups); -N (CH)3)N(C2-6Alkyl) (COC3-6Cycloalkyl groups); -N (CH)3)N(C2-6Alkyl) (coaryl); -N (CH) 3)N(C2-6Alkyl) (CO phenyl); -N (CH)3)N(C2-6Alkyl) (CO heteroaryl); -N (CH)3)N(C3-6Cycloalkyl radicals2;-N(CH3)N(C3-6Cycloalkyl) (aryl); -N (CH)3)N(C3-6Cycloalkyl) (phenyl); -N (CH)3)N(C3-6Cycloalkyl) (heteroaryl); -N (CH)3)N(C3-6Cycloalkyl) (COCH3);-N(CH3)N(C3-6Cycloalkyl) (COC2-6Alkyl groups); -N (CH)3)N(C3-6Cycloalkyl) (COC3-6Cycloalkyl groups); -N (CH)3)N(C3-6Cycloalkyl) (coaryl); -N (CH)3)N(C3-6Cycloalkyl) (CO phenyl); -N (CH)3)N(C3-6Cycloalkyl) (CO heteroaryl); -N (CH)3) N (aryl)2;-N(CH3) N (aryl) (phenyl); -N (CH)3) N (aryl) (heteroaryl); -N (CH)3) N (aryl) (COCH)3);-N(CH3) N (aryl) (COC)2-6Alkyl groups); -N (CH)3) N (aryl) (COC)3-6Cycloalkyl groups); -N (CH)3) N (aryl) (cooaryl); -N (CH)3) N (aryl) (CO phenyl); -N (CH)3) N (aryl) (CO heteroaryl); -N (CH)3) N (phenyl)2;-N(CH3) N (phenyl) (heteroaryl); -N (CH)3) N (phenyl) (COCH)3);-N(CH3) N (phenyl) (COC)2-6Alkyl groups); -N (CH)3) N (phenyl) (COC)3-6Cycloalkyl groups); -N (CH)3) N (phenyl) (cooaryl); -N (CH)3) N (phenyl) (CO phenyl); -N (CH)3) N (phenyl) (CO heteroaryl); -N (CH)3) N (heteroaryl)2;-N(CH3) N (heteroaryl) (COCH)3);-N(CH3) N (heteroaryl) (COC)2-6Alkyl groups); -N (CH)3) N (heteroaryl) (COC)3-6Cycloalkyl groups); -N (CH)3) N (heteroaryl) (coeryl); -N (CH)3) N (heteroaryl) (cofhenyl); -N (CH)3) N (heteroaryl) (CO heteroaryl); -N (CH)3)N(COCH3)2;-N(CH3)N(COCH3)(COC2-6Alkyl groups); -N (CH)3)N(COCH3)(COC3-6Cycloalkyl groups); -N (CH)3)N(COCH3) (CO aryl); -N (CH) 3)N(COCH3) (CO phenyl); -N (CH)3)N(COCH3) (CO heteroaryl); -N (CH)3)N(COC2-6Alkyl radical)2;-N(CH3)N(COC2-6Alkyl) (COC3-6Cycloalkyl groups); -N (CH)3)N(COC2-6Alkyl) (coaryl); -N (CH)3)N(COC2-6Alkyl) (CO phenyl); -N (CH)3)N(COC2-6Alkyl) (CO heteroaryl); -N (CH)3)N(COC3-6Cycloalkyl radicals2;-N(CH3)N(COC3-6Cycloalkyl) (coaryl); -N (CH)3)N(COC3-6Cycloalkyl) (CO phenyl); -N (CH)3)N(COC3-6Cycloalkyl) (CO heteroaryl); -N (CH)3) N (CO aryl)2;-N(CH3) N (coaryl) (coaphenyl); -N (CH)3) N (coaryl) (CO heteroaryl); -N (CH)3) N (CO phenyl)2;-N(CH3) N (cophenyl) (CO heteroaryl); -N (CH)3) N (CO heteroaryl)2(ii) a -N (phenyl) N (CH)3)2(ii) a -N (phenyl) N (CH)3)(C2-6Alkyl groups); -N (phenyl) N (CH)3)(C3-6Cycloalkyl groups); -N (phenyl) N (CH)3) (aryl); -N (phenyl) N (CH)3) (phenyl); -N (phenyl) N (CH)3) (heteroaryl); -N (phenyl) N (CH)3)(COCH3) (ii) a -N (phenyl) N (CH)3)(COC2-6Alkyl groups); -N (phenyl) N (CH)3)(COC3-6Cycloalkyl groups); -N (phenyl) N (CH)3) (CO aryl); -N (phenyl) N (CH)3) (CO phenyl); -N (phenyl) N (CH)3) (CO heteroaryl); -N (phenyl) N (C)2-6Alkyl radical)2(ii) a -N (phenyl) N (C)2-6Alkyl) (C3-6Cycloalkyl groups); -N (phenyl) N (C)2-6Alkyl) (aryl); -N (phenyl) N (C)2-6Alkyl) (phenyl); -N (phenyl) N (C)2-6Alkyl) (heteroaryl); -N (phenyl) N (C)2-6Alkyl) (COCH3) (ii) a -N (phenyl) N (C)2-6Alkyl) (COC2-6Alkyl groups); -N (phenyl) N (C)2-6Alkyl) (COC3-6Cycloalkyl groups); -N (phenyl) N (C)2-6Alkyl) (coaryl); -N (phenyl) N (C) 2-6Alkyl) (CO phenyl); -N (phenyl) N (C)2-6Alkyl) (CO heteroaryl); -N (phenyl) N (C)3-6Cycloalkyl radicals2(ii) a -N (phenyl) N (C)3-6Cycloalkyl) (aryl); -N (phenyl) N (C)3-6Cycloalkyl) (phenyl); -N (phenyl) N (C)3-6Cycloalkyl) (heteroaryl); -N (phenyl) N (C)3-6Cycloalkyl) (COCH3) (ii) a -N (phenyl) N (C)3-6Cycloalkyl) (COC2-6Alkyl groups); -N (phenyl) N (C)3-6Cycloalkyl) (COC3-6Cycloalkyl groups); -N (phenyl) N (C)3-6Cycloalkyl) (coaryl); -N (phenyl) N (C)3-6Cycloalkyl) (CO phenyl); -N (phenyl) N (C)3-6Cycloalkyl) (CO heteroaryl); -N (phenyl) N (aryl)2(ii) a -N (phenyl) N (aryl) (phenyl); -N (phenyl) N (aryl) (heteroaryl); -N (phenyl) N (aryl) (COCH)3) (ii) a -N (phenyl) N (aryl) (COC)2-6Alkyl groups); -N (phenyl) N (aryl) (COC)3-6Cycloalkyl groups); -N (phenyl) N (aryl) (coeryl); -N (phenyl) N (aryl) (cophenyl); -N (phenyl) N (aryl) (CO heteroaryl); -N (phenyl)2(ii) a -N (phenyl) (heteroaryl); -N (phenyl) (COCH)3) (ii) a -N (phenyl) (COC2-6Alkyl groups); -N (phenyl) (COC3-6Cycloalkyl groups); -N (phenyl) (coaryl); -N (phenyl) (cophenyl); -N (phenyl) (CO heteroaryl); -N (phenyl) N (heteroaryl)2(ii) a -N (phenyl) N (heteroaryl) (COCH)3) (ii) a -N (phenyl) N (heteroaryl) (COC) 2-6Alkyl groups); -N (phenyl) N (heteroaryl) (COC)3-6Cycloalkyl groups); -N (phenyl) N (heteroaryl) (oaryl); -N (phenyl) N (heteroaryl) (cofhenyl); -N (phenyl) N (heteroaryl) (CO heteroaryl); -N (phenyl) N (COCH)3)2(ii) a -N (phenyl) N (COCH)3)(COC2-6Alkyl groups); -N (phenyl) N (COCH)3)(COC3-6Cycloalkyl groups); -N (phenyl) N (COCH)3) (CO aryl); -N (phenyl) N (COC)H3) (CO phenyl); -N (phenyl) N (COCH)3) (CO heteroaryl); -N (phenyl) N (COC)2-6Alkyl radical)2(ii) a -N (phenyl) N (COC)2-6Alkyl) (COC3-6Cycloalkyl groups); -N (phenyl) N (COC)2-6Alkyl) (coaryl); -N (phenyl) N (COC)2-6Alkyl) (CO phenyl); -N (phenyl) N (COC)2-6Alkyl) (CO heteroaryl); -N (phenyl) N (COC)3-6Cycloalkyl radicals2(ii) a -N (phenyl) N (COC)3-6Cycloalkyl) (coaryl); -N (phenyl) N (COC)3-6Cycloalkyl) (CO phenyl); -N (phenyl) N (COC)3-6Cycloalkyl) (CO heteroaryl); -N (phenyl) N (COaryl)2(ii) a -N (phenyl) N (coaryl) (coaph); -N (phenyl) N (coeryl) (coeheteroaryl); -N (phenyl) N (Cophenyl)2(ii) a -N (phenyl) N (cophenyl) (CO heteroaryl); -N (phenyl) N (CO heteroaryl)2(ii) a A hydrazone group; NNH (NNH)2;=NNH(CH3);=NNH(C2-6Alkyl groups); NNH (C)3-6Cycloalkyl groups); NNH (aryl); NNH (phenyl); NNH (heteroaryl); NNH (COCH)3);=NNH(COC2-6Alkyl groups); NNH (COC)3-6Cycloalkyl groups); NNH (aryl CO); NNH (CO phenyl); NNH (CO heteroaryl); NN (CH) 3)2;=NN(CH3)(C2-6Alkyl groups); NN (CH)3)(C3-6Cycloalkyl groups); NN (CH)3) (aryl); NN (CH)3) (phenyl); NN (CH)3) (heteroaryl); NN (CH)3)(COCH3);=NN(CH3)(COC2-6Alkyl groups); NN (CH)3)(COC3-6Cycloalkyl groups); NN (CH)3) (CO aryl); NN (CH)3) (CO phenyl); NN (CH)3) (CO heteroaryl); NN (C)2-6Alkyl radical)2;=NN(C2-6Alkyl) (C3-6Cycloalkyl groups); NN (C)2-6Alkyl) (aryl); NN (C)2-6Alkyl) (phenyl); NN (C)2-6Alkyl) (heteroaryl); NN (C)2-6Alkyl) (COCH3);=NN(C2-6Alkyl) (COC2-6Alkyl groups); NN (C)2-6Alkyl) (COC3-6Cycloalkyl groups); NN (C)2-6Alkyl) (coaryl); NN (C)2-6Alkyl) (CO phenyl));=NN(C2-6Alkyl) (CO heteroaryl); NN (C)3-6Cycloalkyl radicals2;=NN(C3-6Cycloalkyl) (aryl); NN (C)3-6Cycloalkyl) (phenyl); NN (C)3-6Cycloalkyl) (heteroaryl); NN (C)3-6Cycloalkyl) (COCH3);=NN(C3-6Cycloalkyl) (COC2-6Alkyl groups); NN (C)3-6Cycloalkyl) (COC3-6Cycloalkyl groups); NN (C)3-6Cycloalkyl) (coaryl); NN (C)3-6Cycloalkyl) (CO phenyl); NN (C)3-6Cycloalkyl) (CO heteroaryl); NN (aryl)2(ii) a NN (aryl) (phenyl); NN (aryl) (heteroaryl); NN (aryl) (COCH)3) (ii) a NN (aryl) (COC)2-6Alkyl groups); NN (aryl) (COC)3-6Cycloalkyl groups); NN (aryl) (cooaryl); NN (aryl) (CO phenyl); NN (aryl) (CO heteroaryl); NN (phenyl)2(ii) a NN (phenyl) (heteroaryl); NN (phenyl) (COCH) 3) (ii) a NN (phenyl) (COC)2-6Alkyl groups); NN (phenyl) (COC)3-6Cycloalkyl groups); NN (phenyl) (CO aryl); NN (phenyl) (CO phenyl); NN (phenyl) (CO heteroaryl); NN (heteroaryl)2(ii) a NN (heteroaryl) (COCH)3) (ii) a NN (heteroaryl) (COC)2-6Alkyl groups); NN (heteroaryl) (COC)3-6Cycloalkyl groups); NN (heteroaryl) (cooaryl); NN (heteroaryl) (CO phenyl); NN (heteroaryl) (CO heteroaryl); NN (COCH)3)2;=NN(COCH3)(COC2-6Alkyl groups); NN (COCH)3)(COC3-6Cycloalkyl groups); NN (COCH)3) (CO aryl); NN (COCH)3) (CO phenyl); NN (COCH)3) (CO heteroaryl); NN (COC)2-6Alkyl radical)2;=NN(COC2-6Alkyl) (COC3-6Cycloalkyl groups); NN (COC)2-6Alkyl) (coaryl); NN (COC)2-6Alkyl) (CO phenyl); NN (COC)2-6Alkyl) (CO heteroaryl); NN (COC)3-6Cycloalkyl radicals2;=NN(COC3-6Cycloalkyl) (coaryl); NN (COC)3-6Cycloalkyl) (CO phenyl); NN (COC)3-6Cycloalkyl) (CO heteroaryl); NN (CO aryl)2(ii) a NN (CO aryl) (CO phenyl)) (ii) a NN (coaryl) (CO heteroaryl); NN (CO phenyl)2(ii) a NN (CO phenyl) (CO heteroaryl); NN (CO heteroaryl)2(ii) a Haloalkyl (e.g., trifluoromethyl, difluoromethyl, fluoromethyl); substituted aminoacyl and aminoalkyl groups; carbocyclic ring C which may be monocyclic or fused or non-fused polycyclic3-8Cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or heterocycloalkyl which may be monocyclic or fused or non-fused polycyclic (e.g., pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, furanyl, or thiazinyl); carbocyclic or heterocyclic, monocyclic or fused or non-fused polycyclic aryl (e.g., phenyl, naphthyl, pyrrolyl, indolyl, furyl, thienyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, pyrazolyl, pyridyl, quinolyl, isoquinolyl, acridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, benzimidazolyl, benzothienyl, or benzofuryl); -O-aryl; an aryl group; aryl-C 1-Or2-6An alkyl group; an alkoxy group; -OCH3;-OC2-6An alkyl group; -OC3-6A cycloalkyl group; -O phenyl; -O phenyl-C1-Or2-6An alkyl group; -O-heteroaryl; -CO2CH3;-CONH2;-OCH2CONH2;-NH2;-N(C1-4Alkyl radical)2(ii) a An amido group; -NHC (O) CH3;-NHC(O)C2-6An alkyl group; -NHC (O) C3-6A cycloalkyl group; -nhc (o) aryl; -nhc (o) phenyl; -nhc (o) heteroaryl; -N (CH)3)C(O)CH3;-N(CH3)C(O)C2-6An alkyl group; -N (CH)3)C(O)C3-6A cycloalkyl group; -N (CH)3) C (O) aryl; -N (CH)3) C (O) phenyl; -N (CH)3) C (O) heteroaryl; -N (C)2-6Alkyl group C (O) CH3;-N(C2-6Alkyl group(s) C (O) C2-6An alkyl group; -N (C)2-6Alkyl group(s) C (O) C3-6A cycloalkyl group; -N (C)2-6Alkyl) C (O) aryl; -N (C)2-6Alkyl) C (O) phenyl; -N (C)2-6Alkyl) C (O) heteroaryl; -N (C)3-6Cycloalkyl group) C (O) CH3;-N(C3-6Cycloalkyl group) C (O) C2-6An alkyl group; -N (C)3-6Cycloalkyl group) C (O) C3-6A cycloalkyl group; -N (C)3-6Cycloalkyl) C (O) aryl; -N (C)3-6Cycloalkyl) C (O) phenyl; -N (C)3-6Cycloalkyl) C (O) heteroaryl; -N (aryl) C (O) CH3(ii) a -N (aryl) C (O) C2-6An alkyl group; -N (aryl) C (O) C3-6A cycloalkyl group; -N (aryl) c (o) aryl; -N (aryl) c (o) phenyl; -N (aryl) c (o) heteroaryl; -N (phenyl) C (O) CH3(ii) a -N (phenyl) C (O) C2-6An alkyl group; -N (phenyl) C (O) C3-6A cycloalkyl group; -N (phenyl) c (o) aryl; -N (phenyl) c (o) phenyl; -N (phenyl) c (o) heteroaryl; -N (heteroaryl) C (O) CH3(ii) a -N (heteroaryl) C (O) C2-6An alkyl group; -N (heteroaryl) C (O) C3-6A cycloalkyl group; -N (heteroaryl) c (o) aryl; -N (heteroaryl) c (o) phenyl; -N (heteroaryl) c (o) heteroaryl; guanidino; an amidino group; -SO 2NH2;-OCHF2;-CF3;-OCF3(ii) a And these moieties may also be optionally substituted with fused ring structures or bridges, e.g. -OCH2O-or-O-C1-or 2-6alkylene-O-. These substituents may optionally be further substituted with substituents selected from these groups.

Representative compounds which may be mentioned in the present invention are those provided in the examples as free bases or pharmaceutically acceptable salts thereof. The molecular weight of the compounds of the invention (in free base form) is preferably less than 1000g/mol, more preferably less than 900g/mol, most preferably less than 800 g/mol.

When the compounds of the present invention and pharmaceutically acceptable salts thereof exist in solvate or polymorphic forms, the present invention includes any possible solvate and polymorphic forms. The type of the solvent forming the solvate is not particularly limited as long as the solvent is pharmacologically acceptable. For example, water, ethanol, propanol, acetone, or the like can be used.

The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids. When the compounds of the present invention are acidic, their corresponding salts may be conveniently prepared from pharmaceutically acceptable non-toxic bases including inorganic and organic bases. Salts derived from these inorganic bases include aluminum, ammonium, calcium, copper (both copper and cuprous), iron, ferrous, lithium, magnesium, potassium, sodium, zinc, and the like. Particularly preferred are ammonium, calcium, magnesium, potassium and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines as well as cyclic and substituted amines such as naturally occurring and synthetic substituted amines. Other pharmaceutically acceptable organic non-toxic bases from which salts can be formed include arginine, betaine, caffeine, choline, N' -dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, reduced glucamine, histidine, hydrabamine, isopropylamine, lysine, methyl reduced glucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.

When the compounds of the present invention are basic, their corresponding salts may be conveniently prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. These acids include, for example, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid and the like. When the compounds of the invention are for pharmaceutical use, they are preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure, especially at least 98% pure (% are in weight/weight).

The invention also provides a compound of the invention, or a pharmaceutically acceptable salt thereof, for use as a medicament or medicament. The invention also provides a pharmaceutical composition comprising at least one compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. The invention also provides a pharmaceutical composition comprising at least one compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient. Preferably, these compositions consist of a pharmaceutically acceptable carrier or a pharmaceutically acceptable carrier, diluent or excipient, and a non-toxic therapeutically effective amount of the at least one compound of the present invention or a pharmaceutically acceptable salt thereof.

The compounds of the present invention may be used in combination with further antibacterial agents. Accordingly, the present invention also provides a composition comprising at least one compound of the invention or a pharmaceutically acceptable salt thereof and at least one further antibacterial agent, wherein the at least one further antibacterial agent is different from the compound of the invention or a pharmaceutically acceptable salt thereof. Preferably, the at least one further antibacterial agent is a regulatory approved antibiotic, in particular a regulatory approved member of the antibiotic family selected from the group consisting of penicillins, cephalosporins, carbapenems, monobactams, polymyxins, rifamycins, lipiarmycins, quinolones, sulfonamides, macrolides, lincosamides, tetracyclines, aminoglycosides, lipopeptides, glycylcyclines, glycopeptides, oxazolidinones, and lipiarmycins. When the present invention relates to a composition comprising at least one compound of the invention or a pharmaceutically acceptable salt thereof and at least one further antibacterial agent, said at least one further antibacterial agent may be a β -lactam antibiotic, in particular a β -lactam antibiotic selected from the group consisting of penicillin antibiotics, cephalosporin antibiotics, and carbapenem antibiotics, or any combination thereof. In the foregoing context, the composition may further comprise at least one additional agent capable of overcoming one or more bacterial resistance mechanisms. Examples of such additional agents include clavulanic acid, sulbactam, tazobactam, avibactam, rilebabactam, faribabactam, and any combination thereof.

Accordingly, such a composition may preferably comprise (i) at least one further antibacterial agent which is a β -lactam antibiotic, in particular an existing regulatory approved β -lactam antibiotic, more in particular an existing regulatory approved β -lactam antibiotic selected from the group consisting of penicillin antibiotics, cephalosporin antibiotics, and carbapenem antibiotics, or any combination thereof; and (ii) at least one additional agent capable of overcoming one or more bacterial resistance mechanisms, said additional agent preferably selected from the group consisting of clavulanic acid, sulbactam, tazobactam, avibactam, releptian, fubactam, and any combination thereof. (i) The specific combination of a beta-lactam antibiotic and said (ii) at least one additional agent capable of overcoming one or more bacterial resistance mechanisms of particular interest comprises

(i) Amoxicillin and clavulanic acid;

(ii) ticarcillin and clavulanic acid;

(iii) ampicillin and sulbactam;

(iv) cefoperazone and sulbactam;

(v) piperacillin and tazobactam;

(vi) ceftizolidine and tazobactam;

(vii) ceftazidime and avibactam;

(viii) ceftaroline and abamectin;

(ix) Carbapenems, in particular epetionam or meropenem, and releptin; and

(x) Carbapenems, in particular epeniam or meropenem, and faropenem.

Accordingly, the present invention provides a composition comprising at least one compound of the present invention or a pharmaceutically acceptable salt thereof and any one of combinations (i) to (x). These compositions may further comprise a pharmaceutically acceptable carrier. These compositions may further comprise pharmaceutically acceptable carriers, diluents and/or excipients.

The pharmaceutical compositions disclosed herein may optionally include additional therapeutic ingredients or adjuvants. The compositions include those suitable for oral, rectal, topical, pulmonary and parenteral (including subcutaneous, intramuscular and intravenous) administration, although the most suitable route in any given case will depend on the particular host and the nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions may conveniently be presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.

In practice, the compounds of the present invention or pharmaceutically acceptable salts thereof may be employed as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral, parenteral (including intravenous, intramuscular, subcutaneous), topical, and pulmonary.

Thus, the pharmaceutical compositions may be presented as discrete units suitable for oral administration, such as capsules, cachets, or tablets each containing a predetermined amount of the active ingredient. Furthermore, the composition may be in the form of a powder, granules, a solution, a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion. In addition to the usual dosage forms described above, the compounds of the present invention or pharmaceutically acceptable salts thereof may also be administered by controlled release means and/or delivery devices. The compositions may be prepared by any of the methods of pharmacy. Generally, such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more necessary ingredients. Generally, compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both. The product can then be conveniently shaped to the desired appearance. The compounds of the present invention or pharmaceutically acceptable salts thereof may also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds. The pharmaceutical carrier used may be, for example, a solid, liquid, or gas. Examples of solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Examples of liquid carriers are syrup, peanut oil, olive oil, and water. Examples of gas carriers include carbon dioxide and nitrogen.

In preparing the compositions for oral dosage form, any convenient pharmaceutical medium may be employed. For example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like can be used to form oral liquid preparations such as suspensions, elixirs, and solutions; and carriers such as starch, sugar, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like may be used to form oral solid preparations such as powders, capsules and tablets. Because of their ease of administration, tablets and capsules are the preferred oral dosage units wherein solid pharmaceutical carriers are employed. Optionally, the tablets may be coated by standard aqueous or non-aqueous techniques. Oral administration of the compounds of the invention may be particularly useful in situations where bioavailability of the compounds is not required, such as in the treatment of intestinal infections such as colonic infections. Alternatively, the compounds of the present invention may be combined with drug delivery techniques known in the art to make aminoglycosides bioavailable.

Tablets containing the composition of the invention may be prepared by compression or molding, optionally together with one or more accessory ingredients (excipients) or adjuvants. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be prepared by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. Each tablet preferably contains from about 0.05mg to about 5g of active ingredient, and each cachet or capsule preferably contains from about 0.05mg to about 5g of active ingredient. For example, a formulation for oral administration to humans may contain from about 0.5mg to about 5g of the active agent, in admixture with a suitable and convenient amount of carrier material which may comprise from about 5% to about 95% of the total composition. Unit dosage forms typically contain from about 1mg to about 2g of active ingredient, typically 2mg, 5mg, 10mg, 25mg, 50mg, 100mg, 200mg, 300mg, 400mg, 500mg, 600mg, 800mg, or 1000 mg.

Pharmaceutical compositions of the invention suitable for parenteral administration may be prepared as aqueous solutions or suspensions of the active compounds. Suitable surfactants, such as hydroxypropyl cellulose, may be included. Dispersants may also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. In addition, preservatives may be included to prevent the unwanted growth of microorganisms.

Pharmaceutical compositions of the invention suitable for injectable use include sterile aqueous solutions or dispersions. In addition, the compositions may be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be fluid to be effective for easy injection. The pharmaceutical compositions must be stable under the conditions of manufacture and storage; therefore, it should preferably be protected against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof. The pharmaceutical compositions of the present invention may be in a form suitable for topical use, for example, aerosols, creams, ointments, lotions, dusting powders and the like. Further, the composition may be in a form suitable for use in a transdermal device. These formulations may be prepared by conventional processing methods using the compounds of the present invention or pharmaceutically acceptable salts thereof. For example, a cream or ointment is prepared by mixing a hydrophilic material and water with about 5% by weight to about 10% by weight of a compound to produce a cream or ointment having a desired consistency.

The pharmaceutical compositions of the present invention may be in a form suitable for rectal administration wherein the carrier is a solid. Preferably the mixture is formed into unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. Suppositories may be conveniently formed by first mixing the composition with the softened or molten carrier, followed by cooling and shaping in a mould. Rectal administration of the compounds of the invention may be particularly useful in situations where bioavailability of the compounds is not required, such as in the treatment of intestinal infections such as colonic infections.

The pharmaceutical compositions of the invention may be in a form suitable for pulmonary administration, for example by inhalation using any technique known in the art for delivering drugs to the lungs, including for example administration by a dry powder inhaler or nebulizer. Suitable carriers and formulations for pharmaceutical compositions for administration in this manner are known in the art and may be employed with the compounds of the present invention.

In addition to the above-mentioned carrier ingredients, the above-mentioned pharmaceutical preparations may suitably include one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surfactants, thickeners, lubricants, preservatives (including antioxidants) and the like. In addition, other adjuvants may be included to render the formulation isotonic with the blood of the intended recipient. Compositions containing the compounds of the present invention or pharmaceutically acceptable salts thereof may also be prepared in powder or liquid concentrate form. Typically, dosage levels of 0.01mg/kg to about 150mg/kg body weight per day are useful for treating the above conditions, or about 0.5mg to about 7g per patient per day. For example, microbial infections can be effectively treated by administering about 0.01 to 50mg of the compound per kilogram of body weight per day, or about 0.5mg to about 3.5g of the compound per patient per day. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing therapy. For example, it is contemplated that when a compound of the invention is administered by inhalation, a higher dose is required as opposed to, for example, parenteral administration, and in the treatment of microbial infections, the dose when the compound is administered by inhalation (e.g., using a nebulizer) may be from 0.01mg/kg to about 500mg/kg body weight per day. The actual methods of preparing the compositions and dosage forms, such as those mentioned above, are known or will be apparent to those skilled in the art; see, for example, Remington, The Science and Practice of Pharmacy,21st Edition (University of The Sciences in Philadelphia, 2005).

A "pharmaceutically acceptable carrier, diluent or excipient" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersant, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier that has been approved by the U.S. food and drug administration as acceptable for use in humans or livestock at the effective date of this application.

The invention also provides a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in or as a medicament or pharmaceutical.

The terms "compound of the invention" and "compound of formula (I)" are to be understood as interchangeable with respect to the compounds disclosed herein, the uses of the compounds disclosed herein, the compositions comprising the compounds disclosed herein and the uses of the compositions comprising the compounds disclosed herein. Thus, when referring to the use of, for example, "a compound of formula (I)" or a composition comprising "a compound of formula (I)" is to be understood as meaning the compound of formula (I) as set forth herein for its broadest definition, as well as other compounds that belong to the broadest definition herein and for example further limit one or more of the structural features of the "compound of formula (I)" for the broadest definition (e.g., compounds of formula (I) or compounds of formulae (Ia) to (If), (Ig) to (Ip) and preferred forms thereof, as defined in narrower fashion in the dependent claims).

When reference is made herein to a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in or as a medicament or pharmaceutical product, or to a method of treatment comprising administration of a compound of the invention, or a pharmaceutically acceptable salt thereof, it is to be understood that all compositions disclosed herein, which comprise a compound of the invention, or a pharmaceutically acceptable salt thereof, may also be used in, or administered in, said treatment.

Accordingly, the present invention also provides any of the compositions disclosed herein, including at least one compound of the invention, or a pharmaceutically acceptable salt thereof, for use in or as a medicament or pharmaceutical.

The compounds of the invention, or pharmaceutically acceptable salts thereof, or compositions comprising at least one compound of the invention, or pharmaceutically acceptable salts thereof, are useful for treating a microbial infection and/or a condition, affliction, or disease caused, at least in part, by a microbial infection. When reference is made herein to the treatment of a microbial infection, this is to be understood as also encompassing a condition, affliction or disease caused at least in part by the microbial infection.

As used herein, the term "microbial infection" preferably refers to a bacterial infection. When reference is made herein to the treatment of a bacterial infection, this is to be understood as also encompassing a condition, affliction or disease caused at least in part by the bacterial infection.

As used herein, the term "treatment" includes both therapeutic and prophylactic treatment. The compounds of the present invention may exhibit advantageous properties compared to known compounds or combination therapies used to treat microbial infections. The compounds of the present invention or pharmaceutically acceptable salts thereof may be administered alone or in combination with one or more other therapeutically active compounds, including those combinations comprising at least one further antibacterial agent disclosed herein above. Other therapeutically active compounds may be used to treat the same disease or condition as the compounds of the invention or a different disease or condition, for example in immunocompromised patients. The therapeutically active compounds may be administered simultaneously, sequentially or separately.

The compounds of the invention may be administered with other active compounds useful in the treatment of microbial infections, for example with penicillins, cephalosporins, polymyxins, rifamycins, quinolones, sulfonamides, macrolide antibiotics, lincosamides, tetracyclines, aminoglycosides, cyclic lipopeptides (e.g. daptomycin), glycylcyclines, oxazolidinones (e.g. linezolid). Bacterial efflux pump inhibitors, such as AcrAB-TolC pump or CmeABC efflux pump, can also be administered simultaneously (co-administration), sequentially or separately with a compound or composition of the invention. Thus, any of the compositions disclosed herein above may further comprise at least one bacterial efflux pump inhibitor, such as an AcrAB-TolC pump inhibitor, a MexAB, MexC-, MexEF and mexy pump inhibitor, an AcrD pump inhibitor, or a cmecabc efflux pump inhibitor. Examples of such inhibitors are described in x, z.li, p.plesiat, h.nikaido, clin.microbiol.rev.2015,28(2),337-418 and j.sun, z.deng, a.yan, biochem.biophysis.res.commun.2014, 453,254-267, the contents of each of which are incorporated herein in their entirety.

Co-administration, within the meaning of the present invention, includes administration of a formulation comprising both a compound of the present invention or a pharmaceutically acceptable salt thereof and the other agent, or administration of different formulations of each agent, either simultaneously or separately. Co-administration of the two agents is preferred when the pharmacological properties of the compound of the invention or a pharmaceutically acceptable salt thereof and the other agent allow. The invention also provides the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, and another agent in the manufacture of a medicament for the treatment of a microbial infection. The present invention also provides a pharmaceutical composition comprising at least one compound of the invention, or a pharmaceutically acceptable salt thereof, and another antimicrobial agent, i.e. an additional active compound for the treatment of a microbial infection (as listed elsewhere herein) and a pharmaceutically acceptable carrier.

Certain aminoglycoside antibacterial agents, e.g., gentamicin, are known to be associated with the induction of renal toxicity in subjects administered with the agents. For example, gentamicin is associated with the induction of, inter alia, renal tubular necrosis, edema of the proximal tubular epithelium, cell desquamation, renal tubular fibrosis, glomerular congestion, perivascular edema and inflammation, which in turn can lead to renal dysfunction. Co-administration of aminoglycoside antibacterial agents with adjuvants capable of inhibiting or preventing undesirable nephrotoxic side effects can greatly increase the usefulness of the agents, thereby enabling them to be developed without observing these side effects and/or potentially allowing them to be administered at higher levels than would be possible without co-administration of such adjuvants. Examples of pharmacological classes of such adjuvants include other antibiotics (such as fosfomycin and/or fleroxacin), calcium channel blockers (such as nifedipine and/or amlodipine), beta blockers (such as carvedilol), cytoprotective antianginals (such as tramazine), iNOS inhibitors (such as L-NIL), Nitric Oxide (NO) precursors (such as L-arginine), hormones (such as melatonin and/or thyroxine), antiplatelet drugs (such as trapidil), statins (such as atorvastatin), PPAR-gamma agonists (such as rosiglitazone), TNF-alpha synthesis inhibitors (such as pentoxifylline), biguanides (such as metformin), antioxidants (such as probucol, aminoguanidine, L-carnitine, ibustine, N-acetylcysteine, lycopene, curcumin, and mixtures thereof, Thymoquinone, fish oil, vitamin E, vitamin C, sesame oil, halofuginone, quercetin, or any combination thereof), a free radical scavenger (e.g., S-allylcysteine, diallyl sulfide, phenethyl caffeate, S-allylmercaptocysteine, or any combination thereof), an antioxidant enzyme (e.g., superoxide dismutase), a superoxide dismutase mimetic (e.g., superoxide dismutase mimetic M40403), and a herbal extract (e.g., Rhazya stricta, garlic, Cassia auriculata, soybean, morchella, green tea, nigella sativa, Ligusticum chuanxiong, Viscum album, or any combination thereof). Other examples of such adjuvants include acacia, pongamia, nigella sativa, smilax indica, PESB (phenolic extract of soybean), green tea extract, cercis, dahliand, oleanolicin, and piperacillin, as well as agents and drugs that reduce the tumor toxicity of gentamicin as taught in Ali, b.h.et al.basic and Clinical pharmacy & Toxicology,09, 225-. Thus, the present invention also relates to a composition comprising at least one compound of the invention or a pharmaceutically acceptable salt thereof and at least one adjuvant capable of inhibiting or preventing aminoglycoside-induced nephrotoxicity, in particular wherein said at least one adjuvant is selected from the pharmacological classes of such adjuvants and/or the specific adjuvants listed above, or any combination thereof. The composition may be any other composition of the invention disclosed herein, which further comprises at least one adjuvant capable of inhibiting or preventing aminoglycoside-induced nephrotoxicity, in particular wherein said at least one adjuvant is selected from the pharmacological classes of such adjuvants and/or the specific adjuvants listed above, or any combination thereof. Furthermore, a compound of the invention or a pharmaceutically acceptable salt thereof, or any composition disclosed herein comprising at least one compound of the invention or a pharmaceutically acceptable salt thereof, may therefore be co-administered with one or more members of any one or more of the above-listed pharmacological classes of adjuvants in any of the uses or methods of treatment disclosed herein.

The compounds of the present invention or pharmaceutically acceptable salts thereof or any composition disclosed herein that includes at least one compound of the present invention or pharmaceutically acceptable salt thereof can be used to treat any microbial or bacterial infection. Such microbial or bacterial infections may be caused, or at least partially caused, by gram-positive bacteria, gram-negative bacteria, or a combination of both. Such microbial or bacterial infections may be caused by gram-positive or gram-negative bacteria.

The gram positive bacterium that causes, at least in part, an infection that can be treated by a compound of the present invention, a pharmaceutically acceptable salt thereof, or a composition comprising at least one compound of formula (I), or a pharmaceutically acceptable salt thereof, can be any gram positive bacterium. The gram-positive bacteria may be selected from the group consisting of staphylococcus aureus, streptococcus pneumoniae, enterococcus faecalis, enterococcus faecium, and mycobacterium tuberculosis, among others.

The gram-negative bacterium that causes, at least in part, an infection that can be treated by a compound of the present invention, a pharmaceutically acceptable salt thereof, or a composition comprising at least one compound of the present invention or a pharmaceutically acceptable salt thereof can be any gram-negative bacterium. These gram-negative bacteria may in particular be selected from species of the genus acinetobacter; a species of the enterobacteriaceae family, in particular a species of the genus escherichia, a species of the genus klebsiella, or a species of the genus enterobacteriaceae; species of the genus Morganella; species of the genus providencia; and a species of Pseudomonas, particularly selected from the group consisting of Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, Klebsiella oxytoca, Enterobacter cloacae, Enterobacter aerogenes, Morganella morganii, providencia stuartii, and Pseudomonas aeruginosa. The E.coli may be selected from wild-type E.coli and E.coli strains expressing one or more aminoglycoside modifying enzymes. Furthermore, in this context, each of said acinetobacter species; a species of the enterobacteriaceae family, in particular a species of the genus escherichia, a species of the genus klebsiella, or a species of the genus enterobacteriaceae; species of the genus Morganella; species of the genus providencia; a species of the genus Pseudomonas; acinetobacter baumannii, escherichia coli, klebsiella pneumoniae, klebsiella oxytoca, enterobacter cloacae, enterobacter aerogenes, morganella, providencia stuartii, and pseudomonas aeruginosa can be non-drug resistant strains or strains expressing one or more aminoglycoside modifying enzymes, particularly one or more AME belonging to any of the AME families disclosed elsewhere herein, or any of the particular individual AME disclosed elsewhere herein.

Aminoglycoside modifying enzymes are drug-resistance-causing enzymes that are expressed in a given bacterial species and are capable of modifying the structure of aminoglycoside molecules, thereby potentially "inactivating" their antibacterial ability by reducing their affinity for the target protein, i.e. the 30S ribosomal subunit. In particular, in the present invention, the aminoglycoside modifying enzyme present in a given bacterial species may be selected from the group consisting of Aminoglycoside Acetyltransferase (AAC), Aminoglycoside Phosphotransferase (APH), and Aminoglycoside Nucleotidyl Transferase (ANTs), bifunctional aminoglycoside modifying enzymes, and any combination thereof. In this context, AAC may include AAC (3) -I (e.g., AAC (3) Ia, AAC (3) Ic), AAC (3) -II (e.g., AAC (3) IId), AAC (3) -III, AAC (3) -IV, AAC (3) -VI, AAC (6 ') -I, AAC (6 ') -II, AAC (6 ') Ie-APH (2 ') Ia, and AAC (2 ' -I). APHs can include APH (2 ') -I, APH (3 ') -I (e.g., APH (3 ') Ia), APH (3 ') -II (e.g., APH (3 ') IIa, APH (3 ') IIb), APH (3 ') -III (e.g., APH (3 ') IIIa), APH (3 ') -IV, APH (3 ') -V, APH (3 ') -VI (e.g., APH (3 ') VIa), APH (3 ') -VII, APH (3 ') -I (e.g., APH (3 ') Ib), and APH (6) -I (e.g., APH (6) Ic, APH (6) Id). ANTs may include ANT (2 ') I (e.g., ANT (2') Ia), ANT (3 ') I (e.g., ANT (3') Ia), ANT (4 ') -I, ANT (4') -II, and ANT (6) -I. Preferably, the AME present in the bacterial species against which a compound of the invention exhibits activity may be selected from APH (3 ') IIIa, APH (3') Ia, APH (3 ') IIb, APH (3') VIa, APH (3 ') Ib, APH (6) Ic, APH (6) Id, AAC (6') Ib, AAC (3) III, AAC (3) IV, AAC (3) Ia, AAC (3) Ic, AAC (3) IId, ANT (2 ') Ia, ANT (3') Ia and AAC (6 ') Ie-APH (2') Ia, most preferably from APH (3 ') IIIa, APH (3') Ia, AAC (3) III, AAC (3) IV and AAC (6 ') Ie-APH (2') Ia.

Accordingly, the present invention further provides a compound of the invention, a pharmaceutically acceptable salt thereof or a composition comprising at least one compound of the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of at least in part by expression of one or more AME, in particular by expression of one or more compounds selected from the group consisting of AAC (3) -I, AAC (3) -II, AAC (3) -III, AAC (3) -IV, AAC (3) -VI, AAC (6 ') -I, AAC (6 ') -II, AAC (6 ') -APH (2 "), AAC (2 ' -I), APH (2") -I, APH (3 ') -I, APH (3 ') -II, APH (3 ') -III, APH (3 ') -IV, APH (3 ') -V, APH (3 ') -VI, APH (3 ') -VII, APH (3 ") -I, APH (3") -II, Microbial infection by bacteria of the group consisting of APH (6) -I, ANT (2 ') -I, ANT (3') -I, ANT (4 ') -I, ANT (4') -II, ANT (6) -I, more particularly one or more AMEs selected from the group consisting of APH (3 ') IIIa, APH (3') Ia, AAC (3) OOO, AAC (3) IV and AAC (6 ') Ie-APH (2') Ia. The AME expressed in the bacteria causing at least in part a microbial infection treated by the above compounds, pharmaceutically acceptable salts or compositions may also be selected from the group consisting of APH (3 ') IIIa, APH (3 ') Ia, APH (3 ') IIa, APH (3 ') IIb, APH (3 ') VIa, APH (3 ') Ib, APH (6) Ic, APH (6) Id, AAC (6 ') Ib, AAC (3) III, AAC (3) IV, AAC (3) Ia, AAC (3) Ic, AAC (3) IId, ANT (2 ') Ia, ANT (3 ') Ia, AAC (6 ') Ie-APH (2 ') Ia, or any combination thereof. The bacterial species expressing such AME may be any of the bacterial species described herein, in particular escherichia coli.

The compounds of the invention, pharmaceutically acceptable salts thereof, or compositions comprising the compounds of the invention or pharmaceutically acceptable salts thereof are also useful for treating microbial infections caused, at least in part, by bacteria exhibiting other resistance mechanisms, e.g., other efflux and/or target modification enzymes. Examples of such target-modifying enzymes include 16S RNA methylases (e.g., armA, rmtA, rmtB, etc.).

The compounds of formula (I), pharmaceutically acceptable salts thereof, or compositions comprising at least one compound of formula (I) or pharmaceutically acceptable salt thereof, are also useful for treating at least in part a bacterial species selected from the group consisting of Escherichia species, enterococcus species, Staphylococcus species, Klebsiella species, and Acinetobacter species. Bacterial infections caused by bacterial species of the group consisting of Pseudomonas species, Enterobacter species, Mycobacterium species, Morganella species, providencia species, and any combination thereof. The species of the genus escherichia coli may be escherichia coli, in particular a wild-type escherichia coli or a strain of escherichia coli expressing one or more aminoglycoside modifying enzymes as defined above. The species of enterococcus may be enterococcus faecium or enterococcus faecalis, preferably enterococcus faecalis. The Staphylococcus species may be Staphylococcus aureus and the Klebsiella species may be Klebsiella pneumoniae. The species of Acinetobacter may be Acinetobacter baumannii. The species of pseudomonas may be pseudomonas aeruginosa. The species of enterobacter may be enterobacter cloacae. The Morganella species can be Morganella morganii. The species of the genus providencia may be providencia stuartii. The strain of each of said species and bacteria may be a non-drug resistant strain or a strain expressing one or more aminoglycoside modifying enzymes as defined elsewhere herein.

The compounds of the present invention, pharmaceutically acceptable salts thereof, or compositions comprising at least one compound of the present invention or pharmaceutically acceptable salts thereof, are also useful for treating bacterial infections, wherein the bacterial infection is caused at least in part by a bacterial species that exhibits resistance to at least one member of the existing regulatory-approved antibiotic family. As used in this context and elsewhere herein, "existing regulatory approved antibiotics" (also referred to herein as "regulatory approved antibiotics") should be understood to mean existing antibiotics that have been approved by at least one national, regional, or international regulatory agency responsible for permitting the use of the drug, or at least antibiotics used in the corresponding country, region, or international region, at the application date of the present application or at the priority date of the claims for priority. The regulatory approved antibiotic to which the bacterial species exhibits resistance may be selected from the group consisting of penicillin antibiotics, cephalosporin antibiotics, carbapenem antibiotics, monobactam antibiotics, polymyxin antibiotics, rifamycin antibiotics, lipiarmycin antibiotics, quinolone antibiotics, sulfonamide antibiotics, macrolide antibiotics, lincosamide antibiotics, tetracycline antibiotics, aminoglycoside antibiotics, lipopeptide antibiotics, glycylcycline antibiotics, glycopeptide antibiotics, oxazolidinone antibiotics, and lipiarmycin.

Examples of existing regulatory approved antibiotics include penicillin G, penicillin K, penicillin N, penicillin O, penicillin V, methicillin, nafcillin, oxacillin, cloxacillin, dicloxacillin, flucloxacillin, ampicillin, amoxicillin, pivampicillin, patatin, methicillin, oxacillin, carbenicillin, ticarcillin, temocillin, mezlocillin, piperacillin, clavulanic acid, sulbactam, tazobactam, and the like.

Examples of currently available regulatory approved cephalosporins include cefazolin, cephalexin, cefadroxil, cefapirin, cefazedone, cefradine, cephradine, cefadroxil, ceftezole, cefoletirizine, cefaclor, ceftazidime, cephalothin, cefterazine, cefaclor, cefotetan, cefoxitin, cefprozil, cefuroxime axetil, cefamandole, cefminox, cefonicid, ceforanide, cefotiam, cefpezil, cefmetazole, chlorocefixime, cefixime, ceftriaxone, ceftazidime, cefoperazone, cefdinir, cefcapene, cefixime, cefepime, cefixime, cefpiramide, cefpodoxime, cefteram, cefetamet, cefdizime, cefepime, Cefsulodin, cefteram, cefoselin, flomoxef, Laloxacillus, cefepime, cefozopran, cefpirome, cefquinome, ceftaroline fosamil, ceftizoxime, ceftibuten, ceftiofur, cefquinome, and cefovivax.

Examples of existing regulatory approved carbapenem antibiotics include imipenem, doripenem, meropenem, ertapenem, faropenem, and biapenem.

Examples of existing regulatory approved monobactam antibiotics include aztreonam.

Examples of existing regulatory approved polymyxin antibiotics include polymyxin B and polymyxin E (colistin).

Examples of currently regulatory approved rifamycin antibiotics include rifamycin B, rifamycin SV, rifampin, rifabutin, and rifaximin.

Examples of lipiarmycin antibiotics approved by existing regulatory agencies include lipiarmycin a and lipiarmycin B.

Examples of currently available regulatory approved quinolone antibiotics include ciprofloxacin, fleroxacin, lomefloxacin, nadifloxacin, norfloxacin, ofloxacin, pefloxacin, rufloxacin, balofloxacin, glafloxacin, levofloxacin, pazufloxacin, sparfloxacin, temafloxacin, clinafloxacin, gatifloxacin, moxifloxacin, sitafloxacin, prulifloxacin, besifloxacin, delafloxacin, and ozafloxacin.

Examples of existing regulatory approved sulfa antibiotics include mafenide, sulfacetamide, sulfadiazine, sulfadoxine, sulfamethylthiazole, sulfamethoxazole, benzenesulfonamide, sulfasalazine, and sulfisoxazole.

Examples of currently regulatory approved macrolide antibiotics include azithromycin, boramycin, clarithromycin, dirithromycin, erythromycin, fluoromycin, josamycin, midecamycin, mehmycin, oleandomycin, rotamycin, roxithromycin, spiramycin, tranudomycin, and tylosin.

Examples of currently available regulatory approved lincosamide antibiotics include lincomycin, clindamycin, and pirlimycin.

Examples of existing approved regulatory tetracycline antibiotics include doxycycline, chlortetracycline, lomycycline, demeclocycline, lymecycline, meclocycline, methacycline, minocycline, omacycline, oxytetracycline, papcycline, rolitetracycline, sarracecycline, tetracycline, and elafin.

Examples of aminoglycoside antibiotics approved by existing regulatory agencies include gentamicin, tobramycin, amikacin, puromycin, streptomycin, neomycin, and paromomycin, apramycin, arbekacin, and dibekacin.

Examples of currently approved lipopeptide antibiotics include daptomycin.

Examples of current regulatory agency approved glycylcycline antibiotics include tigecycline.

Examples of currently regulatory approved glycopeptide antibiotics include vancomycin.

Examples of oxazolidinone antibiotics approved by the current regulatory authorities include linezolid and tedizolid.

The compounds of formula (I), pharmaceutically acceptable salts thereof, or any composition disclosed herein that includes at least one compound of formula (I), pharmaceutically acceptable salts thereof, can be used to treat any bacterial infection. Specific examples of such bacterial infections include one or more of infections and infectious diseases selected from the list consisting of respiratory tract infections, complicated skin and soft tissue infections, complicated abdominal cavity infections, community-acquired pneumonia, hospital-acquired pneumonia, ventilator-associated pneumonia, urinary tract infections, bacterial meningitis, infectious endocarditis, septicemia, osteomyelitis, septic arthritis, septicemia, anthrax, osteomyelitis, tuberculosis, leprosy, necrotizing fasciitis, scarlet fever, rheumatic fever, postpartum fever, and strep toxic shock syndrome, and additional nosocomial infections, such as infections caused by the use of intravascular catheters.

The present invention also provides a method of treating a disease or condition in which a microorganism plays a role, said method comprising the step of administering to a subject in need thereof, said subject being a human or animal, in particular a mammal, more in particular a human, a therapeutically effective amount of a compound of formula (I), a pharmaceutically acceptable salt thereof or a composition comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof as defined herein. Thus, when reference is made herein to a compound of formula (I), a pharmaceutically acceptable salt thereof, or a composition comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof, for use as a medicament or for the treatment of a particular type of infection, disorder, affliction or disease, this may also encompass a method of treating said infection, disorder, affliction or disease, comprising the step of administering to a subject in need thereof, which is a human or animal, in particular a mammal, more particularly a human, a therapeutically effective amount of a compound of formula (I), a pharmaceutically acceptable salt thereof, or a composition comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof.

Accordingly, the present invention also provides a method of treating a microbial infection, said method comprising the step of administering to a subject in need thereof, said subject being a human or animal, in particular a mammal, more particularly a human, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.

The compounds of the present invention may exhibit activity against microbial infections caused by gram-positive and/or gram-negative bacteria and are therefore useful in the treatment of microbial infections caused by gram-positive and/or gram-negative bacteria. The compounds of the invention, pharmaceutically acceptable salts thereof or compositions comprising the compounds of the invention or pharmaceutically acceptable salts thereof are useful for treating one or more members selected from the group consisting of, or at least partially consisting of, species of Escherichia coli, species of enterococcus, species of Staphylococcus, species of Klebsiella, species of Acinetobacter, species of Pseudomonas, and species of Enterobacter, species of Mycobacterium, species of Morganella, species of providencia, and any combination thereof, particularly, one or more members selected from the group consisting of wild-type Escherichia coli, Escherichia coli APH (3 ') IIIa, Escherichia coli APH (3') Ia, Escherichia coli AAC (6 ') -APH (2'), Escherichia coli AAC (6 ') Ib, Escherichia coli AAC (3) III, Escherichia coli AAC (3) IV, Escherichia coli APH (3') IIb, Escherichia coli APH (3 ') VIa, Escherichia coli AAC (6') III, Escherichia coli AAC (3) IV, Escherichia coli APH (3 ') IIb, Escherichia coli APH (3') VIA), Coli APH (3 ') Ib, E.coli APH (6) Ic, E.coli APH (6) Id, E.coli AAC (3) Ia, E.coli AAC (3) Ic, E.coli AAC (3) IId, E.coli ANT (2 ') Ia, E.coli ANT (3 ') Ia, E.coli AAC (6 ') Ie-APH (2 '), enterococcus faecalis, staphylococci, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter cloacae, Morganella morganii, providencia stuartii, any bacterium expressing any one of the AMEs disclosed elsewhere herein, and any combination thereof. For example, the bacterial infection may be caused, at least in part, by one or more of Acinetobacter baumannii, Escherichia coli, Morganella morganii, and providencia stuartii, wherein each of the bacteria expresses an AME independently selected from one or more of AAC (3) Ia, ANT (3 ') Ia, APH (3 ') Ib, APH (3 ') VI, APH (6) Id, ANT (2 ') Ia, APH (3 ') -VIa, APH (3 ') -Ia, AAC (3) -IId, ANT (3 '), APH (3 ') -IIa, APH (3 ') -IIb, APH (6) -Ic, and AAC (3) -Ic. Examples thereof include Acinetobacter baumannii expressing one or more AMEs selected from AAC (3) Ia, ANT (3 ') Ia, APH (3') Ib, APH (3 ') VI, and APH (6) Id (e.g., Acinetobacter baumannii expressing AAC (3) Ia, ANT (3') Ia, APH (3 ') Ib, APH (3') VI, and APH (6) Id; or Acinetobacter baumannii expressing ANT (2 ') Ia, APH (3') Ib, APH (3 ') -VIa, and APH (6) -Id; further examples thereof include E.coli expressing one or more AMEs selected from ANT (3') -Ia, and APH (3 ') -Ia (e.coli expressing APH (3') -Ia; or E.coli expressing ANT (3 ') -Ia and APH (3') -Ia); further examples thereof include E., Morganella morganii of one or more AMEs of ANT (3 '), APH (3') -Ib, APH (3 ') -Ia, and APH (6) -Id (e.g., Morganella morganii expressing AAC (3) -IId, ANT (3'), APH (3 ') -Ib, APH (3') -Ia, and APH (6) -Id). Further examples thereof include providencia stuartii expressing one or more AMEs selected from AAC (3) -Ic, ANT (3 ') Ia, ANT (3'), APH (3 ') -IIa, APH (3') -IIb, APH (3 ') -VI, and APH (6) -Ic (e.g., providencia stuartii expressing AAC (3) -Ic, ANT (3') Ia, ANT (3 '), APH (3') -IIa, APH (3 ') -IIb, APH (3') -VI, and APH (6) -Ic).

The compounds of the invention and their pharmaceutically acceptable salts exhibit particularly high potency and cross-group activity against microbial infections caused at least in part by bacteria of the ESKAPE group and carbapenem-resistant enterobacteriaceae (CRE) infections. Accordingly, the compounds of the present invention, or pharmaceutically acceptable salts thereof, may be used to treat microbial infections and/or carbapenem-resistant enterobacteriaceae infections caused at least in part by bacteria from the ESKAPE group. Accordingly, the present invention also relates to a method of treating a subject suffering from a microbial infection and/or a carbapenem-resistant enterobacteriaceae infection caused at least in part by a bacterium of the ESKAPE group, comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof. In particular, the compounds of the present invention, or pharmaceutically acceptable salts thereof, may be used to treat microbial infections caused at least in part by enterobacter cloacae. When used to treat a carbapenem-resistant enterobacteriaceae infection, the compounds of the present invention, or pharmaceutically acceptable salts thereof, can be used to treat a carbapenem-resistant enterobacteriaceae infection caused, at least in part, by at least one of escherichia coli, klebsiella pneumoniae, and enterobacteriaceae.

The compounds of the invention or pharmaceutically acceptable salts thereof also exhibit excellent cross-group activity against clinical isolates of selected multi-drug resistant bacteria of the ESKAPE group and enhanced activity against enterococcus faecalis. The compounds of the present invention, or pharmaceutically acceptable salts thereof, are particularly useful for treating microbial infections caused at least in part by klebsiella pneumoniae, acinetobacter baumannii, pseudomonas aeruginosa, or any combination thereof, including MDR microbial infections caused at least in part by klebsiella pneumoniae, acinetobacter baumannii, pseudomonas aeruginosa, or any combination thereof. Examples thereof include infections caused at least in part by klebsiella pneumoniae strains expressing carbapenemases such as OXA type carbapenemases (e.g., OXA-48, etc.), NDM type carbapenemases, VIM type carbapenemases, and/or Klebsiella Pneumoniae Carbapenemases (KPC), and/or broad-spectrum beta-lactamases (ESBL). A single klebsiella pneumoniae strain can express more than one drug resistance causing enzyme. Further examples thereof include infections caused at least in part by strains of klebsiella pneumoniae which exhibit colistin resistance. The klebsiella pneumoniae bacterial strain can also have drug resistance to one or more of amikacin, gentamicin, tobramycin, ceftazidime and meropenem. Further examples thereof include infections caused at least in part by acinetobacter baumannii strains that are resistant to carbapenems (so-called carbapenem-resistant acinetobacter baumannii-CRAB) and MDR acinetobacter baumannii strains. The acinetobacter baumannii strain may also be resistant to one or more of amikacin, gentamicin, tobramycin, ceftizoxime and meropenem in particular. Further examples thereof include infections caused at least in part by strains of pseudomonas aeruginosa, and MDR pseudomonas aeruginosa strains expressing carbapenemases, such as an OXA type carbapenemase (e.g., OXA-2), a VIM type carbapenemase. The pseudomonas aeruginosa may also be resistant to one or more of amikacin, gentamicin, tobramycin, ceftazidime and meropenem in particular. Further examples thereof include infections caused at least in part by methicillin-resistant staphylococcus aureus (MRSA) strains.

The compounds of the invention or pharmaceutically acceptable salts thereof are particularly useful for treating microbial infections caused at least in part by multidrug-resistant, particularly aminoglycoside-resistant (e.g., amikacin, gentamicin, and/or tobramycin), or pan-resistant, strains of E.coli.

The compounds of the invention or pharmaceutically acceptable salts thereof are particularly useful for treating microbial infections caused at least in part by multidrug-resistant, particularly aminoglycoside-resistant (e.g., amikacin, gentamicin, and/or tobramycin) or pan-resistant, enterococcus faecalis strains.

The compounds of the present invention or pharmaceutically acceptable salts thereof, or any composition disclosed herein that includes at least one compound of the present invention or pharmaceutically acceptable salt thereof, are particularly useful for treating microbial infections caused, at least in part, by vancomycin-resistant enterococcus faecium (VRE).

The compounds of the invention or pharmaceutically acceptable salts thereof or any composition disclosed herein comprising at least one compound of the invention or pharmaceutically acceptable salt thereof are particularly useful for treating microbial infections caused at least in part by strains of morganella morganii, in particular multidrug-resistant (in particular to aminoglycosides (e.g. amikacin, gentamicin and/or tobramycin), or pan-resistant).

The compounds of the invention or pharmaceutically acceptable salts thereof or any composition disclosed herein comprising at least one compound of the invention or pharmaceutically acceptable salt thereof are particularly useful for treating microbial infections caused at least in part by providencia stuartii strains, in particular multidrug-resistant (in particular to aminoglycosides (e.g. amikacin, gentamicin and/or tobramycin), or pan-resistant).

The compounds of the invention or pharmaceutically acceptable salts thereof are particularly useful for treating microbial infections caused at least in part by multidrug-resistant, particularly aminoglycoside-resistant (e.g., amikacin, gentamicin, and/or tobramycin), or pan-resistant, strains of klebsiella pneumoniae.

The compounds of the invention or pharmaceutically acceptable salts thereof are particularly useful for treating microbial infections caused at least in part by multidrug-resistant, particularly aminoglycoside-resistant (e.g., amikacin, gentamicin, and/or tobramycin), or pan-resistant, acinetobacter baumannii strains.

The compounds of the invention or pharmaceutically acceptable salts thereof are particularly useful for treating microbial infections caused at least in part by multidrug-resistant, particularly aminoglycoside-resistant (e.g., amikacin, gentamicin, and/or tobramycin) or pan-resistant, enterobacter cloacae strains.

The compounds of the invention or pharmaceutically acceptable salts thereof are particularly useful for treating microbial infections caused at least in part by multidrug-resistant, particularly aminoglycoside-resistant (e.g., amikacin, gentamicin, and/or tobramycin) or pan-resistant, pseudomonas aeruginosa strains.

The compounds of the invention or pharmaceutically acceptable salts thereof are particularly useful for treating microbial infections caused at least in part by multidrug-resistant, particularly aminoglycoside-resistant (e.g., amikacin, gentamicin, and/or tobramycin) or pan-resistant, strains of staphylococcus aureus.

In one embodiment, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment of a medical condition, i.e. for use as a medicament or agent, in particular for the treatment of a microbial infection and/or an infectious disease. Accordingly, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment of a microbial infection and/or a condition, affliction or disease caused, at least in part, by a microbial infection. Accordingly, the present invention also provides a method of treating a patient suffering from a microbial infection and/or a condition, affliction or disease caused, at least in part, by a microbial infection, which comprises administering to said patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof. Thus, the subject to be treated is a human or an animal, in particular a mammal, more particularly a human. The microbial infection may be, and preferably is, a bacterial infection. Such bacterial infection may be caused, at least in part, by one or more gram-positive bacteria and/or one or more gram-negative bacteria. Such gram-positive species may be one or more species selected from the group consisting of staphylococcus aureus, streptococcus pneumoniae, enterococcus faecalis, enterococcus faecium, mycobacterium tuberculosis. Such gram-negative species may be selected from the group consisting of Klebsiella species, Escherichia species, Acinetobacter species. Morganella species, providencia species, Enterobacter species. Thus, the bacterial infection may be caused, at least in part, by a species selected from the group consisting of an escherichia species, an enterococcus species, a staphylococcus species, a klebsiella species, an acinetobacter species, a pseudomonas species, an enterobacter species, a mycobacterium species, a morgana species, a providencia species, or a combination thereof. In this context, the species of the genus Escherichia coli may be Escherichia coli, preferably selected from the group consisting of wild-type Escherichia coli and a strain of Escherichia coli expressing one or more aminoglycoside modifying enzymes selected from the group consisting of aminoglycoside phosphotransferases, in particular APH (3 ') IIIa and APH (3') Ia, aminoglycoside acetyltransferases, in particular AAC (3) III and AAC (3) IV, and bifunctional aminoglycoside modifying enzymes, in particular AAC (6 ') Ie-APH (2') Ia, or combinations thereof. The aminoglycoside modifying enzyme may further be selected from the group consisting of APH (3 ') IIIa, APH (3 ') Ia, AAC (6 ') -APH (2 '), AAC (6 ') Ib, AAC (3) III, AAC (3) IV, APH (3 ') IIa, APH (3 ') IIb, APH (3 ') VIa, APH (3 ') Ib, APH (6) Ic, APH (6) Id, AAC (3) Ia, AAC (3) Ic, AAC (3) IId, ANT (2 ') Ia, ANT (3 ') Ia, AAC (6 ') Ie-APH (2 ') Ia, and any combination thereof. In this context, the enterococcus species may be enterococcus faecium or enterococcus faecalis, preferably enterococcus faecalis, the Staphylococcus species may be Staphylococcus aureus, the Klebsiella species may be Klebsiella pneumoniae, the Acinetobacter acinetobacter baumannii, the Pseudomonas species may be Pseudomonas aeruginosa, the Morganella species may be Morganella morganii, the providencia species may be providencia stuartii, and the Enterobacter species may be Enterobacter cloacae. Any of these bacteria may be a non-drug resistant strain or a strain expressing one or more of the AME described above. In this context, the bacterial infection may be caused at least in part by a bacterial species that exhibits resistance to at least one member of an existing regulatory-approved antibiotic family, in particular to a bacterial species that exhibits resistance to at least one member of an antibiotic family selected from the group consisting of penicillins, cephalosporins, carbapenems, monobactams, polymyxins, rifamycins, lipiarmycins, quinolones, sulfonamides, macrolides, lincosamides, tetracyclines, aminoglycosides, lipopeptides, glycylcyclines, glycopeptides, oxazolidinones, and lipiarmycins. In this context, the bacterial infection may be one or more of an infection and an infectious disease selected from the list consisting of respiratory tract infection, complicated skin and soft tissue infection, complicated abdominal cavity infection, community-acquired pneumonia, hospital-acquired pneumonia, ventilator-associated pneumonia, urinary tract infection, bacterial meningitis, infectious endocarditis, sepsis, osteomyelitis, septic arthritis, sepsis, anthrax, osteomyelitis, tuberculosis, leprosy, necrotizing fasciitis, scarlet fever, rheumatic fever, postpartum fever, and streptococcal toxic shock syndrome, and further nosocomial infections, such as infections caused by the use of intravascular catheters.

A common disadvantage associated with known aminoglycoside antibacterial agents such as gentamicin is their small volume of distribution when administered in vivo. This low level of tissue distribution limits the applicability of conventional aminoglycoside antibacterial agents such as gentamicin in the treatment of microbial infections requiring high tissue penetration (e.g., pneumonia, cystic fibrosis, skin infections, and soft tissue infections). Consequently, aminoglycosides are not generally used as the first antimicrobial agents to treat such infections. It is therefore another object of the present invention to provide compositions having improved tissue distribution (as measured by, for example, pharmacokinetic parameters) relative to conventional aminoglycoside antimicrobials, such as gentamicinVolume of cloth (V)d) Measured) aminoglycoside antimicrobial agent. In this regard, the compounds of the present invention exhibit significantly increased volume of distribution over conventional aminoglycoside antimicrobials, such as gentamicin, the increase typically being in the range of 2 to 5 fold. Accordingly, the compounds of the present invention, pharmaceutically acceptable salts thereof, or any composition described herein comprising at least one compound of the present invention, or pharmaceutically acceptable salt thereof, can be used to treat a bacterial infection and/or a condition, affliction, or disease caused, at least in part, by a bacterial infection, wherein the treatment requires a high degree of tissue penetration, particularly in the treatment of pneumonia, cystic fibrosis, skin infection, or soft tissue infection, particularly in the treatment of pneumonia.

It is another object of the present invention to provide an aminoglycoside antibacterial agent, at least one family or strain of bacteria exhibiting a reduced tendency to develop resistance as compared to other known aminoglycoside antibacterial agents, for example amikacin, gentamicin or any other aminoglycoside antibacterial agent described herein. Various bacterial strains exhibit a reduced ability to develop resistance to the compounds of the invention relative to known aminoglycosides, e.g., amikacin or gentamicin. This effect has been observed in several in vitro resistance studies with serial passages for 30 days, in particular in acinetobacter baumannii strains, klebsiella pneumoniae strains (including gentamicin-resistant clinical isolates) and escherichia coli strains.

The compounds of the present invention, or pharmaceutically acceptable salts thereof, may be used to treat a bacterial infection and/or a condition, affliction, or disease caused, at least in part, by a bacterial infection, particularly wherein the bacterial infection is caused by any one of the bacterial strains disclosed herein (or any combination of any of them), or by data directed thereto as provided in the experimental section herein.

Another aspect of the invention relates to a method of preventing, inhibiting or stopping the growth of bacteria on a surface comprising the step of applying at least one compound of the invention to said surface. The compounds of the invention may be applied to the surface directly or in other forms, such as solutions. Thus, the compounds of the present invention may be formulated as bactericides, disinfectants, or antimicrobials and used, for example, to prevent, inhibit, or stop the growth of bacteria on a surface.

These surfaces may constitute hard surfaces, e.g. floors, countertops, bathroom surfaces, kitchen surfaces, stone surfaces, wood surfaces, concrete surfaces, ceramic surfaces, plastic surfaces, glass surfaces, crockery, tableware, pots, trays, equipment such as household equipment or medical equipment including contact lenses, or soft surfaces, e.g. skin, hair, clothing, contact lenses, etc. Thus, the compounds of the present invention may be formulated and used as disinfectants, sanitizers, and the like, and/or as components, additives, or preservatives for medical/surgical devices, disinfectants, soaps, shampoos, hand washes, denitrifiers, household cleaning formulations, laundry and dish washing agents, in cleaning and treatment solutions for topical use, devices and apparatus including contact lenses, and other disinfecting and antimicrobial applications.

Thus, the invention also relates to the use of the compounds of the invention in preventing, inhibiting or stopping the growth of bacteria on surfaces, including both hard surfaces, such as floors, countertops, bathroom surfaces, kitchen surfaces, stone surfaces, wood surfaces, concrete surfaces, ceramic surfaces, plastic surfaces, glass surfaces, crockery, tableware, pots, dishes, equipment such as household equipment or medical equipment including contact lenses, or soft surfaces, such as skin, hair, clothing, contact lenses and the like.

As used herein, the term "comprising" should be understood to also encompass alternatives in which the product/method/use to which the term "comprising" is applied may also "consist exclusively of the subsequently described elements.

As used herein, the term "comprising" should be understood to also encompass alternatives in which a product/method/use in connection with the use of the term "comprising" may also "consist essentially of" the subsequently described elements.

Unless otherwise indicated, each chemical or composition referred to herein should be interpreted as being a commercial grade material, which may include isomers, by-products, derivatives, and other such materials that are normally understood to be present in the commercial grade. However, unless otherwise specified, the amount of each chemical component does not include any solvent or diluent oil, which may typically be present in commercial materials.

Unless otherwise indicated, all synthetic methods and parameter measurements are understood to be carried out at room/ambient temperature, i.e. at 21 ± 1 ℃.

The term "percent" or "%" as used with respect to the amount of material refers to weight percent (i.e., (w/w)) unless otherwise indicated.

As used in this application, the word "or any combination thereof" should be understood to mean that all possible combinations of the list members immediately preceding the word are directly and explicitly disclosed in the list. This expression represents a simple means of expressing the corresponding text (specification or claims) in as concise a manner as possible as required by the patent laws, clearly setting forth what is to be understood and distinctly understood that each and every possible combination resulting therefrom is directly and unequivocally disclosed. For example, the list "selected from A, B, C and any combination thereof" should be understood to disclose directly and explicitly a single list of the following alternatives: A. b, C, A + B, A + C, B + C, A + B + C.

Since the numbers, values, and/or expressions of all specified materials, ingredients, reaction conditions, molecular weights, numbers of carbon atoms, and the like, used herein and in the appended claims are affected by the various uncertainty encountered in obtaining such values, unless otherwise specified, all such values are to be understood as being modified in all instances by the term "about" as used herein. As used herein, the term "about" used in connection with a numerical value should be understood to be within the degree of error of the instrumentation typically used by those of ordinary skill in the art to measure the value in the context of the present disclosure, and more particularly, within the range of values where no discernible function or property is affected so as to differ in the same function or property that is precisely displayed under the value. The term "about" used in conjunction with a numerical value may mean that the value is ± 20%. The term "about" used in conjunction with a numerical value may mean that the value is ± 10%. The term "about" used in conjunction with a numerical value may mean that the value is ± 5%. The term "about" used in conjunction with a numerical value may mean that the value is ± 1%. The term "about" used in conjunction with a numerical value may mean that the value is ± 0.5%.

In the present invention, certain R groups may be substituted or unsubstituted phenyl or 5 or 6 membered heteroaryl (as defined in the claims and/or the description). Thus, these groups are aromatic. In the context of the present invention, "aromatic" means that the corresponding ring is present in aromatic form (in the case of tautomers, in aromatic tautomeric form) at ambient (room temperature) (21 ± 1 ℃) and atmospheric pressure (101.325kPa), preferably to an extent of at least 10, 20, 30, 40, 50, 60, 70, 80, or 90%. Preferably, "aromatic" means that the corresponding ring is present in aromatic form to an extent of at least 95%, more preferably at least 99%, still more preferably at least 99.5% or at least 99.9% at room temperature and atmospheric pressure.

During the synthesis of the compounds of formula (I), labile functional groups such as hydroxyl, carboxyl and amino groups in intermediate compounds may be protected. The protecting group may be removed at any stage of the synthesis of the compound of formula (I) or may be present on the final compound of formula (I). A comprehensive discussion of the manner in which various labile functional groups can be protected and the methods by which the resulting derivatives can be cleaved is found, for example, in Protective groups in Organic Synthesis, T.W.Greene and P.G.M Wuts, (2006) John Wiley&Sons,Inc.,New York,4th edition。

The preferred groups described herein for compounds of formula (I) also apply to the variant compounds.

Although the preferred groups for each variant are listed above separately for each variant, preferred compounds of the invention include those in which several or each variant of formula (I) is selected from the preferred groups for each variant. Thus, the present invention is intended to include all combinations of the groups listed as preferred.

All publications, including but not limited to patents and patent applications cited in this specification, are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth. The invention will now be described with reference to the following examples, which are for the purpose of illustration and are not to be construed as limiting the scope of the invention.

The compounds of the present invention can be synthesized using the synthetic procedures and methods outlined herein. For example, the synthetic procedures and methods outlined in the examples section of this application illustrate the synthesis of exemplary compounds of the invention, and these methods and schemes may be used to obtain other compounds of the invention. It will be clear to those skilled in the art of organic chemistry that the synthetic route may have to be changed, or the order of the synthetic steps changed, in order to most easily introduce the necessary functional groups to provide the particular molecules of the present invention. Modifications necessary to the synthetic routes explicitly set forth herein, for example, providing specific groups to provide reagents required for the compounds of the invention, and the like, are within the knowledge of those skilled in the art.

For example, the compounds of the present invention may be obtained from compounds of the following starting formula:

starting from a compound of the above formula (e.g. tobramycin, kanamycin a or kanamycin B), one method of obtaining a compound of the invention may comprise the steps of:

1) protecting group strategies based on known chemistry are used (e.g., in Protective Groups in Organic Synthesis, T.W.Greene and P.G.M Wuts, (2006) John Wiley &Sons,Inc.,New York,4thedition) and use of the known reactivity of the various heteroatoms in the above compounds to give the only unprotected NH therein2Compounds in which the radical is in position 1. An example of such a protecting group strategy may be the protection of 3-, 2 '-and 6' -NH, when present, with a tert-butyloxycarbonyl (Boc) protecting group2Group and protection of 3' -NH with an orthogonal protecting group such as trifluoromethylacetoxy2A group;

2) using known chemical methods, R is4In the final form in which the radicals are defined for the compounds of the formula (I) or in the appropriate form known to the person skilled in the artIntroduction of protected form into unprotected NH2In position 1 of the group, to enable the remainder of the synthetic procedures required to provide the compounds of the invention;

3) compounds in which the only unprotected OH group is the OH group at the 5 position are prepared using known protecting group chemistry. For example, this may include protecting all other hydroxyl groups in the molecule to their respective acetates (using, for example, acetic anhydride and a suitable base);

4) substitution of the OH at position 5 with an F atom using known chemistry for such fluorination reactions, in particular using fluorinating agents such as X-TalFluor-E (also known as tetrafluoroboric acid (diethylamino) difluorosulfonium);

5) Selective removal of NH at the 3' position2N-protecting groups of radicals, so as to be the only free NH2The groups are groups at the positions, and the OH protecting groups are optionally removed simultaneously under the same reaction conditions. NH when in the 3' position2When the group is protected with a trifluoromethyl acetate and the OH group is protected as an ester, such as its respective acetate, this can be achieved using, for example, nucleophilic basic conditions such as methoxide (e.g., sodium methoxide) in a suitable solvent (e.g., methanol);

6) free NH at position 3' using known chemical methods2The groups reacting to introduce the desired free radical (R) at the 3' position5H) or substituted/derived (R)5Not equal to H; comprising R with protected amino and/or hydroxy functions5Residue) guanidino radical, NH2The group is converted to the corresponding free or substituted/derivatized guanidine. Suitable chemistry for establishing the desired guanidino group may include the free 3 "-NH2Reaction of the group with a suitable N, N '- (bis-protected) -1-guanidinopyrazole, for example N, N' -bis-Boc-1-guanidinopyrazole;

7) using known chemical methods (e.g., in Protective Groups in Organic Synthesis, T.W.Greene and P.G.M Wuts, (2006) John Wiley&Sons,Inc.,New York,4thAs taught in edition) to provide a compound of the invention or salt thereof.

When other functional groups or moieties are to be incorporated into the inventionWhen a compound is present (e.g. when R is1When R is not H3To OH or ether moieties, etc.), the other synthetic steps required to introduce these moieties and ultimately achieve the final compounds of the invention may again be based on chemistry well known to the skilled artisan. Examples of such chemistries are given in the synthesis section herein. These additional steps may, for example, be incorporated into the basic methods outlined above (steps 1) through 7)), the order of the steps in the resulting methods being selected according to the exact chemistry to be performed and belonging to synthetic route planning routines well known to the skilled person. For example, introduction of OH groups or ethers as R3Can be prepared by first oxidizing the 3' -OH group to the corresponding carbonyl group, and then contacting the carbonyl group with a suitable nucleophile (e.g., when R is2H is hydride; when R is2An organometallic compound suitable for an alkyl group such as a grignard reagent). Then, R is obtained3The OH groups can be further carried out using known chemical methods to produce, inter alia, 3' -ether compounds. The oxidation of the above-mentioned 3' -OH group to the corresponding carbonyl group can generally be effected directly after the above-mentioned step 2), since the OH group can generally be selectively oxidized without protecting the OH groups remaining in the molecule (suitable oxidation conditions for this conversion can be found, for example, in WO 2013/195149 a 1). The subsequent reaction of the carbonyl group is preferably carried out after step 3 and before deprotection of the resulting protected OH group, with, for example, a hydride (carbonyl reduction), or with, for example, a nucleophilic organometallic reagent before step 3), since free OH groups may in some cases, for example depending on the particular reagents used and their associated reactivity, interfere with the desired carbonyl reduction/organometallic addition (this synthesis scheme is routine for the skilled person in organic chemistry). However, in general, either the reduction of the carbonyl group with sodium borohydride or the addition of a Grignard reagent to the carbonyl group can be successfully carried out without protecting the hydroxyl groups present.

Similarly, conventional synthesis plans may be used to synthesize R1Group (wherein R1Not H) into the compounds of the invention. This may entail, for example, the use of orthogonal N-protecting groups, so that, for example, after step 1) above, 6' -NH2Radical with NH2Different (and orthogonal) protecting groups are protected at the 2 '-position, 3-position and 3 "-position (e.g. 6' -NH protected with a carboxybenzyl (Cbz) group2Group protection of 2' -NH using Boc group2And 3-NH2Group, and protection of 3' -NH using a trifluoroacetate group2A group). This orthogonality of the protecting groups enables flexible deprotection and further development of the structures at these centers at different stages of the synthetic route. At 6' -NH2Introduction of various R's into the radical1Groups may be achieved using chemical methods well known in the art.

As mentioned above, the general synthetic methods described in the above steps 1) to 7) represent only one possible route for the preparation of the compounds of the invention. Thus, alternative synthetic routes, including those in which the above-described methods are improved in one or more steps, may also provide the compounds of the invention. For example, when NH is in position 12Introduction of R onto radicals4The radicals comprising an amide-forming reaction (NH)2Acylation of the group at position 1), methods known in the art should potentially enable this step with NH at the 3 "-C position 2The group does not need to be protected (i.e. it can be achieved in the presence of other nucleophilic amino groups). For example, 1) PyBOP (benzotriazol-1-yl-oxytripyrrolidinyl hexafluorophosphate) is used) Or 2) a combination of HOMB (N-hydroxy-5-norbornene-2, 3-dicarboximide) and EDC (1-ethyl-3- (3-dimethylaminopropyl) carbodiimide) as an amide coupling agent, together with a suitable acid coupling partner, may allow selective amide formation at the 1-C position above the 3 "-C position (such chemistry and suitable reaction conditions may be found, for example, in US 2013/0217642A 1). This may have the advantage of reducing the total number of steps required in the synthesis. Other alternatives include introducing the guanidine functionality at an earlier stage of the overall synthesis, e.g., prior to introducing the fluorine substituent at the 5-C position. This is well tolerated and can be done throughout the remainder of the synthesis.

Examples of synthetic methods for the compounds of the present invention include each of these two modifications shown in the following reaction schemes.

Examples

Unless otherwise indicated, all synthetic methods and parameter measurements are understood to be carried out at room/ambient temperature, i.e. at 21 ± 1 ℃.

Unless otherwise indicated, each chemical or composition referred to herein should be interpreted as being a commercial grade material, which may include isomers, by-products, derivatives, and other such materials that are normally understood to be present in the commercial grade.

Unless otherwise indicated, all measurements made using the instruments listed or referenced herein should be understood as having been made according to standard procedures for performing such methods, such as those well known in the art or as set forth by the manufacturer of the instrument.

Where the trade name or trademark is used to describe a product mentioned herein, the composition, properties, configuration, etc. of that product should be understood as those of the corresponding product available under that name or trademark on the filing date of this application (as applicable, priority date).

Unless otherwise indicated, all optical density values reported herein were measured at 600nm using uv/vis spectroscopy.

Abbreviations:

CLSI Clinical and Laboratory Standards Institute (Clinical Laboratory Standards Institute)

MIC minimum Inhibitory Concentration (minimum Inhibitory Concentration)

MH Muller-Xindun (Mueller-Hinton)

MH2 Muller-Xindun 2 (Mueller-Hinton 2)

LB Broth Luria-Beltani Broth (Luria Bertani Broth)

CFU Colony Forming Units (Colony Forming Units)

LOD detection Limit (Limit of detection)

OD Optical Density (Optical density)

rH Relative humidity (Relative humidity)

ATCC American Type Culture Collection (American Type Culture Collection)

PBS Phosphate buffer (Phosphate-buffered saline)

DMSO Dimethyl Sulfoxide (Dimethyl Sulfoxide)

DMF Dimethylformamide (dimethyl formamide)

MeOH Methanol (Methanol)

EtOH Ethanol (Ethanol)

EtOAc Ethyl acetate (Ethyl acetate)

DCM Dichloromethane (Dichloromethane)

THF Tetrahydrofuran (Tetrahydrofuran)

LC/MS Liquid chromatography-Mass Spectrometry (Liquid chromatography-Mass Spectrometry)

NMR Nuclear magnetic resonance (Nuclear magnetic resonance)

ATP-linked proton test (Attached proton test)

HSQC Heteronuclear single quantum coherence (Heteronuclear single quantum coherence)

COSY Correlation spectrum (Correlation spectrum)

MS Molecular sieve (Molecular sieve)

KanaA kanamycin A (Kanamycin A)

KanaB kanamycin B (Kanamycin B)

AmikaB Amikacin B (Amikacin B)

Amikaa Amikacin A (Amikacin A)

Tobra Tobramycin (Tobramycin)

AHB (2S) - (-) -4-Amino-2-hydroxybutyric acid ((2S) - (-) -4-Amino-2-hydroxybutanoic acid)

AFB (2S) - (-) -4-Amino-2-fluorobutyric acid ((2S) - (-) -4-Amino-2-fluorobutyric acid)

AHCA (2-aminoethyl) (hydroxy) carbamic acid)

BND N-Benzyloxycarbonyloxy-5-norbornene-2,3-dicarboximide (N-Benzyloxycarbonyloxy-5-norbomene-2, 3-dicarboximide)

TFA Trifluoroacetic acid (Trifluoroacetic acid)

Et3N Triethylamine (Triethylamine)

NaOMe Sodium methoxide (Sodium methoxide)

DIPEA N, N-Diisopropylethylamine (N, N-Diisopropylethylamine)

LDA Lithium diisopropylamide (Lithium diisopropyramide)

Me Methyl (Methyl)

Et Ethyl (Ethyl)

BQ 1,4-Benzoquinone (1,4-Benzoquinone)

NaBH4Sodium borohydride (Sodium borohydrate)

HATU 3-Oxyhexafluorophosphoric acid-1- [ bis (dimethylamino) methylene ] -1H-1,2,3-triazolo [4,5-b ] pyridine (1- [ bis (dimethylamino) methyl ] -1H-1,2,3-triazo [4,5-b ] pyrindinium 3-oxide hexafluorophosphate)

XtalFluor-E Tetrafluoroboric acid (Diethylamino) difluorosulfonium ((diethylenelamino) difluorosulfonium tetrafluoroborate)

HCl Hydrochloric acid (Hydrochloric acid)

Gua Guanidine (Guanidine)

Su Succinimide (Succinimide)

Boc tert-Butyloxycarbonyl (tert-Butyloxycarbonyl)

Cbz Carboxybenzyl (Carboxybenzyl)

TBDMS tert-Butyldimethylsilyl (tert-Butyldimethylsilyl)

Ac Acetate (Acetate)

Tf Triflate (Triflate)

tert (Tertiary)

sec (Secondary)

tRRetention time (Retention time)

RfRetention factor (Retention factor)

ACN Acetonitrile (Acetonitrile)

TSB Tryptic Soybean soup (Tryptic soy broth)

A. Materials and methods

1. Organic synthesis

A method. Using CDCl3、DMSO-d6、MeOD-d4Or D2O as solvent, recorded on a Varian Unity Inova spectrometer (500 MHz and 125.7MHz, respectively) and a Varian AMX400 spectrometer (400,100.59 MHz, respectively)1H、13C. COSY, HSQC spectra.1H and19the F spectra were recorded on a Varian VNMRS NMR 300MHz spectrometer with a 7.05 Tesla magnet equipped with an indirect detection probe from Oxford Instruments, or a Mercury 300 spectrometer with a 7.05 Tesla magnet equipped with 4 nuclear auto-switching probes (300 MHz and 282MHz, respectively) from Oxford Instruments using CDCl3、DMSO-d6、MeOD-d4Or D2O is used as a solvent. Chemical shift values (CDCl) are reported in ppm using solvent resonance as an internal standard31H is delta 7.26, H is beta-beta,13c is delta 77.2; DMSO-d61H is delta 2.50, H is alpha 2.50,13c is delta 39.5; MeOD-d4: the 1H is delta 3.31, the total weight of the catalyst,13c is delta 49.2; d2O:1H is δ 4.80). The data are reported as follows: chemical shift (δ), multiplicities (s ═ singlet, d ═ doublet, t ═ triplet, q ═ quartet, br ═ broad, m ═ multiplet), coupling constants j (hz), and integrals. Liquid chromatography-mass spectrometry (LCMS): the system 1: high performance liquid chromatography-mass spectrometry (HPLC-MS) spectra were recorded on an Agilent 1200 series HPLC system equipped with a degasser (G1379B), a binary pump (G1312B), a multi-column thermostat (G7116A), an autosampler (G1329B), an autosampler thermostat (G1330B), a UV detector (G1315C), an ELSD detector (G7102A), and an Agilent 6130 mass detector (G6130B). And (3) system 2: high performance liquid chromatography-mass spectrometry (HPLC-MS) spectra were recorded on an Agilent 1260Infinity series HPLC system equipped with a degasser (G4225A), binary pump (G1312B), multi-column thermostat (G7116A), autosampler (G1329B), autosampler thermostat (G1330B), UV detector (G4212B), ELSD detector (G4260B), and Agilent 6120 mass detector (G6120B). LCMS method a: utensil for cleaning buttock With a Waters Atlantis T3 column from Krudcatcher (C18 reverse phase silica; 4.6X 100mm, 3 μm); 0.05% TFA (aq)/ACN: 80/20(2.0 min) → (8.0 min) → 10/90(2.0 min) (this description of the LCMS process should be understood to mean 0.05% TFA (aq)/ACN with the solvent ratio stabilized at 80/20 in the first 2 min, followed by a period of 8 min during which the solvent ratio is slowly changed until the final ratio of 0.05% TFA (aq)/ACN of 10/90 is reached, followed by a final period of 2 min during which the solvent ratio is stabilized at 10/90 of 0.05% TFA (aq)/ACN, a flow rate of 1.0ml/min, an ELSD evaporator temperature of 80 ℃; an ELSD atomizer temperature of 80 ℃; a column temperature of 22 ℃. LCMS process B: Waters Atlantis column with Krudcat T3 (C18 reversed phase silica gel; 4.6X 100mm, 3 μm; 0.05% aq/25.25 min.: 100/20 min → 3 min (ELSD) → 3 min; an ELSD evaporator temperature of 0.78 min → 3680 ℃; an ELSD evaporator temperature of 3 min → 364.80 ℃; and a flow rate of 0.80 ℃ Temperature of the mist sprayer: 80 ℃; the column temperature was 22 ℃. LCMS method C: waters Symmetry C18(C18 reverse phase silica; 4.6X 250mm, 5 μm); 0.05% TFA (aq)/ACN: 100/0(5.0 minutes) → (1.0 minute) → 10/90(2.0 minutes); flow rate: 1.0 ml/min; column temperature: 30 ℃; ELSD: 80 ℃, gain: LCMS method D: waters Xbridge BEH C18 XP column (C18 reverse phase column with Bridged Ethylene Hybrid (BEH) technique; 2.1X 50mm, 2.5 μm); mobile phase A: ammonium acetate (10mM), water/methanol/acetonitrile (900:60: 40); mobile phase B: ammonium acetate (10 mM); water/methanol/acetonitrile (100:540: 360); mobile phase a/mobile phase B: 80/20(0.0 min) → (1.5 min) → 0/100(2.5 min); flow rate: 0.6 ml/min; ELSD evaporator temperature: 40 ℃; ELSD nebulizer temperature: 40 ℃; the column temperature was 35 ℃.

Materials all chemicals and reagents were purchased from commercial suppliers (ACROS, Sigma Aldrich, Combi Blocks, Chem impax, LeapChem, shaw sun chemical ltd) and used without further purification. Palladium catalyst [ (copper novanite) PdOAc]2OTf2Prepared according to literature procedures (n.r.conley et al., Organometallics 26(23),5447(2007) and g. -j.ten Brink, adv.synth.catal.345,1341 (2003)). Thin Layer Chromatography (TLC) was performed on Merck silica gel 60, 0.25mm plates by UV, ninhydrin spray (0.2% ninhydrin in ethanol), phosphomolybdic acid (PMA) dip (23.4g phosphomolybdic acid in 300mL ethanol), and/orPotassium permanganate staining (KMnO)4(3g)、K2CO3(10g) And water (300mL) were observed. Column chromatography was performed using silica gel (60-120 mesh). Argon, nitrogen and hydrogen were purchased from Air Products and used as described below.

2. Cloning experiments

The microbial strains are as follows: escherichia coli ATCC 25922.

Media and equipment. LB broth (Lennox, cat # L3022, Sigma); agar-containing LB broth (Lennox, cat # L2897, Sigma); kanamycin A (cat # A1493, AppliChem); CloneJET PCR cloning kit (cat # K1231, Thermo Scientific); pBluescript II KS (+) (cat. 212207, Agilent); pET-9b (+) (cat # 69432-3, Merck); pSET152 (cat # PVT3395, Life Science Market); FastDiget SacI (cat No. FD1133, Thermo Scientific); FastDiget SalI (cat No. FD0644, Thermo Scientific); t4 DNA ligase (cat. No. EL0014, Thermo Scientific); agarose MP (cat # 11388991001, Roche); Tris-borate-EDTA buffer (cat. No. 106177, Merck); illustrla GFX PCR DNA and gel strip purification kit (cat. 28903471, GE Healthcare); GenElute TMHP plasmid miniprep kit (cat No. NA0160, Sigma); a heating block (Eppendorf Thermomixer C); an incubator (Sartorius Certomat BS-1); ultraviolet/visible light meter (Eppendorf BioPhotometer Plus); centrifuge (Beckman Coulter Avanti J-E).

Cloning of drug-resistant enzymes. The E.coli codon-optimized encoding AAC (3) -III gene (GenBank accession X55652.1; SEQ ID NO:1), including promoter and terminator sequences, was synthesized by Integrated DNA Technologies, Belgium. The synthesized gene and pBluescript II KS (+) plasmid were digested with restriction enzymes SacI and SalI and purified from 1% TBE-agarose gel using Illustra gel band purification kit. The purified linear plasmid and T4 DNA ligase were used to ligate aac (3) -III under the control of the constitutive lipoprotein lpp promoter and the ribosomal RNA rrnC terminator to produce pBlue _ lpp _ aac _3_ rrnC.

A competent cell. Coli ATCC from a storage of 20% glycerol (40% v/v glycerol in water diluted with MH 2) at-80 deg.C25922 until the optical density at 600nm in LB broth (measured by UV/Vis spectroscopy) at 37 ℃ and 200rpm reached 0.6, heat shock competent E.coli cells were prepared. Bacterial cells were centrifuged at 5000g for 5 min and resuspended in 1/10 original volume of ice-cold TSS (10% PEG 8,000, 5% DMSO, 50mM MgCl) 2In LB). 100 μ L aliquots were snap frozen in liquid nitrogen and stored at-80 ℃.

And (4) plasmid transformation. About 500ng of pET-9b (+) (Cat 69432-3, Merck), pAT21-1 (provided by Dr. Patricity Corvalin, university of Pasteur Bas, France; prepared according to Trieu-Curt et. al, mol. Gen. Genet.198(2),348(1985), pBluescript:: AAC (6 ') -Ie/APH (2 ") -Ia (provided by Dr. Sergei Vakulenko, Md. J. biol. chem.287(52),43262(2012), pBlue _ lpp _ aacC3_ nC and pST 33152 (Cat PVT3395, Life Science Market; prepared according to Bie Market, 43, 1992) SEQ ID 3, APla: (3-13 '), (AAC) (SEQ ID 3) No. (AAC) (2) (AAC) (2H 3, III) (NO 3, III) (1985), pBlue-II) (AAC 3, III) (AAC) (SEQ ID 3-11: (3, III) (AAC) (SEQ ID 3, III) (NO: 3, III) (NO 2 '), (III) (AAC) (SEQ ID 3, III) and SEQ ID 3) (NO: 1, III) of the plasmid II) of Pastek, III) of the aforementioned, The plasmids of AAC (3) -III (SEQ ID NO:1), and AAC (3) -IV (SEQ ID NO:5) were each incubated in a flask of competent cells on ice for 15 minutes. The cells were then heated to 42 ℃ for 90 seconds and cooled on ice for 1 minute. After addition of 900. mu.L LB, the cells were incubated at 37 ℃ for 1 hour at 200 rpm. Cells were plated on LB agar supplemented with 50. mu.g/mL KanaA and incubated at 37 ℃ for 18 hours. The next day, a single colony was inoculated in 5mL LB supplemented with 50. mu.g/mL and grown at 37 ℃ for 18 hours at 200 rpm. A sample of 200. mu.L of each culture was diluted with 200. mu.L of 40% v/v glycerol in water and stored at-80 ℃. Plasmids were purified from the remaining cultures using a plasmid miniprep kit. Plasmid DNA was studied by Eurofins Genomics, Germany to confirm the identity of aminoglycoside resistance genes.

3. The minimum inhibitory concentration test.

3.1 resistant strains of Enterobacter coli

Reference compound tested. Amikacin sulfate (cat # 01693, Chem-Impex), gentamicin sulfate (cat # G4918, Sigma Aldrich).

A microorganism strain. Escherichia coli ATCC 25922, Escherichia coli ATCC 25922/pET-9b (+), Escherichia coli ATCC 25922/pAT21-1, Escherichia coli ATCC 25922/pBluescript: AAC (6 ') -Ie/APH (2') -Ia, Escherichia coli ATCC 25922/pBlue _ lpp _ aacC3_ rrnC, Escherichia coli ATCC 25922/pSET 152.

Media and equipment. BBLTM Mueller Hinton broth 2 (lot number BCBW1143, cat number 70192, Sigma); kanamycin A (cat # A1493, AppliChem); 96-well deep-well plate, 1mL (cat # 0030506.200, Eppendorf); plate lids (cat # 0030131.525, Eppendorf); 96 well F-plates (cat # 781602); incubators (Infors Multitron Pro); a console (Heraeus KS 12); ultraviolet/visible light meter (Eppendorf BioPhotometer Plus); disk reader (Tecan Sunrise).

A compound preparation. Compounds were prepared at a concentration of 5mM in MH2 medium and stored at 4 ℃. For testing, 128 μ M MH2 solution was prepared. 400 μ L of these working solutions were transferred to wells of the first column of a 96-well deep-well plate. Each compound was transferred in triplicate. In all wells except the first row of wells of the plate, 200 μ L of MH2 medium was pre-filled. After addition of compound and antibiotic, the compound was serially diluted two-fold by transferring 200 μ Ι _ from the first column to the second column, then from the second column to the third column, and so on. Finally, 200 μ L was removed from the last column.

An inoculum preparation. The microorganisms used were recovered from a storage of 20% glycerol (40% v/v glycerol in water diluted with MH 2) at-80 ℃ by inoculating them in 5mL MH 2. Coli inocula containing plasmids were selected by adding 50. mu.g/mL KanaA. The inoculum was grown to an optical density (measured by uv/vis spectroscopy) of 0.6-0.8 at 600 nm. From these suspensions, the actual inocula were prepared by diluting them to an optical density of 0.1 in MH2 medium. From these inocula, 200. mu.L were transferred to each well to give a final compound concentration range of 64. mu.M-31.3 nM. All plates were incubated at 37 ℃ for 18 hours at 900rpm, 70% rH.

And (4) determining MIC. After 18 hours of growth, 200 μ Ι _ were transferred to 96 well F-bottom plates. The optical density was measured by a plate reader at 600nm, shaking for 10s before reading. The three measurements were averaged and the growth curve was plotted in Microsoft Excel. The first column where growth reached half the maximum growth value was determined as the MIC50 value for that particular compound.

3.2 selected bacteria against the ESKAPE group and enterococcus faecalis (DSM Strain)

Reference compound tested. Amikacin sulfate (cat # 01693, Chem-Impex), gentamicin sulfate (cat # G4918, Sigma Aldrich).

A microorganism strain. Enterococcus faecalis DSM 2570, Klebsiella pneumoniae DSM 26371, Acinetobacter baumannii DSM 105126, Enterobacter cloacae 3MRGN (clinical bronchial aspirate isolate).

Media and equipment. BBLTMMueller Hinton broth 2 (lot No. BCBW1143, cat No. 70192, Sigma); trypcase Soy Blood agar plate (cat # 43001, biomerieux); refrigerants (15% glycerol; cat # 822070ZA, VWR International); GasPakTMEZ Standard culture vessel (cat # 260671, Becton Dickinson); GasPakTMEZ campapy container system pouch (cat # 260680, Becton Dickinson); 96-well deep-well plate, 1mL (cat # 0030506.200, Eppendorf); plate lids (cat # 0030131.525, Eppendorf); 96 well F bottom plate (cat No. 260860, Nunc); an incubator (Sartorius Certomat BS-1); a console (Heraeus HS 12); uv/visible light meter (Perkin Elmer Lambda Bio +); a disk reader (Tecan Infinite M200 Pro).

A compound preparation. Compounds were prepared at a concentration of 5mM in MH2 medium and stored at 4 ℃. For testing, 128 μ M solution was prepared in MH2 and 512 μ M solution was prepared for the anti-enterococcus faecalis test. 400 μ L of these working solutions were transferred to wells of the first column of a 96-well deep-well plate. Each compound was transferred in triplicate. In all wells except the first row of wells of the plate, 200 μ L of MH2 medium was pre-filled. After addition of compound and antibiotic, the compound was serially diluted two-fold by transferring 200 μ Ι _ from the first column to the second column, then from the second column to the third column, and so on. Finally, 200 μ L was removed from the last column.

An inoculum preparation. The micro-powder is usedThe organisms were revived from a 15% glycerol stock in croinstan tubes at-28 ℃ by plating onto blood agar plates. Acinetobacter baumannii and Enterobacter cloacae grow aerobically at 37 ℃, enterococcus faecalis grows microaerobically at 37 ℃ and Klebsiella pneumoniae grows aerobically at 37 ℃. The plates were stored at 4 ℃ for up to two weeks, after which new plates were prepared from-28 ℃. A single colony was inoculated in MH2 medium and grown at 37 ℃ at 200rpm to an optical density (measured by UV/Vis spectroscopy) of 0.6-0.8 at 600 nm. In these suspensions, the actual inocula were prepared by diluting them to an optical density of 0.1 in MH2 medium. From these inocula, 200. mu.L were transferred to each well, resulting in a final concentration of compound in the range of 64. mu.M-31.3 nM for Klebsiella pneumoniae, Acinetobacter baumannii and Enterobacter cloacae and 256. mu.M-125 nM for enterococcus faecalis. Place the plates inoculated with enterococcus faecalis in the presence of GasPakTMGasPak for pouch of Campy container systemTMWithin the container system. All plates were incubated for 18 hours at 200rpm under the microbial culture conditions described above.

And (4) determining MIC. After 18 hours of growth, 200. mu.L was transferred to a 96-well F-plate. The optical density was measured by a plate reader at 600nm, shaking for 10s before reading. Triplicate measurements were averaged and growth curves were plotted in Excel. The first column where growth reached half the maximum growth value was determined as the MIC50 value for that particular compound.

3.3 MIC assay protocol (ATCC strain)

Reference compound tested. Amikacin sulfate (cat No. 01693, Chem-Impex), gentamicin sulfate (cat No. G4918, Sigma Aldrich), azithromycin (cat No. 1046056, USP), ceftazidime pentahydrate (cat No. 1098130, USP), ciprofloxacin (cat No. 1134313, USP), meropenem (cat No. 1392454, USP), rifampin (cat No. R0079, TCI), tobramycin (cat No. 1667508, USP), vancomycin (cat No. 1709007, USP).

A microorganism strain. Staphylococcus aureus ATCC 29213, Staphylococcus aureus ATCC BAA-1717, Escherichia coli ATCC BAA-1025, Klebsiella pneumoniae ATCC BAA-1705, Klebsiella pneumoniae ATCC BAA-2524, Pseudomonas aeruginosa ATCC 27853, Pseudomonas aeruginosa ATCC BAA-2108, Acinetobacter baumannii ATCC BAA-1800.

Media and equipment. BBLTMMueller Hinton broth (product No. 275730, batch No. 7009699, Becton Dickinson), Mueller Hinton agar 2 (product No. 97580-500G-F, batch No. BCBV4646, Sigma), Dulbecco's Modified Eagle Medium (DMEM) (product No. 41966-029, Gibco), Minimum Essential Medium (MEM) (product No. 42360-024, Gibco), Fetal Bovine Serum (FBS) (product No. F7524, Sigma), nonessential amino acids (NEAA) (100X, product No. 11140-035, Gibco), sodium pyruvate (100mM, 100X, product No. 11360-039, Gibco), staurosporine (product No. 01BS 88, Biotrend), Cellstar 96 well U plate (product No. E14123, Greie-EB), Titer-Glo (product No. Proc 7573, Protcol 7596 well No. 6596), transparent well plate (product No. 6596-V-96 well No. 3698, product No. 35-6596-G-95, No. 3-6596-95-E, product No. 3-E # 3698, Gei-E # 3-E, Gei-E # 3-E, Gei, and E # 3-E, Sodium pyruvate (100mM, 100X, Gibco, cat # 11360-; DEN-1B (McFarland densitometer, Biosan); McFarland standard (polymer particle suspension, Biosan).

And (3) preparing a compound. Test compounds and reference antibiotics were prepared as 5mg/mL stock solutions in DMSO according to CLSI guidelines (CLSI; Methods for differential antibiotic negative feedback tests for bacterial that grow in bacteria, improved standard-antibiotic injection, M07-A9, Vol.32, No.2,2012). In these DMSO solutions, 43.5 μ L were transferred to 1656.5 μ L of MH medium in deep well plates. These working solutions were transferred 100. mu.L to wells in the third column of the 96-well plate. In all wells except the wells of the third column of the plate, 50 μ L of MH medium was pre-filled. After addition of compound and antibiotic, 50 μ L was transferred from the third column to the fourth column, then from the fourth column to the fifth column, and so on. In this manner, compounds and antibiotics were plated in 96-well plates in serial two-fold dilutions to give a final concentration range of 64-0.125. mu.g/mL.

And (4) preparing the microorganism. Name for clinical isolatesAre referred to as from the Fildelta strain collection library. All microorganisms were recovered from skim milk storage at-70 ℃ by plating on MH agar plates. The next day, a single colony of each microorganism was streaked again on fresh agar plates. The next day, a broth solution was prepared which reached a turbidity equal to the 0.5McFarland standard for each microorganism, using the direct colony suspension method. This results in a suspension containing 1-2X 10 8CFU/mL. In these suspensions, the actual inocula were prepared by diluting them 100-fold with MH medium, yielding 2-8X 105Final microbial count of CFU/mL. For each microbial strain, 20mL of these inoculum solutions were prepared. From the second column to the twelfth column of the 96-well plate, 50. mu.L of these solutions were transferred per well. To the first column 50. mu.L of pure growth medium per well was added. In this way, the first column served as a sterile control for the culture medium, the second column served as a control for the growth of the microorganisms, and the rest of the plate was used for the MIC determination. All plates were incubated at 37 ℃ for 16-24 hours.

And (4) determining MIC. MIC values were determined by visual inspection of bacterial growth in 96-well plates. No bacterial growth was visible in the first column, determined as the MIC90 value for the test compound or the reference antibiotic tested in that particular row. The ATCC strain was used as a reference strain, where a determination of the MIC value of the reference antibiotic was present. The assay is considered to be effective when the MIC value of the reference antibiotic is within the CLSI specification (CLSI; Performance Standards for antibiotic compatibility testing; Twenty-third information supplement, M100-S23, Vol.33, No.1,2013) of the ATCC strain tested.

3.4 MIC determination protocol (clinical isolate)

3.4.1 MIC determination at Uppsala university (Sweden)

Reference compound tested. Amikacin sulfate (cat # A1774, Sigma Aldrich), apramycin sulfate (cat # A2024, Sigma Aldrich), and gentamycin sulfate (cat # G1914, Sigma Aldrich).

Bacterial strains/isolates. All laboratory and clinical strains analyzed (Pseudomonas aeruginosa PA01, Acinetobacter baumannii ATCC 19606, Acinetobacter baumannii 195N (A), Acinetobacter baumannii 48F, Escherichia coli C1162, Escherichia coli C1181, Morganella morganii S49, and providencia stuartii B8-1) were from the IMI ND4BB ENABLE consortium strain collection. ATCC laboratory strains were obtained directly from the American type culture Collection. The clinical strains used have been previously disclosed (M Juhas, et al J. Antiicrob. Chemother.,74 (4)), 944-952 (2019); D.Hughes et al, Antiicrob. Agents Chemother.,47(10),3222-3232(2003)) and drug resistance genotyping was performed in ENABLE consortium.

Media and equipment. Water (Thermo Scientific, R05819); Mueller-Hinton II broth (BD product No. 212322); sterile round bottom 96-well plates (Thermo Scientific, 262162); sensisterie turbidimeter (ThermoFisher Scientific, V3011); 0.5McFarland standard (ThermoFisher Scientific, E1041); sterile polyester film (VWR, 391-1251).

A compound preparation. MIC determination Methods follow the CLSI M07 standard method (Clinical and Laboratory Standards Institute (CLSI); Methods for differential antimicrobial supplementary tests for bacterial that grow in the soil, advanced standard-evaluation, M07-A10, Vol.38, No.2,2018). The test compound was received as a solid and dissolved in water to a concentration of 10mg/mL of active compound. The concentrated compound solution was diluted to 128 μ g/mL by adding 12.8 μ L of the concentrated compound to 1mL of liquid Mueller-Hinton II broth freshly prepared according to the manufacturer's instructions. The solution was serially diluted 2-fold in the first 10 columns of sterile round bottom 96-well plates while the last two columns were filled with Mueller-Hinton II broth, with no compound added, and used as positive and negative growth controls.

An inoculum preparation. The inoculum was prepared by suspending several colonies of fresh streaked test strains in 5mL of sterile 0.9% NaCl. Cell density of the inoculum solution was evaluated and adjusted using a Sensittre turbidimeter and 0.5McFarland standards. The inoculum was diluted 1:100 in Mueller-Hinton II broth and 50. mu.L of this inoculum solution was added to 50. mu.L of the test compound solution to give a concentration range of 64-0.125. mu.g/mL. The plates were sealed with a sterile polyester film and incubated at 37 ℃ for 18 hours on standing.

And (4) determining MIC. MIC values were determined by visual inspection of bacterial growth in 96-well plates. The lowest concentration at which growth was completely inhibited was reported as the MIC for each strain and compound combination.

3.4.2 MIC determination in Crohn's Fidelta

Reference compound tested. Amikacin sulfate (cat No. 01693, Chem-Impex), gentamicin sulfate (cat No. G4918, Sigma Aldrich), ceftazidime pentahydrate (cat No. 1098130, USP), meropenem (cat No. 1392454, USP), tobramycin (cat No. 1667508, USP).

A microorganism strain. Klebsiella pneumoniae KP-1919 (abdomen), Klebsiella pneumoniae KP-1935 (urine), Klebsiella pneumoniae KP-1936 (abscess), Klebsiella pneumoniae KP-1937 (blood), Klebsiella pneumoniae KP-1942 (urine), Klebsiella pneumoniae KP-1944 (blood), Klebsiella pneumoniae KP-2027 (trachea aspirate), Klebsiella pneumoniae KP-2029 (urine), Klebsiella pneumoniae KP-2030 (trachea), Klebsiella pneumoniae KP-2031 (skin), Klebsiella pneumoniae KP-2032 (urine), Klebsiella pneumoniae KP-2033 (urine), Klebsiella pneumoniae KP-2034 (urine), Klebsiella pneumoniae KP-2035 (urine), Klebsiella pneumoniae KP-2036 (urine), urine (abscess, urine (urine), and urine (urine) in the presence of Klebsiella pneumoniae KP-2036 (urine), Klebsiella pneumoniae KP-2037 (urine), Klebsiella pneumoniae ATCC 43816, Pseudomonas aeruginosa PA-1948 (wound), Pseudomonas aeruginosa PA-1949 (urine), Pseudomonas aeruginosa PA-1950 (sputum), Pseudomonas aeruginosa PA-1952 (wound), Pseudomonas aeruginosa PA-1953 (urine), Pseudomonas aeruginosa PA-1954 (sputum), Pseudomonas aeruginosa PA-1967 (blood), Pseudomonas aeruginosa ATCC27853, Acinetobacter baumannii AB-1931 (urine), Acinetobacter baumannii AB-1932 (urine), Acinetobacter baumannii AB-2017 (bronchial aspirate), Acinetobacter baumannii AB-2018 (skin), Acinetobacter baumannii AB-2019 (bronchial aspirate), Acinetobacter baumannii AB-1964 (blood), Acinetobacter baumannii AB-2025 (skin), Acinetobacter baumannii AB-2026 (skin), acinetobacter baumannii ATCC17978, methicillin-resistant Staphylococcus aureus MRSA-1995 (body fluid), methicillin-resistant Staphylococcus aureus MRSA-1998 (wound), methicillin-resistant Staphylococcus aureus MRSA-1999 (skin), methicillin-resistant Staphylococcus aureus MRSA-2003 (wound), vancomycin-resistant enterococcus faecium VRE-2005 (urine), vancomycin-resistant enterococcus faecium VRE-2006 (urine), vancomycin-resistant enterococcus faecium VRE-2007 (urine), vancomycin-resistant enterococcus faecium VRE-2008 (urine), vancomycin-resistant enterococcus faecium VRE-2009 (urine), vancomycin-resistant enterococcus faecium VRE-2010 (urine), vancomycin-resistant enterococcus faecium VRE-2011 (rectum), vancomycin-resistant enterococcus faecium VRE-2012 (rectum), Vancomycin-resistant enterococcus faecium VRE-2013 (rectum).

Media and equipment. BBLTMMueller Hinton broth (product No. 275730, lot No. 7009699, Becton Dickinson); mueller Hinton agar 2 (product number 97580-500G-F, lot number BCBV4646, Sigma); cellstar 96 well U-plate (accession number 650180, batch number E14123EB, Greiner Bio-one); 96-well deep-well plate, 1mL (cat # 278606, Nunc); an incubator (Binder); a console (Thermo Scientific); a spectrophotometer (Eppendorf); DEN-1B (McFarland densitometer, Biosan); McFarland standard (polymer particle suspension, Biosan).

A compound preparation. For testing, 5mg/mL of DMSO solutions of test compound and reference antibiotic were used. In these DMSO solutions, 43.5 μ L were transferred to 1656.5 μ L of MH medium in deep well plates. These working solutions were transferred 100. mu.L to wells in the third column of the 96-well plate. In all wells except the wells of the third column of the plate, 50 μ L of MH medium was pre-filled. After addition of compound and antibiotic, 50 μ L was transferred from the third column to the fourth column, then from the fourth column to the fifth column, and so on. In this manner, compounds and antibiotics were plated in 96-well plates in serial two-fold dilutions to give a final concentration range of 64-0.125. mu.g/mL.

An inoculum preparation. The names used for clinical isolates are from the Fildelta strain depository. All microorganisms were recovered from skim milk storage at-70 ℃ by plating on MH agar plates. The next day, a single colony of each microorganism was streaked again on fresh agar plates. The next day, a broth solution was prepared which reached a turbidity equal to the 0.5McFarland standard for each microorganism, using the direct colony suspension method. This results in a suspension containing 1-2X 108CFU/mL. In these suspensions, the actual inocula were prepared by diluting them 100-fold with MH medium, yielding 2-8X 105Final microbial count of CFU/mL. For each microbial strain, 20mL of these inoculum solutions were prepared. From the second column to the twelfth column of the 96-well plate, 50. mu.L of these solutions were transferred per well. To the first column 50. mu.L of pure growth medium per well was added. In this way, the first column serves as a sterile control for the medium used, the second column serves as a control for the growth of the microorganisms, and the remainder of the plate is used for the MIC determination. All plates were incubated at 37 ℃ for 16-24 hours.

And (4) determining MIC. MIC values were determined by visual inspection of bacterial growth in 96-well plates. No bacterial growth was visible in the first column, determined as the MIC90 value for the compounds tested in that particular row.

3.4.3 MIC determination at the university of Geroning medical center (Netherlands)

Reference compound tested. Amikacin sulfate (cat # 01693, Chem-Impex), gentamicin sulfate (cat # G4918, Sigma Aldrich).

Bacterial strains/isolates. Pseudomonas aeruginosa ATCC27853, escherichia coli ATCC25922, klebsiella pneumoniae ATCC43816, acinetobacter baumannii ATCC17978, 9 clinical isolates of klebsiella pneumoniae (source: 4 unknown, 3 urine, 1 sputum, 1 catheter tip), 7 clinical isolates of pseudomonas aeruginosa (source: 3 unknown, 1 wound, 1 catheter, 1 stool, bronchoalveolar fluid ═ BAL), 8 clinical isolates of escherichia coli (source: 2 bile juice, 2 rectum, 1 throat, 2 blood cultures, 1 sputum), 4 clinical isolates of acinetobacter baumannii (source: 2 blood cultures, 1 sputum, 1 bone).

Media and equipment. Such as The European Commission on Antimicrobial scientific Testing, EUCAST, guidelines for Media preparation for EUCAST disk differentiation and for determination of MIC values by The broth microdialysation method; Version 5.0,2017), Mueller Hinton broth, CM0405, (Oxoid) for broth microdilution; and Mueller Hinton agar, CM0337, (Oxoid) For plating on agar. Assay assays were performed in 96-well U-plates (Greiner Bio-One); all bacterial strains/isolates used were treated in a clean air station (Baker). CO sold using Sanyo2An incubator. Finally, spectrophotometric measurements were performed using a Synergy 2 multi-assay microplate reader (BioTek).

A compound preparation. Compounds were weighed and dissolved in MilliQ water. Aliquots of these concentrated stock solutions were then further diluted to achieve the desired test concentrations. Serial dilutions were made to reach ten desired final concentrations of test compound ranging between 64-0.125 μ g/mL. The concentration was calculated relative to a final volume of 150 μ Ι _ for each of the 96 wells. To these dilutions, 100 μ L was added to each test well of a sterile 96-well plate. All compound dilutions were plated in triplicate and in duplicate for the two reference antibiotics.

An inoculum preparation. Clinical isolates were recovered from Luria Bertani medium stored at-80 ℃ by plating on MH agar plates overnight. The next day, several colonies were picked and inoculated into a small glass bottle containing 10mL MH broth. Growth was then allowed for about 3 hours, shaking at 250rpm at 35 ℃ until the isolate reached late logarithmic growth phase. At this point, OD was prepared 600(optical density measured at 600 nm; measured using a Synergy 2 multi-detection microplate reader from BioTek) 0.05. This results in a starting bacterial suspension containing 2-8X 105CFU/mL. 50 μ L of the culture suspension was added to each well of a 96-well plate previously loaded with the desired antibiotic/culture medium solution to a final total volume of 150 μ L per well. As a control, a blank of MH medium and culture was added to a 96-well plate. The plates were incubated in a plate incubator at 35 ℃ for 18 hours under quiescent conditions.

And (4) determining MIC. After the incubation time, MIC values were determined by visual inspection of bacterial growth in 96-well plates. No bacterial growth was visible in the first column, determined as the MIC90 value for the compounds tested in that particular row. In addition, a spectrophotometer reading at 600nm was recorded for each well.

3.5 MIC value reporting

MIC 50. The MIC50 for the indicated compound was half the maximum growth value reached for bacterial growth at this time.

MIC 90. No visible bacterial growth was observed and the MIC90 value for the compound was determined.

MIC values are given in concentration ranges, e.g., 1-2, 2-4, 4-8, etc., which result from two serial dilution steps, meaning that the actual MIC is above the lower value of the concentration range, but below the upper value of the range, i.e., reported as 1-2 for the MIC range, which is understood to mean "a value between 1 and 2," and thus does not include the explicitly recited endpoints of 1 and 2.

4. In vivo efficacy

4.1 mouse thigh infection model with neutropenia (Klebsiella pneumoniae ATCC 43816)

All animal-related studies were in accordance with 2010/63/EU, and national regulations governing the use of laboratory animals for other purposes in scientific research (official gazette 55/13). The animal research institute Committee (CARE-Zg) supervised animal-related procedures without compromising the health of the animals.

In vivo studies and CFU assays were performed by fiddelta (saxabrib, crohnia), and all mice were obtained from Charles River Laboratories (italy).

In two independent studies (ABX 5006 and ABX5026 in table 12, ABX5026 and ABX5039 in table 13), four compounds, ABX5006, ABX5020, ABX5026 and ABX5039, were tested using a klebsiella pneumoniae ATCC 43816 infected mouse thigh model. A comparative antibiotic (meropenem; cat # 1392454, USP) was used in both studies.

Thigh model for neutropenia: pathogen-free male CD-1 mice weighing 36. + -.4 g (6 mice per administration group) were used. Neutropenia in mice was induced by intraperitoneal administration of two doses of cyclophosphamide four days (150mg/kg body weight) and one day before infection (100mg/kg body weight) prior to infection.

The inoculum size and dose of meropenem (broad spectrum antibiotic) pharmacological standards were selected based on previous titration and validation studies to determine the maximum number of CFUs that can be injected during the course of the experiment without subsequent death. For subcutaneous treatment, pre-weighed test article powder was dissolved in PBS and the pH was adjusted (to achieve a near neutral pH) to achieve the desired final concentration.

In the first study, ABX5006 and ABX5020 were tested at doses of 64mg/kg (ABX5006 is group 5, ABX5020 is group 8), 16mg/kg (ABX5006 is group 4, ABX5020 is group 7) and 4mg/kg (ABX5006 is group 3, ABX5020 is group 6).

In the second study, ABX5026 and ABX5039 were tested at doses of 4mg/kg (ABX5026 for group 5, ABX5039 for group 8), 1mg/kg (ABX5026 for group 4, ABX5039 for group 7) and 0.25mg/kg (ABX5026 for group 3, ABX5039 for group 6).

Under mild ketamine/xylazine anesthesia, 0.10mL of a suspension containing bacteria was administered intramuscularly to both thighs using klebsiella pneumoniae ATCC 43816(6.0 x 10)3Thigh) infected each mouse. Animals were dosed subcutaneously (10mL/kg) twice at 8-hour intervals. Treatment was started 1 hour after infection with vehicle-PBS (group 1) or meropenem 100mg/kg (group 2) or test compound as described above (groups 3-8). All mice were overdosed with ketamine/xylazine 24 hours post-infection. The thighs were removed aseptically and placed individually in sterile precell tubes containing 2mL sterile PBS. The precell tubes were weighed and the weight was recorded before and after sampling. Each thigh was homogenized in sterile PBS using an Ultraturax of IKA. After homogenization, serial dilutions of the homogenate were used for CFU determination. CFU counts per thigh as a single result. For statistical analysis, CFU counts below the limit of detection were replaced by a value of 0 (LOD ═ 1.0 × 10) 2CFU/mL)。

Supplies/chemicals. BBLTMMueller Hinton broth (product No. 275730, Becton Dickinson); BBLTMMueller Hinton agar 2 (product No. 211438, Becton Dickinson); PBS (cat P4417, Sigma); saline (cat # 1422115, HZTM); 2% phenazine (Alfasan, International b.v.); narkamon (100mg/mL ketamine chloride, Bioveta); cyclophosphamide monohydrate (cat # C2236, TCI); sterile, single-use needles (BD MICROLANCE Kanuele, 27G,product number 300220, Becton Dickinson); 5mL syringes (BD Plastipak, product No. 300013, Becton Dickinson).

And (6) anaesthetizing. 1mL Narkamon, 0.2mL Xylazine, and 9mL saline; dose volume: 10mL/kg body weight.

An inoculum formulation for intramuscular infection. The strains were streaked onto MH agar. The next day, Mueller-Hinton broth was inoculated with several colonies of Klebsiella pneumoniae and incubated in an orbital shaker at 37 ℃ until the logarithmic growth phase (OD) was reached6000.6). Log bacteria were centrifuged and suspended in an equal volume of sterile PBS, followed by dilution to obtain a bacterial suspension of sufficient inoculum size.

And (4) CFU determination. Thigh samples were homogenized using a precells Evolution homogenizer (Bertin Instruments) according to the hard tissue program (test tube: 7 mL; speed: 6800 RPM; cycle: 4X 15 s; pause: 5 s). After homogenization, 100 μ Ι _ of the homogenate was transferred to a 96-well U-plate, where serial dilutions in PBS were prepared by transferring 10 μ Ι _ of the homogenate to 90 μ Ι _ of PBS. In this way, 9 serial dilutions were prepared, of which 10 μ L was used to inoculate agar plates, repeated 5 times. The agar plates were then incubated overnight at 37 ℃ and the CFU counts were counted.

4.2 mouse thigh infection model with neutropenia (Klebsiella pneumoniae ATCC BAA-1705)

All animal-related studies were in accordance with 2010/63/EU, and national regulations governing the use of laboratory animals for other purposes in scientific research (official gazette 55/13). The animal research institute Committee (CARE-Zg) supervised animal-related procedures without compromising the health of the animals.

In vivo studies and CFU assays were performed by fiddelta (saxabrib, crohnia), and all mice were obtained from Charles River Laboratories (italy).

Two compounds, ABX5006 and ABX5020, and a comparative antibiotic (amikacin sulfate; cat # 01693, Chem-Impex) were tested in a murine model of thigh infection with Klebsiella pneumoniae ATCC BAA-1705.

Thigh model for neutropenia: pathogen-free male CD-1 mice weighing 36. + -.4 g (6 mice per administration group) were used. Neutropenia in mice was induced by intraperitoneal administration of two doses of cyclophosphamide four days (150mg/kg body weight) and one day before infection (100mg/kg body weight) prior to infection.

Inoculum size and antibiotic dose were selected based on previous titration and validation studies, determining the maximum number of CFUs that can be injected during the course of the experiment without subsequent death. For subcutaneous treatment, pre-weighed test article powder (ABX5006) was dissolved in PBS and the pH was adjusted (to a near neutral pH) to a final concentration of 6.4mg/mL for a 64mg/kg dose (group 3) or 1.6mg/mL for a 16mg/kg dose (group 2). For subcutaneous treatment, pre-weighed test article powder (ABX5020) was dissolved in PBS and the pH was adjusted (to a near neutral pH) to a final concentration of 6.4mg/mL for a 64mg/kg dose (group 7) or 1.6mg/mL for a 16mg/kg dose (group 6). For subcutaneous treatment, a pre-weighed reference compound (amikacin sulfate) was dissolved in PBS and the pH was adjusted (to a near neutral pH) to a final concentration of 6.4mg/mL for a 64mg/kg dose (group 5) or 1.6mg/mL for a 16mg/kg dose (group 4). Under mild ketamine/xylazine anesthesia, 0.10mL of a suspension containing bacteria was administered intramuscularly to both thighs using Klebsiella pneumoniae ATCC BAA-1705 (1.18X 10) 6Thigh) infected each mouse. Animals were dosed subcutaneously twice at 8 hour intervals. Treatment was started 1 hour after infection with vehicle-PBS (group 1) or amikacin sulfate (groups 4-5) or test compounds as described above (groups 2-3 and 6-7). All mice were overdosed with ketamine/xylazine 24 hours post-infection. The thighs were removed aseptically and placed individually in sterile precell tubes containing 2mL sterile PBS. The precell tubes were weighed and the weight was recorded before and after sampling. Each thigh was homogenized in sterile PBS using an Ultraturax of IKA. After homogenization, serial dilutions of the homogenate were used for CFU determination. CFU counts per thigh as a single result.

Supplies/chemicals. BBLTMMueller Hinton broth (product No. 275730, Becton Dickinson); BBLTMMueller Hinton agar 2 (product No. 211438, Becton Dickinson); PBS (cat P4417, Sigma); saline (cat # 1422115, HZTM); 2% phenazine (Alfasan, International b.v.); narkamon (100mg/mL ketamine chloride, Bioveta); cyclophosphamide monohydrate (cat # C2236, TCI); a sterile, single-use needle (BD MICROLANCE Kanuele, 27G, product No. 300220, Becton Dickinson); 5mL syringes (BD Plastipak, product No. 300013, Becton Dickinson).

And (6) anaesthetizing. 1mL Narkamon, 0.2mL Xylazine, and 9mL saline; dose volume: 10mL/kg body weight.

An inoculum formulation for intramuscular infection. The strains were streaked onto MH agar. The next day, Mueller-Hinton broth was inoculated with several colonies of Klebsiella pneumoniae and incubated in an orbital shaker at 37 ℃ until the logarithmic growth phase (OD) was reached6000.6). Log bacteria were centrifuged and suspended in an equal volume of sterile PBS, followed by dilution to obtain a bacterial suspension of sufficient inoculum size.

And (4) CFU determination. Thigh samples were homogenized using a precells Evolution homogenizer (Bertin Instruments) according to the hard tissue program (test tube: 7 mL; speed: 6800 RPM; cycle: 4X 15 s; pause: 5 s). After homogenization, 100 μ Ι _ of the homogenate was transferred to a 96-well U-plate, where serial dilutions in PBS were prepared by transferring 10 μ Ι _ of the homogenate to 90 μ Ι _ of PBS. In this way, 9 serial dilutions were prepared, of which 10 μ L was used to inoculate agar plates, repeated 5 times. The agar plates were then incubated overnight at 37 ℃ and the CFU counts were counted.

4.3 urinary tract infection model (Escherichia coli ATCC 700336)

All animal-related studies were in accordance with 2010/63/EU, and national regulations governing the use of laboratory animals for other purposes in scientific research (official gazette 55/13). The animal research institute Committee (CARE-Zg) supervised animal-related procedures without compromising the health of the animals.

In vivo studies and CFU assays were performed by fiddelta (sagebrush, crohn), and all mice were obtained from Janvier Labs (france).

Two compounds, ABX5006 and ABX5026, and a comparative antibiotic (gentamicin sulfate; cat. No. G4918, Sigma Aldrich) were tested in the E.coli ATCC 700336 mouse model for urinary tract infection.

Urinary tract infection model: pathogen-free female C3H/HeNRj mice weighing 24. + -.4 g (8 mice per administration group) were used.

Inoculum size and antibiotic dose were selected based on previous titration and validation studies, determining the maximum number of CFUs that can be injected during the course of the experiment without subsequent death.

For subcutaneous treatment, pre-weighed test article powder (ABX5006) was dissolved in PBS and the pH was adjusted (to a near neutral pH) to a final concentration of 1.5mg/mL for a 15mg/kg dose (group 5) or 3.0mg/mL for a 30mg/kg dose (group 4). For subcutaneous treatment, pre-weighed test article powder (ABX5026) was dissolved in PBS and the pH was adjusted (to a near neutral pH) to a final concentration of 1.5mg/mL for a 15mg/kg dose (group 7) or 3.0mg/mL for a 30mg/kg dose (group 6). For subcutaneous treatment, a pre-weighed reference compound (gentamicin sulfate) was dissolved in 0.9% sodium chloride solution to a final concentration of 1.5mg/mL for a 15mg/kg dose (group 3), or 3.0mg/mL for a 30mg/kg dose (group 2). Under mild ketamine/xylazine anesthesia, E.coli ATCC 700336 (5.0X 10) 9CFU/100 μ L/mouse) per mouse was infected via the urethra. Mice were starved of water 1.5 hours before infection and 1 hour after infection.

Animals were administered once subcutaneously gentamicin (groups 2-3), ABX5006 (groups 4-5), ABX5026 (groups 6-7) or vehicle-PBS (group 1) 24 hours post infection.

All mice were overdosed with ketamine/xylazine 24 hours post-infection. Both kidneys were aseptically removed and placed in pre-weighed sterile precell tubes containing 2mL sterile PBS. After sampling, the precell tubes were weighed again. Kidneys were homogenized in sterile PBS using a Precellys Evolution homogenizer (BERTIN Technologies) and serially diluted for CFU assay. The kidney pair of one animal will represent one sample.

Supplies/chemicals. Tryptic soy agar (BD Cat 236950, Becton Dickinson), tryptic soy broth (BD Cat 211768, Becton Dickinson), 2mL syringe (BD Plasmopak, product No. 300185, Becton Dickinson), 1mL syringe (BD Plasmopak, product No. 300013, Becton Dickinson), sterile disposable needle (BD MICROLANCE, 27G3/4, Nr.20, product No. 300220, Becton Dickinson), sterile disposable needle (BD MICROLANCE, 25G 1, No.18, No. 300400, Becton Dickinson), Narkamon (100mg/mL chloramine chloride) (Czech republic, Bioveta, a.s KT), Xylazine 2% (Welchon Wu-Dan Alsafan. V), Germany saline (ZTM, series No. PBS, Sigma 1422115, product No. 44961, French-037, product No. Becton-03961)

And (6) anaesthetizing. 1mL Narkamon, 0.2mL Xylazine, and 9mL saline; dose volume: 10mL/kg body weight.

An inoculum formulation for intramuscular infection. Coli ATCC 700336 was grown on tryptic soy agar plates and incubated at 37 ℃ for 18-20 hours. Bacterial agar growth suspension 1.0X 1010Colony Forming Units (CFU)/mL in Tryptic Soy Broth (TSB) in sufficient volume and the inoculum was kept at 4 ℃ until used for infection.

And (4) CFU determination. The kidney pairs were homogenized using a Precellys Evolution homogenizer (Bertin Instruments). After homogenization, 100 μ Ι _ of the homogenate was transferred to a 96-well U-plate, where serial dilutions in PBS were prepared by transferring 10 μ Ι _ of the homogenate to 90 μ Ι _ of PBS. In this way, 9 serial dilutions were prepared, of which 10 μ L was used to inoculate agar plates, repeated 5 times. The agar plates were then incubated overnight at 37 ℃ and the CFU counts were counted.

4.4 Maximum Tolerated Dose (MTD) study in male CD (Sprague Dawley) rats.

The study was performed in an AAALAC I approved apparatus. The standard research program relevant to this study was reviewed by the ethical committee (CARE-sagelub) and animal welfare officials, in accordance with international laws/regulations and requirements of the krydian animal welfare act (animal protection act, official gazette, NN 37/13).

In vivo studies were performed by fiddelta (saxabrib, crohn) and all rats (Sprague Dawley) were obtained from Charles River Laboratories (italy).

Three compounds, ABX5006, ABX5020 and ABX5039, were tested to determine the MTD of the test article in male cd (sprague dawley) rats after one dose (IV).

MTD study: pathogen-free male CD (Sprague Dawley) rats weighing 225-260 g (3 rats per administration group) were used. Five different dosing groups (50mg/kg, 75mg/kg, 100mg/kg, 150mg/kg and 200mg/kg) were used and the test article was given intravenously over 15 minutes (slow injection, 5 mL/kg).

For intravenous administration, pre-weighed test article powder was dissolved in PBS and the pH adjusted (to a near neutral pH) to achieve a final concentration of 10.0mg/mL for a 50mg/kg dose (group 2), 15.0mg/mL for a 75mg/kg dose (group 3), 20.0mg/mL for a 100mg/kg dose (group 4), 30.0mg/mL for a 150mg/kg dose (group 3) or 40.0mg/mL for a 200mg/kg dose (group 6). Observations were made after 30 minutes post-dose, and all rats were again observed after 1 hour, 4 hours and 8 hours, and twice daily for the remaining duration of the study (4 days). Clinical signs, body weight, food consumption and mortality were recorded during the life of the study. All animals surviving to the end of the 4 day monitoring period were euthanized on day 4. The final study included gross necropsy and organ weight recordings of all animals.

Supplies/chemicals. A sterile, single-use needle (25G Microlance, product No. 300400, Becton Dickinson); 2mL syringe (product No. 300185, Becton Dickinson); phosphate buffer (P3813, Sigma Aldrich); isoflurane anesthetic (Holland Abbott)

And (6) anaesthetizing. Animals were deeply anesthetized with isoflurane followed by exsanguination for complete post-mortem examination.

B. Synthesis of

As used herein, "class 1 compounds" refers to aminoglycosides having neither a fluorine substituent at the C-5 position nor a guanidino group at the C-3 "position. "class 2" aminoglycoside compounds have a fluorine substituent at the C-5 position but no guanidino group at the C-3 "position. "class 3" aminoglycoside compounds have no fluorine substituent at the C-5 position but a guanidino group at the C-3 "position. "class 4" compounds are aminoglycoside compounds having a fluorine substituent at the C-5 position and a guanidino group at the C-3 "position, i.e., the compounds of the invention.

B.11 Compounds (comparative example)

B.1.1 Tobra-AHB(ABX5004)/KanaB-AHB(ABX4001)

And (a). To a suspension of tobramycin (30.8g, 65.88mmol) in dimethylsulfoxide (1.8L) was added zinc acetate dihydrate (59.14g, 269.42mmol, 4.1 equiv.). The mixture was stirred at room temperature overnight. Di-tert-butyl dicarbonate (48.88g, 223.96mmol, 3.4 eq.) is added and the reaction mixture is stirred at room temperature for 5 hours. The reaction mixture was poured into water (3L) and applied to Amberlite CG50 (H) washed with water (1L), methanol (1L), and water (1L) prior to use +Type) plug. Dimethyl sulfoxide was eluted with water (6L). With 25% NH3The product was eluted with an aqueous/methanol/water (1:2: 1; 6L) mixture. The solvent was removed in vacuo. The residue was dissolved in methanol and filtered through cotton. The solution was concentrated and dried by co-evaporation with toluene to give the desired product C1(26.74g, 34.84mmol, 53% yield) as a light yellow solid. LC/MS (System 2, method A): t is tR(min) ═ 5.89; measured 768.2(M + H) MS (M/z)+) 768.42(M + H) is calculated+)。

And (b). Tobramycin derivative C1(26.30g, 34.27mmol) was dissolved in dimethyl sulfoxide (50mL) and ethyl trifluoroacetate (4.47mL, 5.34g, 37.56mmol, 1.1 equiv.) was added dropwise. The reaction mixture was stirred at room temperature for 5 hours and poured into brine (1.8L). The precipitate was filtered off (white solid), washed with water (2.5L) and dried in vacuo by co-evaporation of the remaining water with toluene (3 ×) to give product C2(22.31g, 25.83mmol, yield 75%) as a white solid. LC/MS (System 2, method A): t is tR(min) ═ 6.48; measured 864.2(M + H) MS (M/z)+) 864.41(M + H) is calculated+)。

And (c). To a solution of (2S) -Boc-4-amino-2-hydroxybutyric acid (5.13g, 23.41mmol, 1.2 equivalents) in dimethylformamide (160mL) was added triethylamine (3.25mL, 23.31mmol, 1.2 equivalents), and the mixture was stirred at room temperature for 10 min. A solution of compound C2(16.86g, 19.52mmol, 1 eq) in dimethylformamide (160mL) was added followed immediately by HATU (8.89g, 23.38mmol, 1.2 eq) and the reaction mixture was stirred at room temperature overnight. After evaporation of dimethylformamide in vacuo, water (500mL) was added to the residue and the mixture was shaken until a white solid formed without residual oil. The white solid was filtered off, washed with sufficient water (1L) and dried in vacuo by co-evaporation of the remaining water with toluene (3 ×). The resulting product C3(18.58g, 17.44mmol, yield 89%) was isolated as a white solid. LC/MS (System 2, method A): t is t R(min) ═ 8.11; observed MS (M/z) ═ 965.2(M-Boc + H)+) Calculate 965.45(M-Boc + H)+)。

And (d). To a solution of aminoglycoside derivative C3(18.58g, 17.4mmol) in methanol (170mL) was added sodium methoxide (2.31g, 42.8mmol, 2.5 equivalents), and the reaction mixture was stirred at 30 ℃ overnight. The next day, another portion of sodium methoxide (1.39g, 25.7mmol, 1.5 equivalents) was added and the reaction mixture was stirred for another 24 hours. Amberlite CG50 (H) was added portionwise+Form) until a pH of 7 is reached. After removal of Amberlite by filtration, the remaining solution was concentrated to dryness to give the crude product C4(16.02g), which was used in the next step without further purification. LC/MS (System 2, method A): t is tR(min) ═ 6.98; measured 969.2(M + H) MS (M/z)+) 969.52(M + H) is calculated+)。

And (e) step (e). To a solution of crude product C4(50mg) in 1, 4-dioxane (1mL) was added a solution of 4N hydrogen chloride in 1, 4-dioxane (1mL) and the reaction mixture was stirred at room temperature overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/tetrahydrofuran to give the desired product ABX5004(750.93g/mol, 25mg, 0.033mmol, two step yield 61%) as the pentahydrochloride salt as a white solid. LC/MS (System 2, method C): t is t R(min) ═ 2.16; measured 569.0(M + H) MS (M/z)+) 569.31(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.78(1H),5.20(1H),4.31(1H),4.23–4.10(1H),4.10–3.63(10H),3.63–3.51(1H),3.51–3.40(2H),3.40–3.25(1H),3.25–3.11(3H),2.41–2.11(3H),2.11–1.79(3H)。

The pentahydrochloride salt of ABX4001 was synthesized starting from kanamycin B using the same reaction sequence as ABX 5004. ABX4001(766.92g/mol, pentahydrochloride salt): a white solid; LC/MS (System 2, method C): t is tR(min) ═ 2.16; measured 585.0(M + H) MS (M/z)+) 585.31(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.95(1H),5.20(1H),4.30(1H),4.22–4.10(1H),4.10–3.63(10H),3.63–3.44(5H),3.44–3.26(1H),3.20(2H),2.35–2.12(2H),2.12–1.72(2H)。

B.1.23 '-epi-KanaB-AHB (ABX 4002)/3' -epi-KanaA-AHB (ABX3002)

And (a). To a suspension of kanamycin B (30g, 62.05mmol) in dimethylsulfoxide (900mL) was added zinc acetate dihydrate (55.84g, 254.40mmol, 4.1 equiv). The mixture was stirred at room temperature overnight. Di-tert-butyl dicarbonate (46.04g, 210.96mmol, 3.4 eq.) is added and the reaction mixture is stirred at room temperature for 5 hours. In the next step, the entire reaction mixture was poured into water (3L) and applied to Amberlite CG50 (H) washed with water (1L), methanol (1L), and water (1L) prior to use+Type) (about 250g) plug. Dimethyl sulfoxide was eluted with water (6L). With 25% NH3The product was eluted with an aqueous/methanol/water (1:2: 1; 6L) mixture. The solvent was removed in vacuo. The residue was dissolved in methanol and filtered through cotton. The solution was concentrated and dried by co-evaporation with toluene to give the desired product B1(32.3g, 41.24mmol, 66% yield) as a light yellow solid. LC/MS (System 2, method A): t is t R(min) ═ 5.24; measured 784.2(M + H) MS (M/z)+) 784.42(M + H) is calculated+)。

And (b). Kanamycin B derivative B1(57.45g, 73.29mmol) was dissolved in dimethyl sulfoxide (120mL) and ethyl trifluoroacetate (9.56mL, 80.6mmol, 1.1 equiv.) was added dropwise. The reaction mixture was stirred at room temperature for 5 hours and poured into brine (3L). The precipitate was filtered off (white solid), washed with water (3L) and dried in vacuo by co-evaporation of the remaining water with toluene (3 ×) to give product B2(40.57g, 46.13mmol, yield 63%) as a white solid. LC/MS (System 2, method A): t is tR(min) ═ 5.88; MS (M/z) ═ actually measured 880.2(M + H)+) 880.40(M + H) is calculated+)。

And (c). To a solution of (2S) -Boc-4-amino-2-hydroxybutyric acid (12.49g, 56.97mmol, 1.2 eq) in dimethylformamide (150mL) was added triethylamine (7.96mL, 5.78g, 57.12mmol, 1.2 eq) and the mixture was stirred at room temperature for 10 min. A solution of compound B2(40.57g, 46.13mmol, 1 eq) in dimethylformamide (150mL) was added followed immediately by HATU (21.68g, 57.02mmol, 1.2 eq). After the reaction mixture was stirred at room temperature overnight, dimethylformamide was removed in vacuo and water (1L) was added to the oily residue. The mixture was shaken until a white solid formed without residual oil. The white solid was filtered off, washed with sufficient water (2L) and dried in vacuo by coevaporation of the remaining water with toluene (3 ×) to give product B3(37.23g, 34.44mmol, yield 75%) as a white solid. LC/MS (System 2, method A): t is t R(min) ═ 7.51; observed MS (M/z) ═ 981.2(M-Boc + H)+) Calculate 981.45(M-Boc + H)+)。

And (d). Compound B3(23.28g, 21.53mmol) was dissolved in dimethyl sulfoxide (450mL), benzoquinone (7.0g, 64.75mmol, 3 equiv.) and palladium catalyst [ (2, 9-dimethyl-1, 10-phenanthroline) -Pd (. mu. -OAc)]2(OTf)2(1.63g, 1.56mmol, 7.3 mol%; synthesized according to the procedure disclosed in N.R. Conley et al, Organometallics 26(23),5447(2007) and G. -J.ten Brink, Adv.Synth. Cat. 345,1341(2003) -the reaction mixture was stirred at room temperature overnight and then poured into brine (2.5L) -the precipitate was filtered off, washed with water (2-)3L) and dissolved in methanol (500 mL). Insoluble material was filtered off with cotton and the solution was concentrated in vacuo. The residue was dried by co-evaporation with toluene (3 ×) to give the desired product B4a (20.29g, 18.80mmol, yield 87%) as an off-white solid. LC/MS (System 2, method A): t is tR(min) ═ 8.01; observed MS (M/z) ═ 979.4(M-Boc + H)+) Calculate 979.43(M-Boc + H)+)。

And (e) step (e). To a solution of aminoglycoside derivative B4a (17.7g, 16.4mmol) in methanol (320mL) was added sodium borohydride (3.1g, 82mmol, 5 equivalents) portionwise at a temperature between-5 ℃ and 0 ℃. After the reaction mixture was stirred at 0 ℃ for 4 hours, Amberlite CG50 (H) was added +Form) until a pH of 7 is reached. Amberlite was filtered off and washed with methanol, and the filtrate was concentrated to give product B5a (16.6g, 15.4mmol, yield 94%) as a white solid. LC/MS (System 2, method A): t is tR(min) ═ 7.72; observed MS (M/z) ═ 981.2(M-Boc + H)+) Calculate 981.45(M-Boc + H)+)。

And (f). To a solution of aminoglycoside derivative B5a (10g, 9.24mmol) in methanol (90mL) was added sodium methoxide (2.5g, 46.28mmol, 5 equivalents), and the reaction mixture was stirred at 30 ℃ for 7 days. Amberlite CG50 (H) was added portionwise+Form) until a pH of 7 is reached. After removal of Amberlite by filtration, the remaining solution was concentrated to dryness to give crude product B6a (9.1g), which was used in the next step without further purification. LC/MS (System 2, method A): t is tR(min) ═ 6.49; measured 985.2(M + H) MS (M/z)+) 985.47(M + H) is calculated+)。

And (g). To a solution of crude product B6a (50mg) in 1, 4-dioxane (1mL) was added a solution of 4N hydrogen chloride in 1, 4-dioxane (1mL) and the reaction mixture was stirred at room temperature overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/dichloromethane to give the product ABX4002 as pentahydrochloride (766.92g/mol, 32.7mg, 0.043mmol, 84% over two steps) as a white solid. LC/MS (System 2, method C): t is t R(min) ═ 2.14; measured 585.0(M + H) MS (M/z)+) 585.31(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.80(1H),5.16(1H),4.40–4.19(2H),4.19–3.97(2H),3.97–3.62(8H),3.62–3.38(3H),3.38–3.35(3H),3.25–3.05(2H),2.30–1.14(2H),2.01–1.72(2H)。

The tetrahydrochloride salt of ABX3002 was synthesized starting from kanamycin a using the same reaction sequence as ABX 4002:

ABX3002(731.45g/mol, tetrahydrochloride salt): a white solid; LC/MS (System 2, method C): t is tR(min) ═ 2.16; measured 586.0(M + H) MS (M/z)+) 586.29(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.49(1H),5.15(1H),4.26(1H),4.20–3.99(3H),3.96–3.30(13H),3.26–3.09(3H),2.27–2.09(2H),2.09–1.87(1H),1.87–1.63(1H)。

Synthesis of (2S) -Boc-4-amino-2-hydroxybutyric acid (AHB-Boc)

To a solution of (2S) -4-amino-2-hydroxybutyric acid (16g, 134.3mmol) in water (140mL) was added a solution of di-tert-butyl dicarbonate (58.4g, 267.5mmol, 2 equivalents) and triethylamine (37.6mL, 269.8mmol, 2 equivalents) in tetrahydrofuran (800mL), and the reaction mixture was stirred at room temperature overnight. After removal of the tetrahydrofuran in vacuo, the aqueous solution was acidified to pH 1-2 with 1N hydrogen chloride solution and extracted with dichloromethane (3X 150 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated. The residue was dissolved in EtOAc (75mL) and shaken in heptane (800 mL). The organic solution was decanted, leaving the oil adhered to the glass wall. The oil was dissolved in EtOAc and the solution was transferred to a single-necked flask. Concentration to dryness in vacuo afforded the desired product (2S) -Boc-4-amino-2-hydroxybutyric acid (18.97g, 86.58mmol, 64% yield) as a very viscous oil. LC/MS (System 1, method D): t is t R(min) ═ 0.37; found 218.1 (M-H) MS (M/z)+) 218.10 (M-H) is calculated+)。

B.22 Compounds (comparative example)

B.2.1 Tobra-AHB-F(ABX5024)/KanaB-AHB-F(ABX4007)

Synthesis of Compound C3 was as described above for B.1.1 class 1 compounds.

And (d). Tobramycin derivative C3(34.4g, 32.3mmol) was dissolved in pyridine (320mL) and acetic anhydride (30.35mL, 321.1mmol, 10 equiv.) was added at room temperature. After stirring at room temperature overnight, another portion of acetic anhydride (6.07mL, 63.6mmol, 2 equivalents) was added and the reaction mixture was stirred for another day. The crude mixture was poured into heptane (500mL) and the suspension was filtered through a plug of silica gel. The product C5 was eluted with ethyl acetate in heptane (50-70%) (R)f0.29, 70% ethyl acetate in heptane) was used. After removal of volatiles, product C5 was obtained as a white solid (38.3g, 30.0mmol, yield 93%). LC/MS (System 2, method A): t is tR(min) ═ 10.49; observed MS (M/z) ═ 1175.4(M-Boc + H)+) Calculate 1175.51(M-Boc + H)+)。

And (e) step (e). Tobramycin derivative C5(38.3g, 30mmol) was dissolved in dichloromethane (300mL) and triethylamine (8.36mL, 60mmol, 2 equiv.), triethylamine trihydrofluoride salt (39.1mL, 240mmol, 4 equiv.) and X-TalFluor-E (41.22g, 180mmol, 3 equiv.) were added in the exact order. After stirring at room temperature overnight, the reaction mixture was cooled to 0 ℃ and saturated aqueous sodium bicarbonate solution (50mL) was carefully added. The crude mixture was warmed to room temperature with stirring and the aqueous and organic layers were separated. The aqueous solution was extracted with dichloromethane (3X 300mL), all combined organic layers were dried over anhydrous sodium sulfate, concentrated in vacuo, and the residue was taken up in ethyl acetate (30% -70%) in heptane (R) f0.37 using 70% ethyl acetate in heptane) as eluent to give product C6 as a white solid (20.5g, 16mmol, yield 54%). LC/MS (System 2, method A): t is tR(min) ═ 10.70; observed MS (M/z) ═ 1177.4(M-Boc + H)+) Calculate 1177.50(M-Boc + H)+)。

And (f). To a solution of aminoglycoside derivative C6(20.5g, 16mmol) in methanol (160mL) was added sodium methoxide (8.64g, 160mmol,10 equivalents), and the reaction mixture was stirred at 30 ℃ for three days. Amberlite CG50 (H) was added portionwise+Form) until a pH of 7 is reached. After removal of Amberlite by filtration, the remaining solution was concentrated to dryness to give the crude product C7(22.5g) as a white solid which was used in the next step without further purification. LC/MS (System 1, method A): t is tR(min) ═ 7.33; measured 971.4(M + H) MS (M/z)+) 971.52(M + H) is calculated+)。

And (g). To a solution of crude product C7(200mg) in tetrahydrofuran (2mL) were added triethylamine (85. mu.L, 0.6mmol, ca. 3 equivalents) and di-tert-butyl dicarbonate (65mg, 0.3mmol, ca. 1.5 equivalents), and the reaction mixture was stirred at room temperature overnight. After concentration in vacuo, the residue was shaken in water (10mL) and the resulting white precipitate filtered off, washed with water (2X 10mL) and dried by co-evaporation with toluene (3X) to give product C8(153mg, 0.14mmol, 99% yield over two steps). LC/MS (System 1, method B): t is t R(min) ═ 6.31; observed MS (M/z) ═ 971.4(M-Boc + H)+) Calculate 971.52(M-Boc + H)+)。

And (h). To a solution of compound C8(100mg, 0.094mmol) in 1, 4-dioxane (2mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (2mL) and the reaction mixture was stirred at room temperature overnight. The volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/tetrahydrofuran to give the product ABX5024 as pentahydrochloride salt (752.92g/mol, 59mg, 0.078mmol, yield 83%) as a white solid. LC/MS (System 2, method B): t is tR(min) ═ 1.68; measured 571.2(M + H) MS (M/z)+) 571.31(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.62(d,J=51.9Hz,1H),5.49(1H),5.19(1H),4.45–4.17(3H),4.17–4.01(1H),4.01–3.67(8H),3.61(1H),3.54–3.36(2H),3.31–3.23(1H),3.23–3.09(2H),2.47–2.27(2H),2.28–2.11(1H),2.11–1.75(3H)。19F NMR(282MHz,D2O)δ=δ-216.09(dt,2JHF=52Hz,3JHF=28Hz)。

The pentahydrochloride salt of ABX4007 was synthesized starting from kanamycin B using the same reaction sequence as ABX 5024. ABX4007(768.92g/mol,pentahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 1.62; MS (M/z) ═ actually measured 587.2(M + H)+) Calculate 587.30(M + H)+)。1H NMR(300MHz,D2O)δ=5.63(1H),5.62(d,J=51.5Hz,1H),5.19(1H),4.44–4.16(3H),4.09(1H),4.03–3.91(3H),3.91–3.61(5H),3.61–3.37(4H),3.37–3.27(1H),3.20(2H),2.47–2.30(1H),2.30–2.13(1H),2.10–1.77(2H)。19F NMR(282MHz,D2O)δ=δ-216.09(dt,2JHF=52Hz,3JHF=28Hz)。

B.2.23 '-epi-KanaB-AHB-F (ABX 4003)/3' -epi-KanaA-AHB-F (ABX3005)

Synthesis of Compound A4a was as described above for B.1.2 of class 1 compounds.

And (e) step (e). Kanamycin A derivative A4a (12g, 12.2mmol) was dissolved in pyridine (120mL) and acetic anhydride (17.34mL, 183.3mmol, 15 equiv.) was added at room temperature. After stirring at room temperature overnight, another portion of acetic anhydride (2.88mL, 30.4mmol, 2.5 equiv.) was added and the reaction mixture was stirred for another day. The crude mixture was poured into heptane (200mL) and the suspension was filtered through a plug of silica gel. Product A7a was eluted with ethyl acetate (100%) (R) f0.2, 70% EtOAc in heptane). Evaporation of the solvent gave product A7a as a white solid (14.4g, 11.7mmol, 96% yield). LC/MS (System 2, method A): t is tR(min) ═ 9.72; observed MS (M/z) ═ 1132.0(M-Boc + H)+) Calculate 1132.43(M-Boc + H)+)。

And (f). Compound A7A (14g, 11.37mmol) was dissolved in dichloromethane (120mL) and triethylamine (3.16mL, 2.29g, 22.67mmol, 2 equiv.), triethylamine trihydrofluoride salt (7.42mL, 7.34g, 45.52mmol, 4 equiv.) and X-TalFluor-E (7).81g, 34.16mmol, 3 equivalents) were added in the exact order. After stirring at room temperature overnight, the reaction mixture was cooled to 0 ℃ and a saturated aqueous solution of sodium bicarbonate (120mL) was carefully added. The crude mixture was warmed to room temperature with stirring and the aqueous and organic layers were separated. The aqueous solution was extracted with dichloromethane (3X 150mL), all combined organic layers were dried over anhydrous sodium sulfate, concentrated, and the residue was taken up in methanol (0% -4%) in dichloromethane (R)fPurification by silica gel column chromatography using 4% MeOH in DCM) as eluent gave product A8a as a beige solid (3.84g, 3.11mmol, yield 27%). LC/MS (System 2, method A): t is t R(min) ═ 9.83; observed MS (M/z) ═ 1134.0(M-Boc + H)+) Calculate 1134.42(M-Boc + H)+)。

And (g). To a solution of aminoglycoside derivative A8a (3.45g, 2.80mmol) in methanol (60mL) was added sodium borohydride (535mg, 14.02mmol, 5 equivalents) in portions at a temperature between-10 ℃ and 0 ℃. After the reaction mixture was stirred at 0 ℃ for 4 hours, Amberlite CG50 (H) was added+Form) until a pH of 7 is reached. The solution was filtered through cotton and Amberlite was washed with methanol. The solvent was removed in vacuo to give product A9a (3.09g, 2.50mmol, 89% yield) as a white solid. LC/MS (System 2, method A): t is tR(min) ═ 9.70; observed MS (M/z) ═ 1136.4(M-Boc + H)+) Calculate 1136.44(M-Boc + H)+)。

And (h). To a solution of aminoglycoside A9a (3.05g, 2.47mmol) in methanol (25mL) was added sodium methoxide (1.33g, 24.7mmol, 10 equivalents), and the reaction mixture was stirred at 30 ℃ for three days. Amberlite CG50 (H) was added portionwise+Form) until a pH of 7 is reached. After removal of Amberlite by filtration, the remaining solution was concentrated to dryness to give the crude product a10a (2.96g) as a white solid which was used in the next step without further purification. LC/MS (System 2, method A): t is tR(min) 6.00; measured 888.4(M + H) MS (M/z) +) 888.45(M + H) is calculated+)。

And (i). To a solution of crude product A10a (200mg) in tetrahydrofuran (2mL) were added triethylamine (94. mu.L, 0.68mmol, ca. 3 equivalents) and di-tert-butyl dicarbonate (74mg, 0.34mmol, ca. 1)5 equivalents) and the reaction mixture was stirred at room temperature for two days. After concentration in vacuo, the residue was shaken in water (10 mL). The white precipitate formed was filtered off, washed with water (2 × 10mL) and dried by co-evaporation with toluene (3 ×) to give product a11a (143mg, 0.15mmol, two step yield 87%) as a white solid. LC/MS (System 1, method B): t is tR(min) ═ 5.83; observed MS (M/z) ═ 888.2(M-Boc + H)+) Calculate 888.45(M-Boc + H)+)。

And (j) step. To a solution of compound a11A (100mg, 0.10mmol) in 1, 4-dioxane (5mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (2mL) and the reaction mixture was stirred overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/tetrahydrofuran to give the product ABX3005 as the tetrahydrate salt (733.44g/mol, 55mg, 0.075mmol, 75% yield) as a white solid. LC/MS (System 2, method B): t is tR(min) ═ 1.61; measured 588.2(M + H) MS (M/z)+) 588.29(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.47(d,J=51.4Hz,1H),5.22(1H),5.14(1H),4.39–4.21(2H),4.21–3.96(4H),3.96–3.49(8H),3.49–3.33(2H),3.26–3.08(3H),2.39–2.26(1H),2.24–2.06(1H),2.05–1.63(2H)。19F NMR(282MHz,D2O)δ=δ-215.60(dt,2JHF=51.7Hz,3JHF=28Hz)。

The pentahydrochloride salt of ABX4003 was synthesized starting from kanamycin B using the same reaction sequence as ABX 3005. ABX4003(768.91g/mol, pentahydrochloride salt): a white solid; LC/MS (System 2, method C): t is t R(min) ═ 2.15; measured 587.0(M + H) MS (M/z)+) Calculate 587.30(M + H)+)。1H NMR(300MHz,D2O)δ=5.56(d,J=50Hz,1H),5.47(1H),5.16(1H),4.43–4.02(4H),4.02–3.88(2H),3.88–3.67(6H),3.67–3.53(1H),3.53–3.22(3H),3.22–2.99(3H),2.32(1H),2.18(1H),2.09–1.81(2H)。19F NMR(282MHz,D2O)δ=δ-215.95(dt,2JHF=51.8Hz,3JHF=28.4Hz)。

B.33 Compounds (comparative example)

B.3.1 Kanab-AHB-Gua (ABX4009)/Tobra-AHB-Gua (ABX 5005)/Tobra-AHB-Gua-ethyl-p-aniline (ABX 5014)/Tobra-AHB-Gua-2-tert-butyl-ethyl (ABX5015)

Synthesis of Compound C4 was as described above for B.1.1 class 1 compounds.

And (e) step (e). Tobramycin derivative C4(1.9g crude product) was dissolved in 1, 4-dioxane (70mL), triethylamine (2.2mL, 1.60g, 15.77mmol, ca. 8.2 equivalents) and Q1(2.4g, 7.73mmol, ca. 4 equivalents) were added and the reaction mixture was stirred at 50 ℃ for three days. Another portion of triethylamine (2.2mL, 1.60g, 15.77mmol, about 8.2 equivalents) and Q1(2.4g, 7.73mmol, about 4 equivalents) were added and the reaction mixture was stirred at room temperature for an additional three days. After removal of all volatiles, the crude mixture was taken up in ethyl acetate (50-80%) in heptane (R)fEtOAc 0.42) was purified by silica gel column chromatography to give product C9.1 as a white solid (1.06g, 0.88mmol, two step yield 46%). LC/MS (System 2, method A): t is tR(min) ═ 8.90; measured 1211.2(M + H) MS (M/z)+) 1211.65(M + H) is calculated+)。

And (f). To a solution of compound C9.1(1.06g, 0.88mmol) in 1, 4-dioxane (20mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (20mL) and the reaction mixture was stirred at room temperature overnight. The volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/tetrahydrofuran to give the product ABX5005(792.97g/mol, 550mg, 0.69mmol, yield 79%) as the pentahydrochloride salt as a white solid. LC/MS (System 2, method C): t is t R(min) ═ 2.19; measured 611.0(M + H) MS (M/z)+) 611.34(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.79(1H),5.19(1H),4.29(1H),4.23–4.12(1H),4.12(11H),3.64–3.40(3H),3.40–3.24(1H),3.24–3.13(2H),2.42–2.11(3H),2.11–1.74(3H)。

The pentahydrochloride salt of ABX4009 was synthesized starting from kanamycin B using the same reaction sequence as ABX5005. ABX4009(808.96g/mol, pentahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 1.62; measured 627.2(M + H) MS (M/z)+) 627.33(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.89(1H),5.14(1H),4.25(1H),4.12(1H),4.08–3.73(7H),3.73–3.60(2H),3.60–3.39(6H),3.30(1H),3.22–3.08(2H),2.36–2.06(2H),2.06–1.68(2H)。

Accordingly, the hydrochloride salts of ABX5014 and ABX5015 were synthesized using the corresponding guanidine introduction reagents Q4 and Q10:

ABX5014(945.26g/mol, hexachloride): a white solid; LC/MS (System 2, method C): t is tR(min) 3.399; measured 730.2(M + H) MS (M/z)+) 730.41(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=7.42(4H),5.77(1H),5.18(1H),4.29(1H),4.16–3.93(3H),3.93–3.64(9H),3.64–3.36(5H),3.36–3.24(1H),3.19(2H),2.99(2H),2.41–2.13(3H),2.13–1.82(3H)。

ABX5015(877.12g/mol, pentahydrochloride): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 4.703; measured 695.4(M + H) MS (M/z)+) 695.43(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.78(1H),5.19(1H),4.28(1H),4.14(1H),4.11–3.63(11H),3.63–3.37(3H),3.37–3.23(3H),3.23–3.10(2H),2.39–2.10(3H),2.10–1.77(3H),1.54(2H),0.94(9H)。

Carrying a residue R on the guanidine moiety5The corresponding derivative of (2), based on the kanamycin A skeleton (KanaA-AHB-Gua-R)5) It can be obtained by following the same reaction sequence as ABX5005 described above, but starting from kanamycin A.

B.3.23 '-epi-KanaB-AHB-Gua (ABX 4005)/3' -epi-KanaA-AHB-Gua (ABX3004)

Synthesis of Compound B6a was as described above for B.1.2 of class 1 compounds.

And (g). Mixing kanamycin B derivative B6a ( 1.5g, crude mixture) was dissolved in 1, 4-dioxane (90mL), triethylamine (2.54mL, 1.84g, 18.22mmol, about 13.5 equivalents) and N, N' -bis-Boc-1-guanidinopyrazole (Q1) (1.47g, 4.74mmol, about 3.5 equivalents) were added and the reaction mixture was stirred at 50 ℃ for one day. Another portion of triethylamine (1.1mL, 0.80g, 7.89mmol, about 5.8 equivalents) and Q1(1.16g, 3.74mmol, about 2.8 equivalents) were added and the reaction mixture was stirred at 50 ℃ for an additional day. After removal of all volatiles, the crude mixture was purified by silica gel column chromatography using ethyl acetate (50-80%) in heptane to give product B12a as a white solid (613mg, 0.50mmol, 37% yield in two steps). LC/MS (System 2, method A): t is tR(min) ═ 8.43; measured 1227.3(M + H) MS (M/z)+) 1227.65(M + H) is calculated+)。

And (h). To a solution of compound B12a (233mg, 0.19mmol) in 1, 4-dioxane (2mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (2mL) and the reaction mixture was stirred at room temperature overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/diethyl ether to give the product of pentahydrochloride ABX4005(808.96g/mol, 152mg, 0.19mmol, yield 99%) as a white solid. LC/MS (System 2, method C): t is t R(min) ═ 2.32; measured 627.0(M + H) MS (M/z)+) 627.33(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.85(1H),5.20(1H),4.42–4.24(2H),4.24–4.02(2H),4.02–3.64(9H),3.64–3.44(4H),3.41–3.27(1H),3.27–3.11(2H),2.38–2.12(2H),2.12–1.73(2H)。

The tetrahydrochloride salt of ABX3004 was synthesized starting from kanamycin a using the same reaction sequence as ABX 4005: ABX3004(774.49g/mol, tetrahydrochloride salt): a white solid; LC/MS (System 2, method C): t is tR(min) ═ 2.21; measured 627.9(M + H) MS (M/z)+) 628.31(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.50(1H),5.12(1H),4.25(1H),4.19–3.99(3H),3.99–3.49(12H),3.49–3.33(1H),3.30–3.08(3H),2.29–2.07(2H),2.07–1.84(1H),1.84–1.58(1H)。

B.3.3Tobra-AHB-Gua-2-amino-ethyl (ABX 5007)/Tobra-AHB-Gua-3-amino-propyl (ABX5013)

Synthesis of Compound C4 was as described above for B.1.1 class 1 compounds.

And (e) step (e). Tobramycin derivative C4(2.67g, crude compound) was dissolved in 1, 4-dioxane (135mL), triethylamine (2.3mL, 1.67g, 16.5mmol, about 8.2 equivalents) and reagent Q2(4g, 8.27mmol, about 4 equivalents) were added, and the reaction mixture was stirred at room temperature for three days. Another portion of triethylamine (1.15mL, 0.83g, 8.25mmol, about 4.1 equivalents) and reagent Q2(2g, 4.14mmol, about 2 equivalents) were added and the mixture was stirred at 50 ℃ for an additional three days. After removal of all volatiles, the crude mixture was taken up in a solution of up to 5% methanol in ethyl acetate (R)f0.27 in ethyl acetate) was purified by silica gel column chromatography to give product C9.4 as a white solid (1.42g, 1.03mmol, two step yield 50%). LC/MS (System 2, method A): t is t R(min) ═ 9.61; measured 1384.4(M + H) MS (M/z)+) 1384.70(M + H) is calculated+)。

And (f). Compound C9.4(130mg, 0.09mmol) was dissolved in a 9:1 mixture of ethanol and water (7mL) and a solution of hydrazine monohydrate (45. mu.L, 0.90mmol, 10 equivalents) in water (1mL) was added. After stirring at room temperature for one day, the volatiles were removed in vacuo and the residue was taken up in 5% strength methanol in ethyl acetate (R)f0.69 using 5% methanol in ethyl acetate) was purified by silica gel column chromatography to give product C10.4 as a white solid (70mg, 0.056mmol, yield 62%). LC/MS (System 2, method A): t is tR(min) ═ 8.29; measured 1254.4(M + H) MS (M/z)+) 1254.69(M + H) is calculated+)。

And (g). To a solution of compound C10.4(65mg, 0.052mmol) in 1, 4-dioxane (1mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (1mL) and the reaction mixture was stirred at room temperature overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/tetrahydrofuran to give the product ABX5007 as the hexahydrate salt (872.48g/mol, 36mg, 0.041mmol,yield 79%) as a white solid. LC/MS (System 2, method A): t is tR(min) ═ 0.95; MS (M/z) ═ actually measured 654.2(M + H)+) 654.38(M + H) is calculated +)。1H NMR(300MHz,D2O)δ=5.79(1H),5.20(1H),4.36–4.26(1H),4.26–4.14(1H),4.14–3.52(15H),3.52–3.42(1H),3.37–3.25(3H),3.25–3.13(2H),2.42–2.11(3H),2.11–1.75(3H)。

Accordingly, the hexahydrate salt of ABX5013 was synthesized using the corresponding guanidine introduction reagent Q3.

ABX5013(886.51g/mol, hexachloride): a white solid; LC/MS (System 2, method C): t is tR(min) ═ 2.043; measured 668.2(M + H) MS (M/z)+) 668.39(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.78(1H),5.19(1H),4.29(1H),4.16(1H),4.11–3.63(11H),3.63–3.51(2H),3.51–3.23(4H),3.19(2H),3.10(2H),3.39–2.11(3H),2.11–1.75(5H)。

Carrying a residue R on the guanidine moiety5The corresponding derivative of (2), based on the kanamycin A skeleton (KanaA-AHB-Gua-R)5) Or based on the kanamycin B skeleton (KanaB-AHB-Gua-R)5) They can be obtained by following the same reaction sequence as the above ABX5007 but starting from kanamycin A or kanamycin B, respectively.

B.4 Compounds of type 4

B.4.1 Kanab-AHB-F-Gua (ABX4006)/Tobra-AHB-F-Gua (ABX 5006)/Tobra-AHB-F-Gua-2-tert-butyl-ethyl (ABX5025)/Tobra-AHB-F-Gua- (S) -pyrrolidin-2-yl-methyl (ABX 5026)/Tobra-AHB-F-Gua-cyclopropylmethyl (ABX 5027)/Tobra-AHB-F-Gua-2-hydroxy-ethyl (ABX 5029)/Tobra-AHB-F-Gua-methyl (ABX 5031)/Tobra-AHB-F-Gua-ethyl (ABX 5032)/Tobra-AHB-F-Gua-ethyl-p-phenylamine (ABX 5036)/Tobra-F-Gua-2- (methylamino) ethyl (ABX5038) ) /Tobra-AHB-F-Gua- (R) -pyrrolidin-3-yl-methyl (ABX5039)/Tobra-AHB-F-Gua- (S) -morpholin-2-yl-methyl (ABX 5040)/Tobra-AHB-F-Gua-3-hydroxy-propyl (ABX5041)/Tobra-AHB-F-Gua- (S) -pyrrolidin-3-yl-methyl (ABX5042)/Tobra-AHB-F-Gua- (R) -morpholin-2-yl-methyl (ABX5043)/Tobra-AHB-F-Gua- (S) -piperidin-3-yl-methyl (ABX 5044)/Tobra-AHB-F-Gua-tetrahydrofuran-2-yl-methyl (ABX 46 5041) ) /Tobra-AHB-F-Gua-pyridin-4-yl-methyl (ABX 5048)/Tobra-AHB-F-Gua-imidazol-5-yl-methyl (ABX 5050)/Tobra-AHB-F-Gua-tetrahydrofuran-3-yl-methyl (ABX5051)

Synthesis of Compound C7 was as described above for B.2.1 class 2 compounds.

And (g). Tobramycin derivative C7(1g, crude mixture) was dissolved in 1, 4-dioxane (50mL), triethylamine (1.42mL, 1.04g, 10.18mmol, about 13.7 equivalents) and Q1(0.82g, 2.64mmol, about 3.6 equivalents) were added, and the reaction mixture was stirred at 45 ℃ for three days. Another portion of triethylamine (0.6mL, 0.44g, 4.30mmol, about 5.8 equivalents) and Q1(0.65g, 2.09mmol, about 2.8 equivalents) were added and the reaction mixture was stirred at 45 ℃ overnight. After removal of all volatiles, the crude mixture was taken up in ethyl acetate (30-100%) in heptane (R)fEtOAc solution 0.48) was purified by silica gel column chromatography to give product C11.1 as a white solid (520mg, 0.43mmol, 58% over two steps). LC/MS (System 2, method A): t is tR(min) ═ 9.43; measured 1213.2(M + H) MS (M/z)+) 1213.65(M + H) is calculated+)。

And (h). To a solution of compound C11.1(500mg, 0.41mmol) in 1, 4-dioxane (6mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (6mL) and the reaction mixture was stirred at room temperature overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/DCM to give the product ABX5006(794.96g/mol, 267mg, 0.34mmol, yield 82%) as the pentahydrochloride salt as a white solid. LC/MS (System 2, method A): t is t R(min) ═ 0.96; MS (M/z) ═ actually measured 613.0(M + H)+) 613.33(M + H) is calculated+)。

1H NMR(400MHz,D2O):δ=5.64(d,J=52Hz,1H,H-5),5.49(1H,H-1′),5.17(1H,H-1″),4.44–4.24(3H,H-1,H-4,H-8),4.14(1H,H-6),4.00–3.93(2H,H-2″,H-6″),3.90–3.80(2H,H-3,H-5′),3.79–3.67(5H,H-2′,H-4′,H-4″,H-3″,H-6″),3.53–3.42(2H,H-6′,H-5″),3.25(1H,H-6′),3.19(2H,H-10),2.42–2.33(2H,H-2eq,H-3′eq),2.24–2.13(1H,H-9),2.13–2.00(1H,H-3′ax),2.00–1.92(1H,H-9),1.92–1.83(1H,H-2ax)。

13C NMR(100MHz,D2O includes MeOH as internal standard): δ 175.97(C ═ O, C-7),158.36(C ═ NH, C-11),100.35(CH, C-1 "), 89.53(CH, C-1'), 87.45(d, J), and so onCF=181Hz,CH,C-5),78.03(d,JCF=16Hz,CH,C-6),72.91(CH,C-2″),72.43(d,JCF=17Hz,CH,C-4),69.74(3CH,C-8,C-5′,C-4″),68.52(CH,C-5″),64.61(CH,C-4′),61.11(CH2,C-6″),57.28(CH,C-3″),47.61(CH,C-2′),47.37(d,JCF=5Hz,CH,C-3),46.16(d,JCF=4Hz,CH,C-1),40.10(CH2,C-6′),37.13(CH2,C-10),30.89(CH2,C-9),29.87(CH2,C-3′),29.83(CH2,C-2)。

19F NMR(282MHz,D2O):δ=-215.9(dt,2JHF=52Hz,3JHF=27Hz)。

All signals were determined by one-and two-dimensional NMR spectroscopy. Recording and analyzing1H、13C. APT, COSY and HSQC.

Numbering 1H-NMR(ppm) 13C-NMR(ppm)
1 4.39 46.16(3JCF=4Hz)
2 1.87 and 2.36 29.83
3 3.85 47.37(3JCF=5Hz)
4 4.30(3JHF=28Hz) 72.43(2JCF=17Hz)
5 5.65(2JHF=52Hz) 87.45(1JCF=181Hz)
6 4.14(3JHF=28Hz) 78.03(2JCF=16Hz)
7 - 175.97
8 4.30 69.74
9 1.95 and 2.16 30.89
10 3.19 37.13
11 - 158.36
1′ 5.50 89.53
2′ 3.75 47.61
3′ 2.04 and 2.36 29.87
4′ 3.75 64.61
5′ 3.87 69.74
6′ 3.25 and 3.50 40.10
1″ 5.17 100.35
2″ 3.96 72.91
3″ 3.70 57.28
4” 3.70 69.74
5” 3.45 68.52
6” 3.71 and 3.96 61.11

The pentahydrochloride salt of ABX4006 was synthesized starting from kanamycin B using the same reaction sequence as ABX 5006. ABX4006(810.96g/mol, pentahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 1.63; measured 629.2(M + H) MS (M/z)+) 629.33(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.63(d,J=52Hz,1H),5.63(1H),5.17(1H),4.47–4.13(3H),4.13–4.02(1H),4.02–3.91(2H),3.91–3.61(6H),3.61–3.28(4H),3.38–3.26(1H),3.26–3.07(2H),2.46–2.26(1H),2.26–2.09(1H),2.07–1.74(2H)。19F NMR(282MHz,D2O)δ=-215.90(dt,2JHF=53Hz,3JHF=27Hz)。

Accordingly, the hydrochloride salts of ABX5025, ABX5026, ABX5027, ABX5029, ABX5031, ABX5032, ABX5036, ABX5038, ABX5039, ABX5040, ABX5041, ABX5042, ABX5043, ABX5044, ABX5046, ABX5048, ABX5050 and ABX5051 are synthesized using the corresponding guanidine introduction reagents Q4, Q5, Q6, Q7, Q8, Q9, Q10, Q21, Q11, Q17, Q20, Q12, Q18, Q16, Q27, Q24, Q25 and Q28, respectively:

ABX5025(879.12g/mol, pentahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 2.34; measured 697.4(M + H) MS (M/z) +) 697.43(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.63(d,J=53.2Hz,1H),5.49(1H),5.17(1H),4.52–4.04(4H),4.06–3.59(9H),3.50(2H),3.35–3.07(5H),2.44–2.26(2H),2.27–1.75(4H),1.54(2H),0.94(9H)。19F NMR(282MHz,D2O)δ=-215.92(dt,2JHF=52Hz,3JHF=26Hz)。

ABX5026(914.55g/mol, hexahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 3.81; measured 696.3(M + H) MS (M/z)+) 696.41(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.63(d,J=52.5Hz,1H),5.50(1H),5.18(1H),4.51–4.04(4H),4.05–3.55(12H),3.58–3.33(4H),3.34–3.08(3H),2.53–1.67(10H)。19F NMR(282MHz,D2O)δ=-215.85(dt,2JHF=52Hz,3JHF=27Hz)。

ABX5027(849.05g/mol, pentahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 4.99; measured 667.2(M + H) MS (M/z)+) 667.38(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.64(d,J=52.5Hz,1H),5.49(1H),5.18(1H),4.47–4.06(4H),4.02–3.62(9H),3.54–3.40(2H),3.32–3.03(5H),2.46–2.26(2H),2.24–1.75(4H),1.15–0.96(1H),0.58(2H),0.26(2H)。19F NMR(282MHz,D2O)δ=δ-215.85(dt,2JHF=52Hz,3JHF=27Hz)。

ABX5029(839.00g/mol, pentahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 1.64; measured 657.2(M + H) MS (M/z)+) 657 calculation.36(M+H+)。1H NMR(300MHz,D2O)δ=5.62(d,J=53.9Hz,1H),5.49(1H),5.18(1H),4.47–4.05(4H),4.05–3.57(11H),3.57–3.34(4H),3.33–3.11(3H),2.51–2.28(2H),2.28–1.75(4H)。19F NMR(282MHz,D2O)δ=δ-215.85(dt,2JHF=54Hz,3JHF=29Hz)。

ABX5031(808.98g/mol, pentahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 1.65; measured 627.2(M + H) MS (M/z)+) 627.35(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.64(d,J=52.6Hz,1H),5.50(1H),5.18(1H),4.47–4.05(4H),4.05–3.63(9H),3.56–3.41(2H),3.33–3.10(3H),2.88(3H),2.48–2.26(2H),2.26–1.78(4H)。19F NMR(282MHz,D2O)δ=δ-215.85(dt,2JHF=53Hz,3JHF=28Hz)。

ABX5032(823.01g/mol, pentahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 2.20; MS (M/z) ═ actually measured 641.3(M + H)+) 641.36(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=δ5.62(d,J=52.4Hz,1H),5.48(1H),5.18(1H),4.47–4.04(4H),4.053–3.64(9H),3.56–3.42(2H),3.34–3.08(5H),2.48–2.29(2H),2.28–1.71(4H),1.21(3H)。19F NMR(282MHz,D2O)δ=-215.80(dt,2JHF=51Hz,3JHF=29Hz)。

ABX5036(950.57g/mol, hexahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 4.57; measured 732.2(M + H) MS (M/z)+) 732.41(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=7.42(4H),5.64(d,J=52.2Hz,1H),5.50(1H),5.18(1H),4.51–4.04(4H),4.04–3.62(9H),3.62–3.38(4H),3.35–3.11(3H),2.99(2H),2.48–2.29(2H),2.27–1.81(4H)。19F NMR(282MHz,D2O)δ=-215.91(dt,2JHF=53Hz,3JHF=28Hz)。

ABX5038(888.50g/mol, hexahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 1.65; measured 670.4(M + H) MS (M/z)+) 670.39(M + H) is calculated+)。

ABX5039(914.54g/mol, hexahydrochloride salt): a white solid; LC/MS (System 2, method B): t is t R(min) ═ 1.64; measured 696.4(M + H) MS (M/z)+) 696.41(M + H) is calculated+)。

ABX5040(930.54g/mol, hexahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 1.63; measured 712.4(M + H) MS (M/z)+) 712.40(M + H) is calculated+)。

ABX5041(853.03g/mol, pentahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 2.04; measured 671.4(M + H) MS (M/z)+) 671.37(M + H) is calculated+)。

ABX5042(914.54g/mol, hexahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 2.10; measured 696.3(M + H) MS (M/z)+) 696.41(M + H) is calculated+)。

ABX5043(930.54g/mol, hexahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 1.95; measured 712.4(M + H) MS (M/z)+) 712.40(M + H) is calculated+)。

ABX5044(928.57g/mol, hexahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 3.04; measured 710.4(M + H) MS (M/z)+) 710.42(M + H) is calculated+)。

ABX5046(879.07g/mol, pentahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 4.53; measured 697.4(M + H) MS (M/z)+) 697.39(M + H) is calculated+)。

ABX5048(922.52g/mol, hexahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 2.08; measured 704.3(M + H) MS (M/z) +) 704.37(M + H) is calculated+)。

ABX5050(911.50g/mol, hexahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 2.11; MS (m/z) ═ actually measured 693.3(M+H+) 693.37(M + H) is calculated+)。

ABX5051(879.07g/mol, pentahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 5.57; measured 697.4(M + H) MS (M/z)+) 697.39(M + H) is calculated+)。

Carrying a residue R on the guanidine moiety5The corresponding derivative of (2), based on the kanamycin A skeleton (KanaA-AHB-F-Gua-R)5) It can be obtained by following the same reaction sequence as ABX5006 described above, but starting from kanamycin A.

B.4.23 '-epi-KanaB-AHB-F-Gua (ABX 4004)/3' -epi-KanaA-AHB-F-Gua (ABX3003)

Synthesis of Compound B10a was as described above for B.2.2 of class 2 compounds.

And (i). Kanamycin B derivative B10a (435mg, crude compound) was dissolved in 1, 4-dioxane (25mL), triethylamine (0.37mL, 0.27g, 2.65mmol, ca. 8 equivalents) and N, N' -bis-Boc-1-guanidinopyrazole (Q1) (0.41g, 1.32mmol, ca. 4 equivalents) were added and the reaction mixture was stirred at 45 ℃ for one day. Another portion of triethylamine (0.62mL, 0.45g, 4.44mmol, ca. 13.5 equiv.) and N, N' -bis-Boc-1-guanidinopyrazole (Q1) (0.68g, 2.19mmol, ca. 6.6 equiv.) were added and the reaction mixture was stirred at 45 ℃ for an additional day. After removing all volatiles, the reaction mixture was stirred at 45 ℃ for another day and the crude mixture was taken up in ethyl acetate (10-80%) in heptane (R) fEtOAc solution 0.41) was purified by silica gel column chromatography to give product B13a as a white solid (220mg, 0.18mmol, 41% over two steps). LC/MS (System 2, method A): t is tR(min) ═ 8.77; measured 1229.2(M + H) MS (M/z)+) 1229.64(M + H) is calculated+)。

And (j) step. To a solution of compound B13a (200mg, 0.16mmol) in 1, 4-dioxane (2mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (2mL) and the reaction mixture was stirred overnight. Volatiles were removed in vacuo to give a white solid by distillation from methanol/DCThis was purified by recrystallization/precipitation in M to give the product ABX4004 as pentahydrochloride salt (810.96g/mol, 115mg, 0.14mmol, yield 89%) as a white solid. LC/MS (System 2, method C): t is tR(min) ═ 2.06; measured 629.0(M + H) MS (M/z)+) 629.33(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.60(d,J=52Hz,1H),5.50(1H),5.17(1H),4.47–4.21(3H),4.22–4.04(2H),3.96(2H),3.92–3.62(7H),3.62–3.41(2H),3.31(1H),3.19(2H),2.49–2.28(1H),2.29–2.10(1H),2.06–1.76(2H)。19F NMR(300MHz,D2O)δ=-215.77(dt,2JHF=50Hz,3JHF=27Hz)。

The tetrahydrochloride salt of ABX3003 was synthesized starting from kanamycin a using the same reaction sequence as ABX 4004: ABX3003(775.47g/mol, tetrahydrochloride salt): a white solid; LC/MS (System 2, method C): t is tR(min) ═ 2.23; MS (M/z) — 630.0(M + H) measured+) 630.31(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.51(d,J=51.7,1H),5.26(1H),5.15(1H),4.50–4.24(2H),4.24–3.99(4H),3.99–3.53(8H),3.53–3.39(2H),3.33–3.12(3H),2.33(1H),2.15(1H),2.03–1.88(1H),1.79(1H)。19F NMR(300MHz,D2O)δ=-215.32(dt,2JHF=53Hz,3JHF=27Hz)。

B.4.3. Tobra-AHB-F-Gua-2-amino-ethyl (ABX 5020)/Tobra-AHB-F-Gua-3-amino-propyl (ABX5030)/Tobra-AHB-F-Gua- (R) -3-amino-butyl (ABX5047)

Synthesis of Compound C7 was as described above for B.2.1 class 2 compounds.

And (g). Tobramycin derivative C7(1g, crude mixture) was dissolved in 1, 4-dioxane (50mL), triethylamine (1.12mL, 0.81g, 8.0mmol, about 10.8 equivalents) and reagent Q2(2g, 4.14mmol, about 5.6 equivalents) were added, and the reaction mixture was stirred at 45 ℃ overnight. Another portion of triethylamine (0.56mL, 0.41g, 4) was added0mmol, about 5.4 equivalents) and reagent Q2(1g, 2.07mmol, about 2.7 equivalents). After removal of all volatiles, the mixture was stirred at 45 ℃ for an additional 4 days and the crude mixture was taken up in 100% EtOAc in heptane (R)fEtOAc solution 0.58) was purified by silica gel column chromatography to give product C11.16 as a white solid (424mg, 0.31mmol, two step yield 42%). LC/MS (System 2, method A): t is tR(min) ═ 9.43; measured 1386.6(M + H) MS (M/z)+) 1386.69(M + H) is calculated+)。

And (h). Compound C11.16(400mg, 0.29mmol) was dissolved in a 9:1 mixture of ethanol and water (25mL) and a solution of hydrazine hydrate (145. mu.L, 2.9mmol, 10 equivalents) in water (7.5mL) was added. After stirring at room temperature for one day, the volatiles were removed in vacuo and the residue taken up in 5% methanol in EtOAc (R)f0.29 using 5% MeOH in EtOAc) was purified by silica gel column chromatography to give the product C12.16 as a white solid (207mg, 0.16mmol, yield 57%). LC/MS (System 2, method A): t is t R(min) ═ 8.22; measured 1256.6(M + H) MS (M/z)+) 1256.69(M + H) is calculated+)。

And (i). To a solution of compound C12.16(163mg, 0.13mmol) in 1, 4-dioxane (2mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (2mL) and the reaction mixture was stirred at room temperature overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/DCM to give the product ABX5020 as the hexahydrate salt (874.49g/mol, 80mg, 0.091mmol, yield 70%) as a white solid. LC/MS (System 1, method B): t is tR(min) ═ 1.61; measured 656.4(M + H) MS (M/z)+) 656.37(M + H) is calculated+)。

1H NMR(400MHz,D2O):δ=5.65(d,J=52Hz,1H,H-5),5.50(1H,H-1′),5.19(1H,H-1″),4.43–4.25(3H,H-1,H-4,H-8),4.23-4.09(1H,H-6),4.02–3.93(2H,H-2″,H-6″),3.91–3.81(2H,H-3,H-5′),3.81–3.68(5H,H-2′,H-4′,H-3″,H-4″,H-6″),3.68–3.61(2H,H-12),3.55–3.47(2H,H-6′,H-5″),3.33–3.23(3H,H-13,H-6′),3.23–3.17(2H,H-10),2.43–2.35(2H,H-2eq,H-3′eq),2.25–2.14(1H,H-9),2.14–2.02(1H,H-3′ax),2.02–1.84(2H,H-9,H-2ax)。

13C NMR(100MHz,D2O includes MeOH as internal standard): δ 175.90(C ═ O, C-7),157.65(C ═ NH, C-11),100.44(CH, C-1 "), 89.53(CH, C-1'), 87.35(d, J), and so onCF=181Hz,CH,C-5),77.88(d,JCF=17Hz,CH,C-6),72.94(CH,C-2″),72.40(d,JCF=17Hz,CH,C-4),69.74(2CH,C-8,C-5′),69.60(CH,C-4″),68.47(CH,C-5″),64.60(CH,C-4′),61.09(CH2,C-6″),57.47(CH,C-3″),47.61(CH,C-2′),47.37(d,JCF=5Hz,CH,C-3),46.21(d,JCF=5Hz,CH,C-1),40.10(CH2,C-6′),38.95(CH2,C-12),38.16(CH2,C-13),37.13(CH2,C-10),30.93(CH2,C-9),29.86(CH2,C-3′),29.84(CH2,C-2)。

19F NMR(282MHz,D2O):δ=-215.9(dt,2JHF=53Hz,3JHF=27Hz)。

All signals were determined by one-and two-dimensional NMR spectroscopy. Recording and analyzing1H、13C. APT, COSY and HSQC.

Accordingly, the hexahydrochlorides of ABX5030 and ABX5047 were synthesized using the corresponding guanidine introduction reagents Q3 and Q29:

ABX5030(888.50g/mol, hexahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 1.93; MS (M/z) ═ actually measured 670.2(M + H)+) 670.39(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.63(d,J=52.3Hz,1H),5.49(1H),5.18(1H),4.48–4.05(4H),4.05–3.61(9H),3.57–3.42(2H),3.43–3.32(2H),3.32–3.14(3H),3.13–3.02(2H),2.47–2.27(2H),2.27–2.10(1H),2.12–1.75(5H)。19F NMR(282MHz,D2O)δ=-215.9(dt,2JHF=52Hz,3JHF=27Hz)。

ABX5047(902.53g/mol, hexahydrochloride salt): a white solid; LC/MS (System 1, method B): t is t R(min) ═ 2.18; measured 684.4(M + H) MS (M/z)+) 684.41(M + H) is calculated+)。

The pentahydrochloride salt of 3 '-epi-KanaB-AHB-F-Gua-2-amino-ethyl (ABX4008) and the tetrahydrate salt of 3' -epi-KanaA-AHB-F-Gua-2-amino-ethyl (ABX3006) were synthesized from the corresponding precursors B10a and a10a, respectively (see b.2.2 for class 2 compounds), in the same reaction sequence (steps (g) to (i)) as described above for ABX 5020.

ABX4008(890.47g/mol, hexahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 1.63; measured 672.2(M + H) MS (M/z)+) 672.37(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.60(d,J=52.1Hz,1H),5.50(1H),5.18(1H),4.45–4.24(3H),4.24–4.03(2H),4.03-3.90(2H),3.90–3.58(9H),3.56–3.43(2H),3.37–3.24(3H),3.24-3.13(2H),2.46–2.27(1H),2.27–2.08(1H),2.04–1.79(2H)。19F NMR(282MHz,D2O)δ=-215.72(dt,2JHF=52Hz,3JHF=27Hz)。

ABX3006(855.00g/mol, pentahydrochloride salt): a white solid; LC/MS (System 2, method B): t is tR(min) ═ 1.63; measured 673.2(M + H) MS (M/z)+) 673.35(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.50(d,J=51.3Hz,1H),5.26(1H),5.16(1H),4.46–4.24(2H),4.24–4.11(2H),4.11–3.99(2H),3.99–3.59(10H),3.57–3.40(2H),3.33–3.23(3H),3.22–3.11(2H),2.43–2.25(1H),2.26–2.10(1H),2.03–1.70(2H)。19F NMR(282MHz,D2O)δ=-215.30(dt,2JHF=52Hz,3JHF=28Hz)。

Carrying a residue R on the guanidine moiety5The corresponding derivative of (2), based on the kanamycin A skeleton (KanaA-AHB-F-Gua-R)5) Or based on the kanamycin B skeleton (KanaB-AHB-F-Gua-R)5) They can be obtained by following the same reaction sequence as above ABX5020, but starting from kanamycin A or kanamycin B, respectively.

B.4.43 '-methyl-3' -epi-KanaB-AHB-F-Gua (ABX4011)

Synthesis of Compound B4a was as described above for B.1.2 of class 1 compounds.

And (e) step (e). 3' -oxo-kanamycin B derivative B4a (13g, 12.04mmol) was dissolved in tetrahydrofuran (700mL) and cooled to-15 ℃. Methyl magnesium bromide (3M diethyl ether solution, 96.25mL, 288.75mmol, 24 equivalents) was added dropwise and stirred at-10 ℃ for one hour, 0 ℃ for 2 hours, then the reaction mixture was slowly warmed to 15 ℃ over four to five hours. After the reaction mixture was stored in a-20 ℃ refrigerator overnight, the pH was adjusted to 6 by adding 3N aqueous hydrogen chloride solution under cooling (0 ℃) and the solution was extracted with ethyl acetate (3 ×). The combined organic layers were dried over anhydrous sodium sulfate and concentrated. The crude product was taken up in methanol (0-5%) in ethyl acetate (R) f0.74 using 5% methanol in ethyl acetate) was purified by silica gel column chromatography to give product B14a as a white solid (2.2g, 2.01mmol, yield 17%). LC/MS (System 2, method A): t is tR(min) ═ 7.81; observed MS (M/z) ═ 995.4(M-Boc + H)+) Calculate 995.46(M-Boc + H)+)。

And (f). Compound B14a (2g, 1.9mmol) is dissolved in pyridinePyridine (20mL), acetic anhydride (2.15mL, 22.8mmol, 12 equivalents) was added at room temperature and stirred for five days. The crude mixture was diluted with heptane (200mL) and the suspension was filtered through a plug of silica gel and washed with heptane (500 mL). The product B15a (R) was eluted with a mixture of heptane and ethyl acetate (1:1)f0.29, 70% ethyl acetate in heptane) was used. Evaporation of the solvent gave product B15a as a white solid (2.0g, 1.53mmol, 81% yield). LC/MS (System 1, method A): t is tR(min) ═ 11.06; MS (M/z) ═ found 1205.2(M-Boc + H)+) Calculate 1205.52(M-Boc + H)+)。

And (g). And (g). Kanamycin B derivative B15a (2g, 1.53mmol) was dissolved in dichloromethane (15mL) and triethylamine (0.43mL, 3.09mmol, 2 equiv.), triethylamine trihydrofluoride salt (1.0mL, 6.13mmol, 4 equiv.) and X-TalFluor-E (1.05g, 4.6mmol, 3 equiv.) were added in the exact order. After stirring at room temperature overnight, the reaction mixture was cooled to 0 ℃ and a saturated aqueous solution of sodium bicarbonate (20mL) was carefully added. The crude mixture was warmed to room temperature with stirring and the aqueous and organic layers were separated. The aqueous solution was extracted with dichloromethane (3 ×), all combined organic layers were dried over anhydrous sodium sulfate, concentrated in vacuo, and the residue was taken up in ethyl acetate (0% -50%) in heptane (R) f0.52 using 70% ethyl acetate in heptane) as eluent to give product B16a as a white solid (600mg, 0.46mmol, yield 30%). LC/MS (System 2, method A): t is tR(min) ═ 10.46; observed MS (M/z) ═ 1207.4(M-Boc + H)+) Calculate 1207.51(M-Boc + H)+)。

And (h). To a solution of aminoglycoside B16a (600mg, 0.46mmol) in methanol (5mL) was added sodium methoxide (248mg, 4.6mmol, 10 equivalents), and the reaction mixture was stirred at room temperature for four days. Amberlite CG50 (H) was added portionwise+Form) until a pH of 5-6 is reached. After removal of Amberlite by filtration, the remaining solution was concentrated to dryness to give the crude product B17a (700mg) as a white solid, which was used in the next step without further purification. LC/MS (System 2, method A): t is tR(min) ═ 6.77; measured 1023.5(M + Na) for MS (M/z)+) 1023.51(M + Na) was calculated+)。

And (i). Crude product B17a (700mg) was dissolved in 1, 4-dioxane (35mL), triethylamine (0.50mL, 7.8mmol, ca. 11 equivalents) and Q1(1.1g, 7.8mmol, ca. 11 equivalents) were added and the reaction mixture was stirred at 50 ℃ for three days. After removal of all volatiles, the crude mixture was taken up in ethyl acetate (30-100%) in heptane (R)f0.58 in ethyl acetate) was purified by silica gel column chromatography to give product B18a as a white solid (63mg, 0.051mmol, two-step yield 11%). LC/MS (System 1, method A): t is t R(min) ═ 9.23; measured 1243.4(M + H) MS (M/z)+) 1243.66(M + H) is calculated+)。

And (j) step. To a solution of compound B18a (50mg, 0.040mmol) in 1, 4-dioxane (5mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (5mL) and the reaction mixture was stirred overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/dichloromethane to give the product ABX4011 as pentahydrochloride salt (824.97g/mol, 31mg, 0.038mmol, yield 94%) as a white solid. LC/MS (System 1, method B): t is tR(min) ═ 1.63; measured 643.3(M + H) MS (M/z)+) 643.34(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.63(1H),5.62(d,J=52Hz,1H),5.17(1H),4.46–4.03(4H),4.03–3.61(8H),3.61–3.42(3H),3.42–3.25(1H),3.25–3.11(2H),2.43–2.27(1H),2.27–2.08(1H),2.02-1.74(2H),1.53(3H)。19F NMR(282MHz,D2O)δ=-215.70(dt,2JHF=52Hz,3JHF=28Hz)。

The corresponding derivative, based on the 3 ' -epi-kanamycin A skeleton (3 ' -Me-3 ' -epi-KanaA-AHB-F-Gua), can be obtained in the same reaction sequence as above ABX4011, but starting from kanamycin A.

B.4.56N-2-amino-ethyl-Tobra-AHB-F-Gua (ABX5037)

ABX5037 was synthesized according to the procedure described for ABX5020 (section b.4.3 above), except for the modified form of step (a) and the additional steps (h) and (i)), each of which is described below.

And (a). To a suspension of tobramycin (30.0g, 64mmol) in dimethylsulfoxide (1.3L) was added ZnOAc2X2H2O (42.3g, 192.7mmol, 3 equivalents), and the mixture was stirred at room temperature overnight. After addition of BND (20.1g, 64.2mmol, 1 eq.) the reaction was stirred at room temperature for a further day. Di-tert-butyl dicarbonate (30.8g, 141.1mmol, 2.2 equiv.) is added and the reaction stirred at room temperature for a further 4 hours, then poured into water (4.2L) and applied to Amberlite CG50 (H) washed with water (1L), methanol (1L), and water (1L) before use +Form) on the plug. Dimethyl sulfoxide was eluted with water (6L). Product C13 with 25% NH3An aqueous/methanol/water (1:2: 1; 6L) mixture. The solvent was removed in vacuo and the residue was dissolved in methanol. Insoluble matter was filtered off with cotton. The filtrate was concentrated in vacuo and dried by co-evaporation with toluene (3 ×) to give product C13(29.1g, 36.3mmol, 57%) as a white solid. LC/MS (System 2, method A): t is tR(min) ═ 5.84; MS (M/z) — measured 802.2(M + H)+) 802.41(M + H) is calculated+)。

And (h). Compound C19(3.3g, 2.65mmol) was dissolved in methanol (70mL) and carbon-supported palladium hydroxide (20 wt%) (70mg, 0.13mmol, 5 mol%) and acetic acid (0.66mL, 0.69g, 11.5mmol, 4.3 equiv) were added. After flushing the reaction vessel with hydrogen, the reaction was stirred at room temperature under a hydrogen atmosphere (1 atm). After two days, the reaction was filtered over celite and concentrated to dryness to give product C20 as a white solid (2.3g, 2.06mmol, 78%). LC/MS (System 2, method A): t is tR(min) ═ 7.31; measured 1113.4(M + H) MS (M/z)+) 1113.59(M + H) is calculated+)。

And (i). Cesium (II) hydroxide monohydrate (302mg, 1.80mmol, 1 equiv.) and molecular sieve: (molecular sieve600mg) was dissolved in DMF (20mL), the suspension was stirred at 35 ℃ for 10 min, then Compound C20(2g, 1.80mmol, 1 equiv.) was added. After stirring for 2 h at 35 ℃ N- (2-bromoethyl) phthalimide (3.01g, 11.85mmol, 6.6 equivalents) was added and the reaction mixture was stirred for 24 h at 35 ℃. The molecular sieve was filtered off with a glass filter and the solution was concentrated. The crude product was purified by column chromatography on silica gel using 50-100% ethyl acetate in heptane to remove impurities, then product C21(259mg, 0.2mmol, 11%) was eluted using 1-3% methanol in ethyl acetate. LC/MS (System 2, method A): t is t R(min) ═ 8.13; measured 1286.6(M + H) MS (M/z)+) 1286.64(M + H) is calculated+)。

Following step (i) which provides product C21, steps (j) and (k) are carried out under the conditions described for steps (h) and (i) in the synthesis of ABX5020 (section b.4.3, supra), thereby providing the final product ABX 5037.

N-benzyloxycarbonyloxy-5-norbornene-2, 3-dicarboximide (BND)

A solution of 4-benzylchloroformate (39mL, 46.61g, 0.273mol) in tetrahydrofuran (900mL) was cooled to 0 deg.C and N-hydroxy-dicarboximide (47.5g, 0.265mol) was added. After subsequent addition of triethylamine (36.9mL, 26.77g, 0.265mol) at 0 deg.C, the reaction mixture was warmed to room temperature and stirred for an additional 4 hours. The reaction was cooled to-5 ℃ (ice salt bath) and the solid was filtered off. The filtrate was concentrated and the residue was dissolved in methanol (900 mL). The solid formed was filtered through a glass filter and dried in air to give N-benzyloxycarbonyloxy-5-norbornene-2, 3-dicarboximide as a white solid (48.5g, 0.15mol, 58%). LC/MS (System 1, method D): t is tR(min) ═ 1.89; measured 352.1(M + K) MS (M/z)+) 352.06(M + K) is calculated+)。1H NMR(300MHz,CDCl3)δ=7.38(5H),6.17(2H),5.27(2H),3.45(2H),3.31(2H),1.78(1H),1.54(1H)。

The corresponding derivatives carrying a 2-amino-ethyl group at the 6N position, based on the kanamycin A skeleton (6N-2-amino-ethyl-KanaA-AHB-F-Gua) or on the kanamycin B skeleton (6N-2-amino-ethyl-KanaB-AHB-F-Gua), can be obtained following the same reaction sequence as described above for ABX5037, but starting with kanamycin A or kanamycin B, respectively.

B.4.6 Tobra-(AHB-Gua)-F-Gua(ABX5033)/Tobra-(AFB)-F-Gua(ABX5034)

Synthesis of Compound C2 was as described above for B.1.1 class 1 compounds.

And (c). To a solution of AHB-Gua (1.7g, 4.7mmol, 1.2 equivalents) in dimethylformamide (19mL) was added triethylamine (670. mu.L, 4.8mmol, 1.2 equivalents), and the mixture was stirred at room temperature for 10 min. A solution of tobramycin derivative C2(3.45g, 4mmol, 1 eq) in dimethylformamide (19mL) was added followed immediately by HATU (1.54g, 4mmol, 1 eq) and the reaction mixture was stirred at room temperature overnight. After evaporation of dimethylformamide in vacuo, water (60mL) was added to the residue and the mixture was shaken until a white solid formed without residual oil. The white solid was filtered off, washed with sufficient water (120mL) and dried in vacuo by co-evaporation of the remaining water with toluene (3 ×). The resulting product C23(4.14g, 3.43mmol, yield 86%) was isolated as a white solid. LC/MS (System 2, method A): t is tR(min) ═ 8.35; MS (m/z): found 1207.45(M + H)+) 1207.57(M + H) is calculated+)。

And (d). Compound C23(4g, 3.31mmol) was dissolved in pyridine (33mL), acetic anhydride (3.15mL, 33.1mmol, 10 equiv.) was added at room temperature and stirred for two days. The crude mixture was poured into heptane (100mL) and the suspension was filtered through a plug of silica gel. The product C24 was eluted with ethyl acetate (50-70%) in heptane (R) f0.41, 70% ethyl acetate in heptane) was used. After removal of volatiles in vacuo, product C24 was obtained as a white solid (2.05g, 1.45mmol, yield 44%). LC/MS (System 2, method A):tR(min) ═ 10.78; MS (m/z): found 1417.6(M + H)+) 1417.57(M + H) is calculated+)。

And (e) step (e). Tobramycin derivative C24(1.95g, 1.37mmol) was dissolved in dichloromethane (14mL) and triethylamine (382. mu.L, 2.74mmol, 2 equiv.), triethylamine trihydrofluoride salt (893. mu.L, 5.48mmol, 4 equiv.) and X-TalFluor-E (941mg, 4.11mmol, 2 equiv.) were added in the exact order. After stirring at room temperature overnight, the reaction mixture was cooled to 0 ℃ and saturated aqueous sodium bicarbonate solution (50mL) was carefully added. The crude mixture was warmed to room temperature with stirring and the aqueous and organic layers were separated. The aqueous solution was extracted with dichloromethane (3X 50mL) and all combined organic layers were dried over anhydrous sodium sulfate. After removal of the volatiles in vacuo, the residue obtained was taken up in ethyl acetate (60%) in heptane (R)f0.57 using 70% ethyl acetate in heptane) as eluent to give product C25 as a light yellow solid (1.02g, 0.72mmol, yield 52%). LC/MS (System 1, method A): t is t R(min) ═ 10.74; MS (m/z): found 1419.2(M + H)+) 1419.62(M + H) is calculated+)。

And (f). To a solution of aminoglycoside derivative C25(985.6mg, 0.69mmol) in methanol (6.9mL) was added sodium methoxide (373mg, 6.9mmol, 10 equivalents), and the reaction mixture was stirred at room temperature for two days. Amberlite CG50 (H) was added portionwise+Form) until a pH of 7 is reached. After removal of Amberlite by filtration, the remaining solution was concentrated to dryness to give the crude product C26(1g) as a white solid which was used in the next step without further purification. LC/MS (System 2, method A): t is tR(min) ═ 6.31; MS (m/z): 1013.4(M-Boc + H) was observed+) Calculate 1013.53(M-Boc + H)+)。

And (g). Crude product C26(1g) was dissolved in DMF (50mL) and triethylamine (1mL, 7.17mmol, ca. 4.5 equivalents) and reagent Q1(1.2g, 3.87mmol, ca. 2.4 equivalents) were added. The reaction mixture was stirred at room temperature for two days. After removing all volatiles, the crude mixture was taken up in 5% methanol in ethyl acetate (R)f0.23 in 100% EtOAc) was purified by silica gel column chromatography to give product C27 as a white solid(169mg, 0.13mmol, 19% yield over two steps). LC/MS (System 1, method A): t is tR(min) ═ 8.24; MS (m/z): found 1255.4(M + H)+) 1255.67(M + H) is calculated +)。

And (h). To a solution of compound C27(118.6mg, 0.094mmol) in 1, 4-dioxane (12mL) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (12mL) and the reaction mixture was stirred at room temperature overnight. Volatiles were removed in vacuo to give a white solid which was purified by recrystallization/precipitation from methanol/tetrahydrofuran to give the product ABX5033 as pentahydrochloride salt (836.99g/mol, 55mg, 0.066mmol, yield 70%) as a white solid. LC/MS (System 2, method B): t is tR(min) ═ 2.02; MS (m/z): found 655.2(M + H)+) 655.35(M + H) is calculated+)。

Pentahydrochloride synthesis of ABX5034 was as shown for ABX5033 and used (2S) -Boc-4-amino-2-fluoro-butyric acid (AFB-Boc) as the reagent in step (c).

ABX5034(796.94g/mol, pentahydrochloride salt): a white solid; LC/MS (System 1, method B): t is tR(min) ═ 1.77; MS (M/z) ═ 615.2(M + H) measured+) 615.33(M + H) is calculated+)。1H NMR(300MHz,D2O)δ=5.64(d,J=52Hz,1H),5.49(1H),5.18(ddd,J=3.3Hz,J=9.1Hz,J=48.2Hz,1H),5.16(1H),4.51–4.03(3H),4.03–3.62(10H),3.55–3.40(2H),3.34–3.16(3H),2.49–2.16(3H),2.16–1.98(1H),1.98–1.78(1H)。19F NMR(282MHz,D2O)δ=-191.37(ddd,2JHF=48.6Hz,3JHF=33.8Hz,3JHF=18.8Hz),-215.93(dt,2JHF=52.5Hz,3JHF=26.2Hz)。

The corresponding derivative carrying AHB-Gua or AFB at position 1N, based on the kanamycin A skeleton (KanaA- (1N-R)4) -F-Gua) or based on the kanamycin B skeleton (KanaB- (1N-R)4) -F-Gua), which can follow the same reaction sequence as ABX5033 and ABX5034 described aboveBut was obtained starting from kanamycin a or kanamycin B, respectively.

Synthesis of reagents AHB-Gua and AFB-Boc

To a suspension of (2S) -4-amino-2-hydroxybutyric Acid (AHB) (16.8mmol, 2g, 1 eq) in dimethylformamide (20mL) at room temperature was added Q1(4.72g, 15.2mmol, 0.9 eq) followed by triethylamine (25.7mL, 184mmol, 11 eq). After stirring at room temperature for 5 days, the volatiles were removed in vacuo and the residue was purified by column chromatography (9: 1; ethyl acetate/heptane) to give the desired product AHB-Gua as a pale yellow oil (3.82g, 10.58mmol, yield 63%). LC/MS (System 1, method D): t is tR(min) ═ 1.89; measured 362.4(M + H) MS (M/z)+) 362.19(M + H) is calculated+)。1H NMR(300MHz,CD3OD)δ=4.22(1H),3.57(2H),2.11(1H),1.91(1H),1.57(9H),1.52(9H)。

The synthesis of the reagent (2S) -Boc-4-amino-2-fluoro-butyric acid (AFB-Boc) was according to the published procedure (m.e. farkas, b.c. li, c.dose, p.b. dervan, Bioorg. & med.chem.lett.2009,19, 3919-.

AFB-Boc (221.11 g/mol): a colorless oil; LC/MS (System 1, method D): t is tR(min) ═ 0.55; MS (M/z) ═ found 244.4(M + Na +), calculated 244.10(M + Na +).1H NMR(300MHz,DMSO-d6)δ=13.27(1H),6.89(1H),4.96(ddd,2J=49.0Hz,3J=8.3Hz,3J=3.7Hz,1H),3.04(2H),2.08–1.65(2H),1.37(9H)。

B.4.7 Tobra-(AHCA)-F-Gua(ABX5035)

Synthesis of Compound C2 was as described above for B.1.1 class 1 compounds.

And (c). To a solution of tobramycin derivative C2(72mg, 0.084mmol, 1 eq) in DMF (0.5mL) at-40 deg.C was added dropwise a solution of DIPEA (60. mu.L, 0.34mmol, 4 eq) and OSu-AHCA (98mg, 0.21mmol, 2.5 eq) in DMF (1.5 mL). After 1 hour, the reaction mixture was warmed to room temperature and stirred for 16 hours. Volatiles were removed in vacuo and the viscous residue was suspended in water and stirred for several hours to obtain a fine powder. The suspension was filtered on a glass funnel and the remaining product was dried by co-evaporation with toluene (3 ×). The desired product C28 was obtained as a beige powder. (72.4mg, 0.06mmol, 71%) LC/MS (System 1, method A): t is t R(min) ═ 9.23; observed MS (M/z) ═ 1104.2(M-Boc + H)+) Calculate 1104.1(M-Boc + H)+)。

The final compound ABX5035 was obtained from intermediate C28 following the reaction sequence shown in the reaction scheme using the procedure described above for 6N-substituted class 4 compounds as described in b.4.5 (steps (d) to (h) and (k) in b.4.5).

Corresponding derivatives carrying AHCA at position 1N, based on the kanamycin A skeleton (KanaA- (1N-R)4) -F-Gua) or based on the kanamycin B skeleton (KanaB- (1N-R)4) -F-Gua) which can be obtained following the same reaction sequence as described above for ABX5035, but starting with kanamycin a or kanamycin B, respectively.

Synthesis of reagent OSu-AHCA

For the synthesis of OSu-AHCA reagent, the published procedure (p.dozzo, a.a.goldblum, j.b.agnen, m.sheringham Linsell, WO 2010/132768a9,2011) was used as follows (3 steps):

step 1. benzyl chloroformate (7.8mL, 55mmol, 1 equiv.) in dry dichloromethane (180mL) is added dropwise over 7 hours to a well stirred solution of 1, 2-diaminoethane (36mL, 539mmol, 10 equiv.) in dry dichloromethane (540mL) at-78 deg.C under a nitrogen atmosphere. The solution was slowly warmed to 0 ℃ and at the same temperatureStir overnight. The diurethane by-product was removed by filtration, and the filtrate was washed with water (3X 500mL) and Na 2SO4Dried and concentrated to give a mixture of benzyl (2-aminoethyl) carbamate and bis-Cbz-protected ethylenediamine. The mixture was dissolved in dichloromethane and washed with 1M hydrochloric acid solution. The aqueous phase was basified to pH 12 and extracted again with dichloromethane. Na for organic layer2SO4Drying and removal of volatiles in vacuo afforded the desired product as a white solid (8.7g, 44.8mmol, 82%). LC/MS (System 1, method D): t is tR(min) ═ 0.68; MS (M/z) ═ measured 195.4(M + H)+) 195.11(M + H) is calculated+)。

Step 2 benzyl-N- (2-aminoethyl) carbamate benzyl chloride salt (7.9g, 34.25mmol, 1 eq) with NaHCO3(656mL) of saturated aqueous solution 1M NaOH (219mL) was added and the reaction stirred vigorously. Dichloromethane (438mL) was added followed by benzoyl peroxide (25% water, 22.05g, 68.5mmol, 2 eq) and the reaction stirred at room temperature overnight. The organic layer was separated, washed with brine and Na2SO4Dry, filter and concentrate in vacuo until about 100mL of solution remains. The solution was poured into an excess of heptane (400mL) on a silica gel column and washed thoroughly with heptane. Using 35% ethyl acetate in heptane (R)f0.48 using 40% ethyl acetate in heptane) and the volatiles were removed in vacuo to give the product as a colourless oil (5.63g, 17.9mmol, 53%). LC/MS (System 1, method D): t is t R(min) ═ 1.98; MS (M/z) ═ actually measured 315.4(M + H)+) 315.13(M + H) is calculated+)。

Step 3. to a stirred solution of disuccinimidyl carbonate (461mg, 1.8mmol, 1.1 equiv.) in acetonitrile (25mL) is added dropwise a solution of benzyl-2- (benzoyloxyamino) ethylcarbamate (252mg, 1.6mmol, 1 equiv.) in acetonitrile (25 mL). The reaction mixture was stirred at 60 ℃ overnight, then all volatiles were removed in vacuo. The crude product was purified on a small pad of silica gel (1: 1; heptane/EtOAc) to give the desired product as a yellow oil (98mg, 0.21mmol, 13%). LC/MS (System 1, method D): t is tR(min) ═ 1.971; MS (m/z) — actually measured 456.3(M+H+) 456.14(M + H) is calculated+)。1H NMR(300MHz,CDCl3)δ=8.08(2H),7.71–7.61(1H),7.55–7.42(2H),7.41–7.28(5H),5.57(1H),5.09(2H),4.02(2H),3.52(2H),2.77(4H)。

B.4.83 '-epi-3' -O-alkyl-KanaB-AHB-F-Gua (ABX4012)

Synthesis of Compound B9a was as described above for B.2.2 of class 2 compounds.

And (h). LDA solution (2M in THF/heptane/ethylbenzene, 1.2 equiv.) was added dropwise to a pre-cooled solution of kanamycin B derivative B9a (1 equiv.) in tetrahydrofuran (20mL/mmol) at-35 deg.C. After stirring at this temperature for 30 minutes, the reaction was warmed to-20 ℃ and a solution of bromoacetonitrile (2 equivalents) in tetrahydrofuran (1.5mL/mmol bromoacetonitrile) was added dropwise. The reaction was stirred at-10 ℃ for 4 hours and then at room temperature overnight. Saturated aqueous ammonium chloride was added and the aqueous layer was extracted with DCM (3 ×). The combined organic layers were washed with Na 2SO4Drying and concentration gave the desired product B19 a.

And (i). At room temperature under an inert atmosphere (N)2) Next, a solution of aminoglycoside derivative B19a (1 equivalent) in methanolic ammonia (7N NH)320mL/mol) was added to Raney nickel (50% slurry in H2O, 1 mL/mmol). By H2The reaction vessel was rinsed and the reaction stirred at room temperature overnight. The reaction mixture was filtered through celite and concentrated in vacuo to give the desired crude product.

To a solution of the crude product (1 eq) in tetrahydrofuran (10mL/mmol) were added triethylamine (3 eq) and di-tert-butyl dicarbonate (1.5 eq) and the reaction mixture was stirred at room temperature for two days. After concentration in vacuo, the residue was shaken in water. The precipitate formed was filtered off, washed with water (2 ×) and dried by co-evaporation with toluene (3 ×) to give product B20 a.

And (j) step. To a solution of aminoglycoside derivative B20a (1 eq) in methanol (10mL/mmol) was added sodium methoxide (10 eq) and the reaction mixture was stirred at room temperature for two days. Amberlite CG50 (H) was added portionwise+Form) until a pH of 7 is reached. After removal of Amberlite by filtration, the remaining solution was concentrated to dryness to give the crude product B21 a.

And (k). Kanamycin B derivative B21a (1 eq) was dissolved in 1, 4-dioxane (70mL/mmol), triethylamine (10 eq) and N, N' -bis-Boc-1-guanidinopyrazole (Q1) (9 eq) were added and the reaction mixture was stirred at 45 ℃ for three days. After removal of all volatiles, the crude mixture was purified by column chromatography on silica gel using ethyl acetate in heptane to give product B22 a.

And (l). To a solution of compound B22a (1 eq) in 1, 4-dioxane (20mL/mmol) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (20mL/mmol) and the reaction mixture was stirred overnight. The volatiles were removed in vacuo to give the crude product, which was purified by recrystallization/precipitation from methanol/DCM to give the product ABX4012 as the hexahydrate salt.

The corresponding derivative carrying a residue on the epimerized hydroxyl group at the 3 ' position, based on the kanamycin A backbone (3 ' -epi-3 ' -O-alkyl KanaA-AHB-F-Gua), can be obtained according to the same reaction sequence as ABX4012 described above.

B.4.93 ' -methyl-3 ' -epi-3 ' -O-alkyl-kanamycin B-AHB-F-Gua (ABX4013)

Synthesis of Compound B16a was as described above for B.4.4 of class 4 compounds (wherein Compound B4a was obtained using the reaction sequence described for B.1.2 of class 1 compounds).

And (h). At-35 deg.C, mixing LDA solution(2M, 1.2 equiv.) was added dropwise to a previously cooled solution of kanamycin B derivative B16a (1 equiv.) in tetrahydrofuran (20 mL/mmol). After stirring at this temperature for 30 minutes, the reaction was warmed to-20 ℃ and a solution of bromoacetonitrile (2 equivalents) in tetrahydrofuran (1.5mL/mmol bromoacetonitrile) was added dropwise. The reaction was stirred at-10 ℃ for 4 hours and then at room temperature overnight. Saturated aqueous ammonium chloride was added and the aqueous layer was extracted with DCM (3 ×). The combined organic layers were washed with Na 2SO4Drying and concentration gave the desired product B23 a.

And (i). At room temperature under an inert atmosphere (N)2) Next, a methanolic ammonia solution (7N NH) of aminoglycoside derivative B23a (1 equivalent) was added320mL/mol) was added to Raney nickel (50% slurry in H2O, 1 mL/mmol). By H2The reaction vessel was rinsed and the reaction stirred at room temperature overnight. The reaction mixture was filtered through celite and concentrated in vacuo to give the desired crude product.

To a solution of the crude product (1 eq) in tetrahydrofuran (10mL/mmol) were added triethylamine (3 eq) and di-tert-butyl dicarbonate (1.5 eq) and the reaction mixture was stirred at room temperature for two days. After concentration in vacuo, the residue was shaken in water. The precipitate formed was filtered off, washed with water (2 ×) and dried by co-evaporation with toluene (3 ×) to give product B24 a.

And (j) step. To a solution of aminoglycoside derivative B24a (1 eq) in methanol (10mL/mmol) was added sodium methoxide (10 eq) and the reaction mixture was stirred at room temperature for two days. Amberlite CG50 (H) was added portionwise+Form) until a pH of 7 is reached. After removal of Amberlite by filtration, the remaining solution was concentrated to dryness to give the crude product B25 a.

And (k). Kanamycin B derivative B25a (1 eq) was dissolved in 1, 4-dioxane (70mL/mmol), triethylamine (10 eq) and N, N' -bis-Boc-1-guanidinopyrazole (Q1) (9 eq) were added and the reaction mixture was stirred at 45 ℃ for three days. After removal of all volatiles, the crude mixture was purified by silica gel column chromatography using ethyl acetate in heptane to give product B26 a.

And (l). To a solution of compound B26a (1 eq) in 1, 4-dioxane (20mL/mmol) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (20mL/mmol) and the reaction mixture was stirred overnight. The volatiles were removed in vacuo to give the crude product, which was purified by recrystallization/precipitation from methanol/DCM to give the product ABX4013 as the hexahydrate salt.

The corresponding derivative carrying a residue on the epimerized hydroxyl group at the 3 'position, based on the kanamycin A backbone (3' -methyl-3 '-epi-3' -O-alkyl KanaA-AHB-F-Gua), can be obtained according to the same reaction sequence as ABX4013 described above.

B.4.106N-2-hydroxy-ethyl-Tobra-AHB-F-Gua (ABX5045)

Synthesis of Compound C20 was as described above for B.4.5 of class 4 compounds.

And (i). Cesium (II) hydroxide monohydrate (1 eq.) and molecular sievesDissolved in DMF (10mL/mmol), the suspension was stirred at 35 ℃ for 10 min, then Compound C20(1 eq) was added. After stirring for 2 hours at 35 deg.C, (2-bromoethoxy) -tert-butyldimethylsilane (2 eq) was added and the reaction mixture was stirred for 24 hours at 35 deg.C. The molecular sieve was filtered off with a glass filter and the solution was concentrated. The crude product was purified by column chromatography on silica gel using a solution of methanol in ethyl acetate to elute product C23.

And (j) step. To a solution of compound C23(1 eq) in 1, 4-dioxane (20mL/mmol) was added a 4N solution of hydrogen chloride in 1, 4-dioxane (20mL/mmol) and the reaction mixture was stirred overnight. The volatiles were removed in vacuo to give the crude product, which was purified by recrystallization/precipitation from methanol/DCM to give the product ABX5045 as the hydrochloride salt.

ABX5045 as pentahydrochloride salt (839.00g/mol) as a white solid. LC/MS (System 2, method B): t is tR(min) ═ 2.13; MS (m/z): found 657.4(M + H)+) 657.36(M + H) is calculated+)。

The corresponding derivatives carrying a 2-hydroxy-ethyl group at the 6N position, based on the kanamycin A skeleton (6N-2-hydroxy-ethyl-KanaA-AHB-F-Gua) or on the kanamycin B skeleton (6N-2-hydroxy-ethyl-KanaB-AHB-F-Gua), can be obtained following the same reaction sequence as the above ABX5045, but starting with kanamycin A or kanamycin B, respectively.

B.5 guanylating reagents

From Q1 and with R5The general procedure for the synthesis of reagents Q2-Q6 and Q8-Q29 was started with alcohol (II):

example Q2:

n, N' -bis-Boc-1-guanidinopyrazole Q1(10.09g, 32.5mmol) and N- (2-hydroxyethyl) phthalimide (9.32g, 48.75mmol, 1.5 equivalents) were dissolved in tetrahydrofuran (140mL) and triphenylphosphine (12.79g, 48.75mmol, 1.5 equivalents) was added at room temperature. The reaction mixture was cooled to 0 ℃ and diisopropyl azodicarboxylate (15.21g, 14.81mL, 75.22mmol, 2.3 equiv.) was added dropwise over 30 minutes. The reaction mixture was warmed to room temperature and stirred for 2 days. After removal of volatiles, the entire crude residue was taken up in 30% strength ethyl acetate in heptane (R) f0.38 using 30% ethyl acetate in heptane) to afford product Q2 as a white solid (10.33g, 21.36mmol, 66% yield). LC/MS (System 1, method D): t is tR(min) ═ 2.19; measured 484.2(M + H) MS (M/z)+) 484.22(M + H) is calculated+)。1H NMR(300MHz,CDCl3)δ=7.95(1H),7.83(2H),7.69(2H),7.57(1H),6.37(1H),4.05(4H),1.40(9H),1.24(9H)。

From Q1 and with R5General procedure for the synthesis of reagent Q7 starting with bromide:

example Q7:

to a solution of potassium hydroxide (85% pure, 768mg, 11.63mmol, 1 eq) and N, N' -bis-Boc-1-guanidinopyrazole Q1(3.61g, 11.63mmol, 1 eq) in dry DMF (16mL) was added dropwise a solution of (2-bromoethoxy) (tert-butyl) dimethylsilane (2.5mL, 11.63mmol, 1 eq) in dry DMF (20 mL). After stirring at 30 ℃ for 10 days, the reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, dried over anhydrous sodium sulfate and concentrated. The residue was taken up to 10% ethyl acetate in heptane (R)f0.76 using 30% ethyl acetate in heptane) was purified by chromatography on silica gel to give product Q7 as a colorless oil (1.85g, 3.95mmol, yield 34%). LC/MS (System 1, method D): t is tR(min) ═ 2.91; MS (M/z) ═ found 469.4(M + H)+) 469.28(M + H) is calculated+)。1H NMR(300MHz,CDCl3)δ=7.97(1H),7.67(1H),6.37(1H),3.85(4H),1.49(9H),1.26(9H),0.83(9H),0.01(6H)。

Biological data and results

Coli is the most predominant gram-negative bacterium found, in particular, in nosocomial and nosocomial infections. The diversity of diseases caused by E.coli is due to the availability of specific virulence factors, either carried on transferable genetic elements (e.g., plasmids) or within distinct DNA fragments called pathogenicity islands, which are not present in non-pathogenic strains. As a result, there are many strains of E.coli, and some strains are more virulent than others. Such highly adapted bacterial species are often opportunistic when encountered in a hospital environment and are often identified particularly in urinary tract infections, abdominal wound infections and bacteremia. Thus, the provision of novel compounds exhibiting activity against E.coli, including E.coli strains having plasmids encoding different Aminoglycoside Modifying Enzymes (AME) which are known to be responsible for resistance to Aminoglycoside (AG) antibiotics, would constitute a valuable contribution to the art and therefore constitute a potential object of the present invention.

As described above, the compounds of the present invention exhibit excellent activity not only against a range of E.coli strains including wild-type E.coli and E.coli strains expressing a range of aminoglycoside modifying enzymes which are different and known to be responsible for resistance to aminoglycoside antibiotics, but also exhibit synergistic activity characteristics relative to analogous compounds bearing only 5-fluoro or 3 "-guanidino substituents. The particular combination of 5-fluoro and 3 "-guanidino substituents therefore leads to activity levels on the corresponding E.coli strains, which are completely unpredictable based on the known prior art. In this respect, to the best of the applicant's knowledge, there is no teaching in the prior art which would lead to the expectation of tolerating both 5-fluoro (i.e. 5-epi-5-fluoro) and 3 "-guanidino substituents in one molecule, not to mention such a wide range of aminoglycoside structures exhibited by the compounds of the present invention, from the standpoint of antibacterial activity. This combination of substituents not only results in compounds that continue to exhibit activity against the corresponding bacterial strain, but in fact a synergistic (not only additive) increase in efficacy is observed, which is entirely surprising and unexpected.

In this regard, reference is made to the data shown in table 1 below. Four amikacin-like (4, 6-disubstituted-2-DOS AG bearing an AHB group at position N1; 2-DOS ═ 2-deoxytriptolide, AHB ═ 4-amino-2-hydroxybutyrate) antibiotics with neither 5-fluoro nor 3 "-guanidino substituents thereon were selected as scaffolds and tested for the effect of the substituents. These compounds are specifically ABX4001 (1N-AHB-kanamycin B ═ amikacin B), ABX5004 (1N-AHB-tobramycin), ABX3002(1N-AHB-3 '-epi-kanamycin a ═ 3' -epi-amikacin a) and ABX4002(1N-AHB-3 '-epi-kanamycin B ═ 3' -epi-amikacin B), the structures of which are shown above and in fig. 1 below. Analogous compounds were synthesized as described above, each bearing 5-fluoro, 3 "-guanidino, and both 5-fluoro and 3" -guanidino substituents. The antibacterial activity of each compound was studied using the antibacterial susceptibility test using the method described in example section a.3.1 above, during which the Minimum Inhibitory Concentration (MIC) of each compound was determined for each bacterial strain described in table 1. These bacterial strains were further tested for a series of compounds with additional substitutions on the 3 "-guanidino group. Table 1 shows the MIC values of the compounds against American Type Culture Collection (ATCC) strains of escherichia coli. All compounds were tested against E.coli wild type strains (i.e., ATCC25922) which were also used for quality control, and E.coli strains containing plasmids encoding different AMEs.

As can be seen from table 1, whatever the escherichia coli strains tested (wild-type or strains expressing the various AME), the compounds displaying both 5-fluoro and 3 "-guanidino groups (i.e. ABX4006, ABX4004, ABX3003, ABX5006) not only displayed improved activity (totally unexpected in itself) on each strain with respect to the" parent compounds "not carrying said substituents (i.e. ABX4001, ABX4002, ABX3002, ABX5004), but also an improved level of activity when compared to the introduction of any of these substituents alone. Furthermore, most surprisingly, the synergistic effect on activity of the combination of each of these substituents in a single molecule exceeds that expected from the data shown in table 1, when considering the respective effect of analogous compounds having a 5-fluoro or 3 "-guanidino group on their activity relative to the respective" parent compound ". In fact, in several cases it was even observed that the introduction of the 3 "-guanidino substituent alone resulted in a loss of activity, however, when this substituent was introduced into the corresponding molecule with a 5-fluoro substituent, not only the expected loss of activity was not observed, but also a significant improvement in potency was observed. In this respect, with particular reference to escherichia coli expressing APH 3' (IIIa), compounds ABX4009 to ABX4006, ABX4005 to ABX4004, and ABX3004 to ABX 3003; for escherichia coli expressing APH 3' (Ia), ABX4005 vs ABX4004, and ABX3004 vs ABX 3003; data for E.coli expressing AAC (6 ') Ie-APH (2') Ia, ABX4005 vs ABX4004, and ABX3004 vs ABX 3003.

Thus, the data in table 1 further clearly show that compounds with 5-fluoro-and 3 "-guanidino groups, i.e. ABX4006, ABX4004, ABX3003 and ABX5006, overcome bacterial resistance mediated by all tested AME. Although E.coli strains expressing AME APH (3 ') IIIa are resistant to amikacin (according to EUCAST — "The European Committee on Antimicrobial Succinity testing. Breakwoint tables for interpretation of MICs and zone parameters, Version 8.1,2018.http:// www.eucast.org.", breakpoints of amikacin at MIC >16) and strains containing The drug-resistance-causing enzymes AAC (3) III, AAC (6 ') Ie-APH (2") Ia or AAC (3 ') IV are resistant to gentamicin (according to" Breakwoint tables for interpretation of MIC and zone parameters. Version 8.1,2018.http:// www.eucast.org. "of EUCAST), all strains tested for which have increased sensitivity to gentamicin are sensitive to gentamikacin. Furthermore, the compounds of the invention show in almost all cases a much higher efficacy (and never a lower efficacy) against all tested pathogens than the clinical administration of the antibiotic amikacin, whereas all compounds of the invention show significantly improved efficacy compared to gentamicin against e.coli expressing AAC (3) III, AAC (6 ') Ie-APH (2") Ia or AAC (3') IV. Even in the case where the potency of the compound against a given strain is equal to that of amikacin (or gentamicin) and thus is not improved relative to that of amikacin (or gentamicin), the compound of the present invention has a broader spectrum of antibacterial activity against the tested strains as compared to the clinical comparative example. Each compound with 5-fluoro-and 3 "-guanidino groups overcomes class-related bacterial resistance mediated by AME, assuming that the compounds of the present invention have similar or identical breakpoints to amikacin.

The data in table 1 are not only used to demonstrate the above-mentioned effects and advantages of the compounds of the invention having a free guanidino group at the 3 "-C position (also referred to as the 3" position), but also demonstrate the surprising activity of the hitherto unknown substituted (derivatized) guanidines at this position (compounds ABX5020, ABX5025, ABX5026, ABX5027, ABX5029, ABX5030, ABX5034, ABX5038, ABX5039, ABX5040, ABX5041, ABX5042, ABX5043, ABX5044, ABX5045, ABX5046, ABX5047, ABX5048, ABX5050 and ABX 5051). These additional structural changes are not only of interest as tools to address bacterial resistance or to increase the general potency or affinity of a drug for a target molecule, but are also important strategies to modify the physical (e.g., solubility, melting point, lipophilicity, hygroscopicity) and/or pharmacological (e.g., volume of distribution, plasma protein binding, toxicology, metabolic profile, CYP inhibition, hERG activity, etc.) properties of a molecule, which play an extremely important role in drug development. The data in table 1 not only show that this substitution on the guanidine group is tolerated in terms of activity/efficacy against all tested pathogens, but that compounds bearing these substituents continue to exhibit significantly improved activity relative to their respective parent compounds (ABX 5004). In many cases (e.g., for e.coli and e.coli APH (3') IIIa), these compounds continue to show synergistic levels of activity relative to those expected based on individual data for ABX5024(5-F) and ABX5005(3 "-guanidino). Furthermore, all compounds bearing this substituted guanidine moiety at the 3 "-C position showed significantly improved activity against all tested pathogens when compared to the clinically administered antibiotic amikacin. Furthermore, each of those derivatives has an improved or at least equal antibacterial activity when compared to gentamicin.

Furthermore, the results in Table 1 show that the simultaneous introduction of 5-fluoro and 3 "-guanidino substituents can also be used to restore the efficacy of AG derivatives that have lost some activity due to previous structural modifications. For example, epimerization of the hydroxyl group at the 3' -C position was introduced in 4, 6-disubstituted-2-DOS AG (Jaeger, M.Selective oxidation of carbohydrates, Ph.D. thesis, ISBN 978-90-367-. However, this modification resulted in a significant loss of antibacterial activity against wild-type strains of e. As shown in table 1, the derivatives ABX3002 and ABX4002 are direct analogs of amikacin and ABX4001(═ amikacin B), respectively, in which the hydroxyl group at the 3' -C position has been epimerized. MIC values in table 1 show that epimerization at the 3' -C position resulted in a 4-fold and between 8-and 16-fold reduction in the antibacterial activity of the amikacin and amikacin B (ABX4001) scaffolds, respectively, resulting in MIC values of 8 and 16-32 for the AG derivatives ABX4002 and ABX3002, respectively, against wild-type e. However, as shown in table 1, by introducing the 5-fluoro and 3 "-guanidino substituents of the invention (giving ABX3003 and ABX4004, respectively), the potency of the parent compounds Amikacin and ABX4001 was again achieved despite the presence of epimerization at the 3' -C position (table 1). Furthermore, the compounds of the invention not only showed restored antibacterial activity against all tested strains of escherichia coli, but were also found to overcome bacterial resistance mediated by all tested AME.

In the following table (table 1) and all other tables listed below, the values indicated using commas ("decimal point" standard expression, in particular in germany) are to be understood as corresponding to the same values indicated using decimal points, for example in the uk or in the us. For example, the value denoted 0,25 in the following table corresponds to a value denoted 0.25, for example in the uk or the us, and is equal to one quarter (1/4). Further, the values reported in the tables herein are generally expressed in ranges, such as 1-2, 4-8, 16-32, and the like. Such ranges are to be understood to include all values between the explicitly recited endpoints, but not inclusive of the recited endpoints. Thus, a range of 1-2 should be understood to mean a value between 1 and 2, but excluding 1 or 2, i.e. greater than 1 but less than 2. Thus, the reported activity value of 1 represents an improvement over the reported activity values of 1-2, and at value 1 there is no overlap in the activities. Likewise, reported activity values of 1-2 represent an improvement over reported activity value of 2, and at value of 2 there is no overlap in the activities. The same applies to all other activity ranges and values expressed in this way in the tables below herein. Finally, for the sake of clarity, it should be noted that the manner and form used herein to express ranges such as 1-2 or 4-8 may be generally expressed in the art using "slashes" rather than hyphens, namely 1/2 and 4/8, respectively. Each manner of expressing the ranges is standard in the art.

Table 1 antimicrobial susceptibility testing was performed with wild-type e.coli and e.coli strains expressing different AME.

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines as outlined in the materials and methods section above;2the strain ATCC25922, which is a strain,3ATCC25922 strain carrying a plasmid encoding AME.

To determine whether the compounds of the invention additionally show efficacy against other bacterial species, in particular against the above-mentioned selected bacteria of the ESKAPE family, the compounds were first tested for in vitro activity against four Gram-negative and one Gram-positive pathogens of the ESKAPE group, namely staphylococcus aureus (Gram +), klebsiella pneumoniae (Gram-), acinetobacter baumannii (Gram-), pseudomonas aeruginosa (Gram-), and enterobacter cloacae (Gram-). In addition, all compounds were tested against the gram-positive pathogen enterococcus faecalis. The results are shown in tables 2 and 3.

With respect to the data in Table 2, the latter bacterial strain (Enterobacter cloacae) is a clinical isolate, whereas the other bacterial species are "German Collection of microorganisms" ((German Collection of microorganisms) ", (Deutsche Sammlung von Mikroorganismen, DSM). This sensitivity test not only confirms that the combination of fluorination at the 5-C position (also called 5 position) and introduction of guanidine at the 3 "-C position (functionalized or not) surprisingly produces AG derivatives with excellent efficacy for all the bacterial strains tested, but also, totally unexpectedly, that the same synergistic effects as those observed for the escherichia coli strains are again observed (see table 1). In this respect it is noted that a comparison of the data, for example for enterococcus faecalis, Klebsiella pneumoniae and Enterobacter cloacae, wherein the introduction of 5-fluoro and 3 "-guanidino substituents is not only considered to result in additivity (based on the teaching of the known prior art, which is based on the teaching thereof In itself unexpected) and all compounds of the invention (ABX4006, ABX4004, ABX3003 and ABX5006) produce a synergistic effect in terms of activity relative to the activity observed by the introduction of any one of said substituents alone. This is more significant due to the fact that in many cases the introduction of only the 3 "-guanidino substituent results in a loss of activity (see, e.g., activity data for ABX4009, ABX4005 and ABX3004 for enterococcus faecalis, ABX4009 and ABX4005 for klebsiella pneumoniae, ABX4005 and ABX3004 for enterobacter cloacae, and ABX3004 for acinetobacter baumannii). Furthermore, derivatization of the 3 "-guanidino group (ABX5020, ABX5025, ABX5026, ABX5027, ABX5029, ABX5030, ABX5038, ABX5039, ABX5040, ABX5041, ABX5042, ABX5043, ABX5045, ABX5046, ABX5047, ABX5048, ABX5050, and ABX5051) was believed to produce compounds that exhibited superior levels of activity against all tested pathogens, exhibited improved activity in almost all cases over their respective parent compounds (ABX5004), and exhibited improved activity in all cases over the clinical comparative amikacin. In the case of klebsiella pneumoniae, acinetobacter baumannii and enterobacter cloacae, all compounds of the invention showed efficacy levels at least equal to, and in most cases much higher than, the clinically relevant aminoglycosides amikacin and gentamicin, respectively. The results show not only a completely surprising level of activity against a single ESKAPE group pathogen and enterococcus faecalis, but also a completely unexpected and significantly improved activity profile (cross-group activity) against the entire group of pathogens tested, relative to amikacin and gentamicin.

Table 2 antimicrobial susceptibility testing was performed with selected pathogens of the bacterial ESKAPE group and enterococcus faecalis.

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines as outlined in the materials and methods section above;2a clinical isolate.

In addition to the bacterial strains for which the data set out in table 2 above are directed, the compounds of the invention (based on scaffolds ABX4002, ABX3002 and ABX5004) were tested against laboratory ATCC strains of pseudomonas aeruginosa (ATCC 29212) and staphylococcus aureus (ATCC 29213) as well as ATCC strains of escherichia coli (ATCC BAA-1025 ═ BL 1). In contrast to the sensitivity tests described above (tables 1 and 2), MIC90 values were determined in this case instead of MIC50 values (these values represent the MIC of the bacterial growth inhibition level of 90% and 50%, respectively). The results are shown in Table 3.

All compounds of the invention (ABX4004, ABX3003, ABX5006, ABX5030, ABX5020, ABX5026) are believed to have overall superior antibacterial efficacy relative to their respective parent compounds (ABX4002, ABX3002, ABX 5004). Furthermore, upon introduction of both 5-fluoro and 3 "-guanidino substituents, an unexpected synergistic effect on the activity of these targets was again observed, relative to the single one. Again, this is considered to be more significant, since in the case of, for example, ABX3004, the introduction of a guanidino group only at the 3 "-C position would result in a reduction of the antibacterial activity. However, when this substituent was introduced into a compound carrying a 5-fluoro group (ABX3005), yielding the derivative ABX3003, the negative effect on the 3 "-guanidino activity was completely eliminated and, in the case of E.coli ATCC strains and Staphylococcus aureus strains, was even observed to result in an increase in activity. Thus, ABX3003 was observed to have 4/8-fold increased activity against all three bacterial strains compared to the parent compound ABX3002 lacking both structural changes.

Table 3 antimicrobial susceptibility testing was performed with selected pathogens and escherichia coli strains of the bacterial ESKAPE group.

1The method used for antimicrobial testing was the broth microdilution method performed according to the CLSI guidelines, as outlined in the materials and methods section above.

Further significant, based on the data shown in tables 2 and 3, was the observed improvement in efficacy against enterococcus faecalis and staphylococcus aureus. These species are gram-positive bacteria, which is particularly important because AG antibiotics are not currently used as monotherapies for infections caused by gram-positive bacteria. One reason for this is the low antibacterial activity of AG, as shown by amikacin (tables 2 and 3). In contrast, the compounds of the present invention show significantly increased activity levels against enterococcus faecalis and staphylococcus aureus, respectively, thus making them potential candidates for Aminoglycoside (AG) monotherapy against these gram-positive targets, or potentially more effective combination therapy when used in combination with other regulatory-approved antibiotics, in particular with (regulatory-approved) β -lactam antibiotics.

Antibacterial activity against selected wild-type bacterial strains and clinical isolates expressing various aminoglycoside modifying enzymes

Compounds ABX5006, ABX5026 and ABX5039 of the present invention were tested against two wild-type bacterial strains of the ESKAPE group, pseudomonas aeruginosa (PAO1) and acinetobacter baumannii (ATCC19606), as well as against a series of clinical isolates containing various AME, i.e. acinetobacter baumannii (195n (a) and 48F), escherichia coli (C1162 and C1181), morganrhizobium morgani (S49) and providencia stuartii (B8-1). While E.coli isolate C1162 contained a single AME, i.e., APH (3') Ia, the remaining isolates expressed multiple distinct subclasses of AME enzymes, i.e., AAC, ANT and APH. Table 4 shows the MIC values of the compounds of the invention against the bacterial isolates, compared to the aminoglycosides amikacin, apramycin and gentamycin. Notably, all compounds of the present invention showed significantly increased activity levels compared to all of the comparative aminoglycosides (table 4). It must also be emphasized that all the bacterial strains tested are sensitive to the compounds of the invention. In contrast, five strains were resistant to gentamicin (MIC >4) and three strains were resistant to amikacin according to the clinical Breakpoint defined in EUCAST (Breakwoint tables for interpretation of MICs and zone diameters. version 8.1,2018.http:// www.eucast.org). Since apramycin is still being developed for human use, the breakpoint for this aminoglycoside has not been defined. However, the MIC values for apramycin were two to sixteen times higher than for the compounds of the invention.

TABLE 4 antimicrobial susceptibility testing with selected wild-type pathogens and bacterial strains expressing various AMEs

1The method used for antimicrobial testing was the broth microdilution method performed according to the CLSI guidelines, as outlined in the materials and methods section above.

Antibacterial activity against ESKAPE group strains showing drug resistance

The provision of new compounds capable of treating bacterial strains exhibiting resistance to clinically relevant antibiotics, in particular aminoglycoside antibiotics, in particular amikacin, would constitute a major contribution to the art (and therefore constitute a potential object of the present invention). In view of this, a series of compounds of the invention were further evaluated to test their efficacy against selected resistant strains of representative gram negative and gram positive pathogens in the ESKAPE group (table 5). In particular, the ATCC strain of Staphylococcus aureus (ATCC BAA-1717), Klebsiella pneumoniae (ATCC BAA-1705, ATCC BAA-2524), Pseudomonas aeruginosa (ATCC BAA-2108) and Acinetobacter baumannii (ATCC BAA-1800) which have genes for various clinically relevant resistance mechanisms to antibiotics belonging to different antibiotic classes were used in this sensitivity study.

As shown in table 5, all compounds of the present invention have improved efficacy against both gram positive and gram negative pathogens compared to the clinical comparative example amikacin. All compounds of the present invention are more effective against amikacin resistant pathogens of Klebsiella pneumoniae (ATCC BAA-1705) and Acinetobacter baumannii (ATCC BAA-1800) and have up to 16-fold more extensive antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA) strain ATCC BAA-1717 than amikacin.

Clinical isolation of the gram-negative pathogens Pseudomonas aeruginosa, Acinetobacter baumannii and Klebsiella pneumoniae Antibacterial activity of strains and gram-positive pathogens enterococcus faecium and staphylococcus aureus.

Pseudomonas aeruginosa, acinetobacter baumannii and klebsiella pneumoniae are the main nosocomial gram-negative bacterial strains in the hospital environment that cause most nosocomial infections such as urinary tract infections, nosocomial pneumonia, ventilator-associated pneumonia, bloodstream infections, abdominal cavity infections and skin structure infections. Clinicians have very limited treatment options for patients infected with those pathogens.

Pseudomonas aeruginosa is the most commonly isolated pathogen in hospitals and has been found in ice making machines, pharmaceuticals, plasters, mouthwashes, sprayers, whirlpools, mattresses, sinks, potted plants, and many other locations and materials. It is a multifunctional pathogen with the ability to cause different types of infections. Mainly because the pathogen causes nosocomial pneumonia and neonates, and infects wounds after surgery. Due to the widespread use of broad-spectrum antibiotics, pseudomonas aeruginosa is the leading pathogen in hospitalized patients who are resistant to the majority of available antibiotics. Toxic drugs, such as polymyxins, are often the last treatment option, but are associated with significant side effects. In addition, most P.aeruginosa infections are endemic and sporadic. Severely contaminated fluids or medical equipment, and particularly virulent strains of pseudomonas aeruginosa, can cause epidemics.

Acinetobacter baumannii causes nosocomial bloodstream infections, pneumonia, soft tissue infections, urinary tract infections, abdominal infections, meningitis, and endocarditis. This pathogen is predominant in patients in intensive care units and is one of the major causes of ventilator-associated bacterial pneumonia. Its ability to rapidly develop resistance to major antibiotic types is responsible for the emergence of multidrug-resistant acinetobacter baumannii. In fact, this pathogen has become one of the critical gram-negative bacterial strains that needs to be handled in hospitals. Although carbapenems have been effective in the past against multi-drug resistant infections caused by acinetobacter baumannii, carbapenem-resistant acinetobacter baumannii (CRAB) species have emerged in the past decade. Currently, CRAB infections are usually treatable only with toxic antibiotics such as colistin and polymyxin B.

Klebsiella Pneumoniae (KP) is one of the most prominent gram-negative bacteria isolated from patients in intensive care units due to outbreaks associated with multiple drug resistant strains. In fact, klebsiella pneumoniae is the clinically most important member of klebsiella pneumoniae of the enterobacteriaceae family and causes various hospital-acquired infections, such as hospital-acquired pneumonia, urinary tract infections, wound/burn infections and bloodstream infections, particularly in patients with weakened immune systems. In addition, the opportunistic pathogens have developed resistance to third generation cephalosporin antibiotics. This resistance is mediated by the expression of extended spectrum beta-lactamases (ESBLs). In particular, infections caused by ceftazidime-resistant klebsiella pneumoniae have become a real challenge in hospital settings as they are associated with adverse clinical outcomes. For example, sepsis-associated mortality rates for those infected with third generation cephalosporin-resistant pathogens are more than 30% higher than with cephalosporin-sensitive strains of klebsiella pneumoniae. In 2006, 6-33% of the European clinical isolates were resistant to third-generation cephalosporins, and in 2013, 14-55% of the European clinical isolates were resistant to third-generation cephalosporins (European Center for Disease Control and preservation, Antimicrobial Surveillance Report,2008and 2013).

Staphylococcus aureus and enterococcus faecium are the major nosocomial gram-positive strains responsible for a variety of nosocomial infections, such as endocarditis, urinary tract infections, lung infections, bacteremia, and skin structure infections. Clinicians have only very limited treatment options for those patients infected with pathogens, particularly when infected with drug resistant isolates of vancomycin-resistant enterococcus faecium and methicillin-resistant staphylococcus aureus.

Staphylococcus aureus is a major bacterial human pathogen causing infections in community-and hospital-acquired settings. Multidrug resistant strain MRSA is a major cause of bacteremia, bone and joint infections, skin and soft tissue infections, endocarditis, gastroenteritis, meningitis, toxic shock syndrome and urinary tract infections. MRSA infections are common in hospitalized patients with open wounds, invasive devices such as catheters, and weakened immune systems. Hospital-acquired MRSA infections result in longer hospital stays and higher economic costs. Furthermore, MRSA, which is characterized by the continuous emergence of epidemic strains, is a dangerous clinical threat, causing infections with a continuing high morbidity and mortality. There is no doubt that new antimicrobial agents and adjunctive care aspects such as infectious disease counseling, echocardiography and source control are urgently needed for successful treatment of hospitalized patients with MRSA infections.

Since 1990 enterococcus faecium has become one of the major causes of multiple drug resistant enterococcus infections. About 50% of pathogenic isolates of enterococcus faecium are resistant to vancomycin, ampicillin and aminoglycosides. Currently, treatment of infections caused by enterococcus faecium pathogens is extremely challenging due to the increased mortality rate. VRE and ampicillin resistant enterococcus faecium cause 80% and 90.4% hospital acquired infections, respectively, associated with the use of devices such as ventilators and catheters. VRE has become a major nosocomial pathogen worldwide due to its colonization strategy, persistence in the environment, and genomic plasticity. In immunosuppressed patients, it causes a wide range of infections such as bacteremia, infectious endocarditis, abdominal and pelvic infections, urinary tract infections, central nervous system infections and skin structure infections. In the treatment of VRE infections in intensive care units, new agents, improved dosing regimens and combination therapies are urgently needed.

In view of the above, the provision of new compounds capable of treating infections caused by these bacterial strains (in particular those exhibiting resistance) would constitute a further major contribution to the art and therefore constitute the underlying object of the present invention. Thus, the study was extended to test the compounds of the invention against clinical isolates of Pseudomonas aeruginosa, Acinetobacter baumannii, Klebsiella pneumoniae, Staphylococcus aureus and enterococcus faecium.

To this end, a panel of clinical isolates of Pseudomonas Aeruginosa (PA), all from the university of crosstie, crotalaria hospital, was assembled and tested for sensitivity to a variety of antibiotic species, including aminoglycosides, carbapenems, third generation cephalosporins, macrolides, and quinolones, as outlined in the materials and methods section above. According to the clinical Breakpoint defined by EUCAST at 2018, 5, 16 (Breakwoint tables for interpretation of MICs and zone diameters, Version 8.1,2018.http:// www.eucast.org), the tested clinical isolates were multidrug resistant and insensitive to most of the available antibiotics. The group of six clinical isolates included five pathogens with moderate resistance and one with high levels of resistance to aminoglycosides. Pseudomonas aeruginosa was quality controlled using the corresponding non-resistant ATCC27853 strain. The results in table 6 show that the compounds of the invention (ABX5006, ABX5020, ABX5026 and ABX5039) have superior properties compared to other aminoglycoside antibiotics (amikacin and gentamicin) and representatives of other classes of antibiotics, namely third generation cephalosporins (ceftazidime) and carbapenems (meropenem).

In addition, as outlined in the materials and methods section above, a panel of Acinetobacter Baumannii (AB) clinical isolates, also from the university of crosstie, crohn's disease hospital, were also assembled and tested for sensitivity to a variety of antibiotic species, including aminoglycosides, carbapenems, third generation cephalosporins, macrolides, and quinolones. According to the clinical Breakpoint defined by EUCAST (Breakwoint tables for interpretation of MICs and zone diameters, Version 8.1,2018.http:// www.eucast.org; 5 months and 16 days 2018), the tested clinical isolates were multidrug resistant and insensitive to the majority of available antibiotics. All isolates tested were CRAB strains and therefore resistant to carbapenems, the last resort drugs. The panel of five clinical isolates included one strain with moderate resistance and four strains with high levels of resistance to aminoglycosides. Quality control was carried out using the corresponding non-drug-resistant A.baumannii ATCC17978 strain. The MIC values in table 7 demonstrate that the compounds of the invention (ABX5006, ABX5020, ABX5026 and ABX5039) have the highest efficacy and are effective against the entire pathogen group, including isolates that are resistant to the comparative compounds.

Furthermore, as outlined in the materials and methods section above, a group of clinical isolates of Klebsiella Pneumoniae (KP), all from the university of saxabriber hospital, crohn, were assembled and tested for sensitivity to a variety of antibiotic species, including aminoglycosides, carbapenems, third generation cephalosporins, macrolides, and quinolones. Pathogens are isolated from hospitalized patients with urinary tract infections, nosocomial pneumonia, bloodstream infections, and abdominal infections. According to the clinical Breakpoint defined by EUCAST (Breakwoint tables for interpretation of MICs and zone diameters, Version 8.1,2018.http:// www.eucast.org; 5 months and 16 days 2018), the tested clinical isolates were multidrug resistant and insensitive to the majority of available antibiotics. These bacterial isolates express carbapenemases of the OXA, NDM or VIM type and extended spectrum beta-lactamases (ESBLs). In addition, the selected group of seven clinical isolates included six strains with moderate resistance and one with high levels of resistance to aminoglycosides. Quality control was carried out using the corresponding non-resistant ATCC43816 Klebsiella pneumoniae strain. The results in table 8 show the antibacterial activity profile of selected compounds of the invention (ABX5006, ABX5020, ABX5026, ABX5039 and ABX 4006). It is noteworthy that all compounds of the invention not only show improved activity characteristics with respect to amikacin, but are also effective against multidrug-resistant klebsiella pneumoniae strains, which are resistant to the aminoglycoside amikacin of the last resort.

In addition, as outlined in the materials and methods section above, a panel of methicillin-resistant staphylococcus aureus (MRSA) clinical isolates, all from the university of crotalaria hospital, were assembled and tested for sensitivity to a variety of antibiotic species, including aminoglycosides, carbapenems, and third-generation cephalosporins. According to the clinical Breakpoint defined by EUCAST at 2018, 5, 16 (Breakwoint tables for interpretation of MICs and zone diameters, Version 8.1,2018.http:// www.eucast.org), the tested clinical isolates were multidrug resistant and insensitive to most carbapenems and third generation cephalosporins. The results in table 9 show that all three compounds ABX5006, ABX5026 and ABX5039 of the present invention have superior performance compared to other aminoglycoside antibiotics (amikacin and gentamicin) and representatives of other classes of antibiotics, third generation cephalosporins (ceftazidime) and carbapenems (meropenem).

In a further study, a panel of vancomycin-resistant enterococcus faecium (VRE) clinical isolates, all from the university of saxabriber hospital, crohn, were assembled and tested for sensitivity to a variety of antibiotic species, including aminoglycosides, carbapenems, and third generation cephalosporins, as outlined in the materials and methods section above. According to the clinical Breakpoint defined by EUCAST at 2018, 5, 16 (Breakwoint tables for interpretation of MICs and zone diameters, Version 8.1,2018.http:// www.eucast.org), the tested clinical isolates were multidrug resistant and insensitive to most aminoglycosides, carbapenems and third generation cephalosporins. Of particular note (see table 10) is that the entire group of nine clinical isolates had high levels of aminoglycoside resistance. The results in table 10 show that all three compounds ABX5006, ABX5026 and ABX5039 of the present invention have superior performance compared to other aminoglycoside antibiotics (amikacin and gentamicin) and representatives of other classes of antibiotics, third generation cephalosporins (ceftazidime) and carbapenems (meropenem).

Table 5 antibacterial activity of compounds of the invention and amikacin against selected resistant strains of the ESKAPE group.

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines, as outlined in the materials and methods section above; clinical breakpoints defined by EUCAST (The European Committee on Antimicrobial surgery testing. BreakPoint tables for interpretation of MICs and zone parameters. version 8.1,2018.http:// www.eucast.org.), resistant to amikacin; OXA-48 is a carbapenemase; KPC refers to Klebsiella pneumoniae carbapenemase; MDR refers to multiple drug resistance; MRSA refers to methicillin-resistant staphylococcus aureus.

Table 6 antibacterial activity of the compounds of the invention and clinical comparative examples against multiple drug-resistant pseudomonas aeruginosa clinical isolates.

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines, as outlined in the materials and methods section above;2ATCC27853 strain; clinical breakpoint defined according to EUCAST (The European Committee on Antimicrobial surgery testing. Breakwoint tables for interpretation of MICs and zone parameters. version 8.1,2018.http:// www.eucast.org.: for amikacin (MIC) >16) P-gentamicin (MIC)>4) P-ceftazidime (MIC)>8) Para Meropenem (MIC)>8) The comparative example had drug resistance); OXA-2 is a carbapenemase; MDR refers to multiple drug resistance.

Table 7 antibacterial activity of the compounds of the present invention and clinical comparative examples against multiple drug-resistant acinetobacter baumannii clinical isolates.

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines, as outlined in the materials and methods section above;2ATCC17978 strain; clinical breakpoint defined according to EUCAST (The European Committee on Antimicrobial surgery testing. Breakwoint tables for interpretation of MICs and zone parameters. version 8.1,2018.http:// www.eucast.org.: for amikacin (MIC)>16) P-gentamicin (MIC)>4) Para Meropenem (MIC)>8) The comparative example had drug resistance); CRAB refers to carbapenem-resistant acinetobacter baumannii.

Table 8 antibacterial activity of the compounds of the invention and clinical comparative examples against multiple drug resistant klebsiella pneumoniae clinical isolates.

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines, as outlined in the materials and methods section above;2ATCC 43816 strain; clinical breakpoint defined according to EUCAST (The European Committee on Antimicrobial surgery testing. Breakwoint tables for interpretation of MICs and zone parameters. version 8.1,2018.http:// www.eucast.org.: for amikacin (MIC) >16) P-gentamicin (MIC)>4) P-ceftazidime (MIC)>4) Para Meropenem (MIC)>8) The comparative example had drug resistance); VIM, NDM, OXA-48 are carbapenemases, ESBL refers to extended spectrum beta-lactamases; ColR refers to antiadherin.

Table 9 antibacterial activity of compounds of the invention and clinical comparative examples against clinical isolates of methicillin-resistant staphylococcus aureus (MRSA).

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines, as outlined in the materials and methods section above; comparative examples were resistant according to The clinical breakpoint defined in EUCAST (The European Committee on Antimicrobial Suadaptability testing. BreakPoint tables for interpretation of MICs and zone parameters. version 8.1,2018.http:// www.eucast.org).

Table 10 antibacterial activity of the compounds of the invention and clinical comparative examples against clinical isolates of vancomycin-resistant enterococcus faecium (VRE).

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines, as outlined in the materials and methods section above; comparative examples were resistant according to The clinical breakpoint defined in EUCAST (The European Committee on Antimicrobial Suadaptability testing. BreakPoint tables for interpretation of MICs and zone parameters. version 8.1,2018.http:// www.eucast.org).

To demonstrate the potential of the compounds of the invention for clinical use, ABX5006 was further tested against a clinical isolate from the medical center of grand university of gening, netherlands (UMCG). Eight E.coli AG-resistant strains (MIC > the breaking point of amikacin, tobramycin and gentamicin, as defined by EUCAST (BreakPoint tables for interpretation of MICs and zone diameters. version 8.1,2018.http:// www.eucast.org; 2018, 5/16 th), one pan-resistant Pseudomonas aeruginosa strain, six aminoglycoside-sensitive Pseudomonas aeruginosa strains, nine pan-resistant Klebsiella Pneumoniae Carbapenemase (KPC) producing bacterial isolates, and four aminoglycoside-sensitive Acinetobacter baumannii strains were selected and tested as outlined in the materials and methods section above. Pan-resistant bacteria are an emerging group of highly resistant gram-negative bacilli that cause infections associated with significant morbidity and mortality. Selected pan-resistant pathogens are isolated from patients with infections that are resistant or insensitive to all available antibiotics. In this experiment, the corresponding non-drug resistant ATCC strains of the corresponding bacterial species were used for quality control. The MIC values shown in ABX5006 in table 11 significantly demonstrate that all isolates, including the highly aminoglycoside resistant and pan-resistant bacteria tested, were sensitive to the compounds of the present invention.

Table 11 antibacterial activity of ABX5006 and ABX5020 against multiple drug resistant clinical isolates.

1The method for antimicrobial testing was broth microdilution performed according to CLSI guidelines, as outlined in the materials and methods section above;2ATCC25922,3ATCC27853 strain;4ATCC43816 strain;5ATCC17978 strain.

In vivo efficacy testing

Compounds of the invention were further tested in vivo, ABX5006, ABX5020, ABX5026 and ABX 5039. The in vivo efficacy of the compounds was evaluated using a mouse models of thigh infection with neutropenia as outlined above in the materials and methods section. Pathogen-free male CD-1 (outbred) mice (6 animals per group) and a non-resistant ATCC strain of Klebsiella pneumoniae (ATCC 43816) were used. The MIC 90(μ g/mL) value of ABX5006 to this ATCC strain was 0.25, while the MIC 90(μ g/mL) value of each of ABX5020, ABX5026 and ABX5039 was 1. In vivo efficacy studies were performed using meropenem (a broad spectrum antibiotic) as a reference compound (for quality control), and each drug was administered twice a day (BID) Subcutaneously (SC), one and nine hours after infection.

During the study, one dose of meropenem, i.e. 100mg/kg, and three different doses of each new aminoglycoside were tested. Compounds ABX5006 and ABX5020 were first tested using doses of 4, 16 or 64mg/kg (table 12). Two thighs were infected, and a total of 12 samples were obtained for determination of Colony Forming Units (CFU) per gram of thigh. However, the control group (vehicle) had a mortality rate of 50%, resulting in only 6 sample reads. The survival of the group treated with meropenem and the two compounds of the invention was 100%. All mice were overdosed with ketamine + xylazine 24 hours post-infection. Each thigh was homogenized in sterile PBS using an Ultraturax of IKA. After homogenization, serial dilutions of the homogenate were used for CFU determination. The CFU count for each thigh will be a single result.

The results of the in vivo efficacy studies are summarized in table 12. ABX5006 and ABX5020 showed excellent in vivo activity, reaching 7 logs already at the lowest total dose of 4mg/kg compared with the vehicle control group respectively10And>7log10significant CFU count reduction. At the highest tested dose, 64mg/kg, both compounds were achieved compared to the vehicle control group>7log10The CFU count of (a) is decreased. In fact, multiple samples of the group treated with ABX5006 and ABX5020 resulted in CFU counts below the limit of detection (LOD ═ 1.0 × 10)2CFU/mL), demonstrating high in vivo activity of both compounds. Also surprising is that compound ABX5020 has similar in vivo activity to ABX5006 when the same dose of each is applied. As described above, ABX5020 had 4 times lower antibacterial activity against klebsiella pneumoniae strain in vitro than ABX5006, which was also used for in vivo studies (table 12). Without being bound by any theory, it appears that R on the 3 "-guanidino group in ABX50205The residues have an effect on in vivo performance, compensating for the reduced in vitro activity compared to ABX 5006.

Table 12 results of in vivo neutropenic mouse thigh model infected with klebsiella pneumoniae (ATCC 43816).

Due to the high in vivo efficacy of ABX5006 and ABX5020, in subsequent studies, 4-fold lower doses of compounds ABX5026 and ABX5039, i.e. 0.25, 1 or 4mg/kg, were administered (table 13). Exactly the same protocol was used for this study as described above for ABX5006 and ABX 5020. The results of the in vivo efficacy studies are summarized in table 13. Remarkably, ABX5026 and ABX5039 showed excellent in vivo activity, respectively at a dose of 1mg/kg, compared with the vehicle control group >6log10And>4log10significant CFU count reduction. At the highest tested dose, i.e. 4mg/kg, both compounds reached compared to the vehicle control group>7log10The CFU count of (a) is decreased.

Table 13 results of in vivo neutropenic mouse thigh model infected with klebsiella pneumoniae (ATCC 43816).

The compounds of the invention ABX5006 and ABX020 were further tested against drug-resistant Klebsiella pneumoniae strain ATCC BAA-1705. The Klebsiella pneumoniae strain carrying KPC is resistant or insensitive to aminoglycoside antibiotics tobramycin and amikacin. The MIC values for this strain are shown in Table 5. Detailed experimental procedures are described in the experimental and methods section above. Pathogen-free male CD-1 (outbred) mice (6 animals per group) and amikacin as a reference compound were used in this study. Amikacin (clinical comparative, also used for quality control) and each compound of the invention were administered twice a day (BID) Subcutaneously (SC) one and nine hours after infection. Two different doses (16 or 64mg/kg) of each compound were studied (Table 10). Two thighs were infected, and a total of 12 samples were obtained for determination of CFU per gram of thigh. Due to poor clinical condition, two animals of the group treated with ABX5020 were euthanized prior to the second dose, yielding eight samples for CFU determination. Survival rates for all other animal groups were 100%. All mice were overdosed with ketamine + xylazine 24 hours post-infection. Each thigh was homogenized in sterile PBS using an Ultraturax of IKA. After homogenization, serial dilutions of the homogenate were used for CFU determination. The CFU count for each thigh will be a single result.

The results of the in vivo efficacy studies are summarized in table 14. Furthermore, ABX5006 and ABX5020 showed superior in vivo activity against drug-resistant infection compared to amikacin. Treatment with ABX5006 at doses of 16 and 64mg/kg (SC, BID) resulted in a statistically significant reduction (-2.8-3.2 log) in the number of CFUs in the thigh compared to the vehicle control group10Decrease). Furthermore, treatment with ABX5020 at 16 and 64mg/kg (SC, BID) doses resulted in a statistically significant reduction (-2.7-3.6 log) in the number of CFUs in the thigh compared to the vehicle control group10Decrease). In contrast, amikacin treatment did not significantly reduce CFU counts in the thighs when compared to vehicle control groups administered either dose.

When comparing the in vitro and in vivo activity of each compound of the present invention, the same effects as those observed in the case of klebsiella pneumoniae above were also observed in this study. Also, compound ABX5020 surprisingly showed similar in vivo activity as ABX5006 when the same dose was applied. As shown in table 5, ABX5020 had 4-fold lower in vitro antibacterial activity against the klebsiella pneumoniae strain tested in this in vivo study than ABX5006 (table 14). Without being bound by any theory, it appears that R on the 3 "-guanidino group in ABX5020 5The residues have an effect on in vivo performance, compensating for the reduced in vitro activity compared to ABX 5006.

TABLE 14 results of neutropenic mouse thigh model in vivo infected with amikacin resistant Klebsiella pneumoniae (ATCC BAA-1705).

To further evaluate the in vivo efficacy of the compounds of the invention, ABX5006 and ABX5026 were also tested in vivo using a murine model of urinary tract infection. Urinary Tract Infections (UTIs) are among the most common bacterial infections in humans. Each year, UTI infections caused by gram-negative pathogens alone, which are treated in approximately 7 million hospitals worldwide. Therefore, the E.coli infection model has been used to evaluate the efficacy of the compounds of the invention compared to gentamicin, a broad spectrum antibiotic. Of all aminoglycosides, gentamicin is the most commonly used aminoglycoside against gram-negative nosocomial infections. The detailed protocol of this study is outlined in the materials and methods section above. Pathogen-free female C3H/HeNRj (inbred) mice (8 animals per group) and non-resistant ATCC strains of e.coli (ATCC 700336) were used. MIC (μ g/mL) values of ABX5006 and ABX5026 for this ATCC strain were 0.25 and 1, respectively. The MIC (. mu.g/mL) value of gentamicin against the same E.coli was 1.

Comparative examples gentamicin and each of the compounds ABX5006 and ABX5020 of the present invention were tested at doses of 15 and 30mg/kg (table 15). Each drug was delivered once daily (QD) Subcutaneously (SC) 24 hours post-infection. Each mouse was infected under mild ketamine + xylazine anesthesia by transurethral administration of 100 μ L/mouse of bacterial suspension. Mice were starved for water 90 minutes prior to infection and one hour post infection. Colony Forming Units (CFU) were determined in the kidney 24 hours after treatment. Survival in the vehicle and treatment groups was 100%. All mice were overdosed with ketamine + xylazine 24 hours post-infection. Kidneys were homogenized in sterile PBS using Ultraturax, IKA. After homogenization, serial dilutions of the homogenate were used for CFU determination. The CFU count for each kidney pair will be a single result.

The results of the in vivo efficacy studies are summarized in table 15. Two were used in comparison with the vehicle control groupAn effective and significant reduction in CFU counts in the kidney was observed after subcutaneous treatment with ABX5006 and ABX5026 at doses, i.e. 30 and 15 mg/kg. ABX5006 and ABX5026 showed excellent in vivo activity reaching 1.53log at a dose of 30mg/kg, respectively, as compared to the vehicle control group 10And 1.72log10And reached 1.38log at 15mg/kg dose, respectively10And 1.58log10The CFU count of (a) is decreased. A significant reduction in CFU count in the kidney was observed in ABX5006 at the 30mg/kg dose compared to the gentamicin-treated group at the same dose. Notably, a significant reduction in CFU counts in the kidney was observed at both doses of ABX5026 compared to the gentamicin-treated group at the same dose. In fact, five of the eight samples of the group treated with ABX5026 resulted in CFU counts below the limit of detection (LOD ═ 2.0 × 10)2CFU/mL), demonstrating high in vivo efficacy. Of particular note is that the in vivo efficacy of ABX5026 was significantly higher than that of gentamicin, although ABX5026 had the same MIC value as the reference antibiotic for the e. Also surprising is that compound ABX5026 had slightly higher in vivo activity than ABX5006 when the same dose of each was applied. As described above, ABX5026 had 4-fold lower in vitro antibacterial activity against the enterobacter coli strain than ABX5006 (i.e., 1 versus 0.25), which was also used for in vivo studies (table 15). Without being bound by any theory, it appears that R on the 3 "-guanidino group in ABX5026 5The residues have an effect on in vivo performance that compensates for the reduced in vitro activity compared to ABX 5006. This result also confirmed the observations in the in vivo studies of ABX5006 and ABX5026 using a mouse thigh infection model (table 12).

Table 15 results of in vivo mouse model of e.coli (ATCC700336) urinary tract infection.

In vivo toxicity test

Compounds of the invention ABX5006, ABX5026 and ABX5039 were further tested for acute toxicity in vivo in rats.During this study, the Maximum Tolerated Dose (MTD) and the median Lethal Dose (LD) of all compounds in male cd (spraque dawley) rats were determined by using the Intravenous (IV) route50). Three animals were used per group and each group was treated once daily at a dose of 50, 75, 100, 150 or 200 mg/kg/day. Each compound was delivered intravenously by slow injection over 15 minutes in a dose volume of 5 mL/kg. Mortality was recorded twice daily during the study (4 days), i.e. in the morning and at the end of the working day. Animals were clinically examined before dosing, 30 minutes after dosing, one, four and eight more hours, and then twice daily during the study. Between 30 and 90 minutes after dosing on day 1 and on day 4, animals were subjected to comprehensive clinical examinations outside the cages, including complete external examinations, posture and movement observations and behavioral examinations. All surviving animals were euthanized at the end of day 4. Gross necropsy was performed on all animals and organ weights (adrenal, brain, heart, kidney, liver, spleen, epididymis, thymus, testis) were recorded for all animals per group and compared to those of the vehicle group. Table 16 shows the results of the in vivo acute toxicity study. Notably, the pathology did not result in macroscopic findings in the treatment groups. In addition, absolute and relative organ weights were in the same range as in the vehicle group (data not shown). ABX5026 and ABX5049 have the same MTD75, their LD 50Values have been determined to be above 75mg/kg (no mortality occurred), but below 100mg/kg (all animals died). In contrast, ABX5006 was prominently shown to have significantly (at least 2-fold) higher MTD and LD than ABX5026 and ABX503950Values (table 16). However, all compounds of the present invention have significantly lower acute toxicity compared to gentamicin. Previous studies (Robbins et al, 1971) reported the LD of gentamicin in rats50The value was 67 mg/kg. In contrast, ABX5006 has>LD of 200mg/kg50The value (only one of the three rats died) was approximately 3-fold higher than that reported for gentamicin. Furthermore, the compounds ABX5026 and ABX5039 of the invention have>LD 75mg/kg50I.e. with significantly lower acute toxicity compared to the reference antibiotic.

Table 16 results of acute toxicity studies in rats.

1 Robbins et al.,1971

Sequence listing

<110> personal company of Eitao and Leibiotics (AGILeBiotics B.V.)

<120> novel antibacterial 3' -derivatives of 4, 6-disubstituted 2, 5-dideoxytetracycline aminoglycoside antibiotics

<130> 192143wo

<150> EP18210470.3

<151> 2018-12-05

<160> 6

<170> PatentIn version 3.3

<210> 1

<211> 816

<212> DNA

<213> E.coli codon optimized AAC (3) -III Gene from Pseudomonas aeruginosa

<400> 1

atgaccgatc tgaacattcc gcatacccat gcgcatctgg tggatgcgtt tcaggcgctg 60

ggcattcgtg cgggccaggc gctgatgctg catgcgagcg tgaaagcggt gggcgcggtg 120

atgggcggcc cgaacgtgat tctgcaagcg ctgatggatg cgctgacccc ggatggcacc 180

ctgatgatgt atgcgggctg gcaggatatt ccggatttta ttgatagcct gccggatgcg 240

ctgaaagcgg tgtatctgga acagcatccg ccgtttgatc cggcgaccgc gcgtgcggtg 300

cgtgaaaaca gcgtgctggc ggaatttctg cgtacctggc cgtgcgtgca tcgtagcgcg 360

aacccggaag cgagcatggt ggcggtgggc cgtcaggcgg cgctgctgac cgcgaaccat 420

gcgctggatt atggctatgg cgtggaaagc ccgctggcga aactggtggc gattgaaggc 480

tatgtgctga tgctgggcgc gccgctggat accattaccc tgctgcatca tgcggaatat 540

ctggcgaaaa tgcgtcataa aaacgtggtg cgttatccgt gcccgattct gcgtgatggc 600

cgtaaagtgt gggtgaccgt ggaagattat gataccggcg atccgcatga tgattatagc 660

tttgaacaga ttgcgcgtga ttatgtggcg cagggcggcg gcacccgtgg caaagtgggc 720

gatgcggatg cgtatctgtt tgcggcgcag gatctgaccc gttttgcggt gcagtggctg 780

gaaagccgct ttggcgatag cgcgagctat ggctga 816

<210> 2

<211> 816

<212> DNA

<213> synthetic APH (3') -Ia Gene

<400> 2

atgagccata ttcaacggga aacgtcttgc tcgaggccgc gattaaattc caacatggat 60

gctgatttat atgggtataa atgggctcgc gataatgtcg ggcaatcagg tgcgacaatc 120

tatcgattgt atgggaagcc cgatgcgcca gagttgtttc tgaaacatgg caaaggtagc 180

gttgccaatg atgttacaga tgagatggtc agactaaact ggctgacgga atttatgcct 240

cttccgacca tcaagcattt tatccgtact cctgatgatg catggttact caccactgcg 300

atccccggga aaacagcatt ccaggtatta gaagaatatc ctgattcagg tgaaaatatt 360

gttgatgcgc tggcagtgtt cctgcgccgg ttgcattcga ttcctgtttg taattgtcct 420

tttaacagcg atcgcgtatt tcgtctcgct caggcgcaat cacgaatgaa taacggtttg 480

gttgatgcga gtgattttga tgacgagcgt aatggctggc ctgttgaaca agtctggaaa 540

gaaatgcata agcttttgcc attctcaccg gattcagtcg tcactcatgg tgatttctca 600

cttgataacc ttatttttga cgaggggaaa ttaataggtt gtattgatgt tggacgagtc 660

ggaatcgcag accgatacca ggatcttgcc atcctatgga actgcctcgg tgagttttct 720

ccttcattac agaaacggct ttttcaaaaa tatggtattg ataatcctga tatgaataaa 780

ttgcagtttc atttgatgct cgatgagttt ttctaa 816

<210> 3

<211> 795

<212> DNA

<213> APH (3') -IIIa Gene from Streptococcus faecalis

<400> 3

atggctaaaa tgagaatatc accggaattg aaaaaactga tcgaaaaata ccgctgcgta 60

aaagatacgg aaggaatgtc tcctgctaag gtatataagc tggtgggaga aaatgaaaac 120

ctatatttaa aaatgacgga cagccggtat aaagggacca cctatgatgt ggaacgggaa 180

aaggacatga tgctatggct ggaaggaaag ctgcctgttc caaaggtcct gcactttgaa 240

cggcatgatg gctggagcaa tctgctcatg agtgaggccg atggcgtcct ttgctcggaa 300

gagtatgaag atgaacaaag ccctgaaaag attatcgagc tgtatgcgga gtgcatcagg 360

ctctttcact ccatcgacat atcggattgt ccctatacga atagcttaga cagccgctta 420

gccgaattgg attacttact gaataacgat ctggccgatg tggattgcga aaactgggaa 480

gaagacactc catttaaaga tccgcgcgag ctgtatgatt ttttaaagac ggaaaagccc 540

gaagaggaac ttgtcttttc ccacggcgac ctgggagaca gcaacatctt tgtgaaagat 600

ggcaaagtaa gtggctttat tgatcttggg agaagcggca gggcggacaa gtggtatgac 660

attgccttct gcgtccggtc gatcagggag gatatcgggg aagaacagta tgtcgagcta 720

ttttttgact tactggggat caagcctgat tgggagaaaa taaaatatta tattttactg 780

gatgaattgt tttag 795

<210> 4

<211> 1440

<212> DNA

<213> synthetic AAC (6') Ie-APH (2' ') Ia genes from Staphylococcus aureus

<400> 4

atgaatatag ttgaaaatga aatatgtata agaactttaa tagatgatga ttttcctttg 60

atgttaaaat ggttaactga tgaaagagta ttagaatttt atggtggtag agataaaaaa 120

tatacattag aatcattaaa aaaacattat acagagcctt gggaagatga agtttttaga 180

gtaattattg aatataacaa tgttcctatt ggatatggac aaatatataa aatgtatgat 240

gagttatata ctgattatca ttatccaaaa actgatgaga tagtctatgg tatggatcaa 300

tttataggag agccaaatta ttggagtaaa ggaattggta caagatatat taaattgatt 360

tttgaatttt tgaaaaaaga aagaaatgct aatgcagtta ttttagaccc tcataaaaat 420

aatccaagag caataagggc ataccaaaaa tctggtttta gaattattga agatttgcca 480

gaacatgaat tacacgaggg caaaaaagaa gattgttatt taatggaata tagatatgat 540

gataatgcca caaatgttaa ggcaatgaaa tatttaattg agcattactt tgataatttc 600

aaagtagata gtattgaaat aatcggtagt ggttatgata gtgtggcata tttagttaat 660

aatgaataca tttttaaaac aaaatttagt actaataaga aaaaaggtta tgcaaaagaa 720

aaagcaatat ataatttttt aaatacaaat ttagaaacta atgtaaaaat tcctaatatt 780

gaatattcgt atattagtga tgaattatct atactaggtt ataaagaaat taaaggaact 840

tttttaacac cagaaattta ttctactatg tcagaagaag aacaaaattt gttaaaacga 900

gatattgcca gttttttaag acaaatgcac ggtttagatt atacagatat tagtgaatgt 960

actattgata ataaacaaaa tgtattagaa gagtatatat tgttgcgtga aactatttat 1020

aatgatttaa ctgatataga aaaagattat atagaaagtt ttatggaaag actaaatgca 1080

acaacagttt ttgagggtaa aaagtgttta tgccataatg attttagttg taatcatcta 1140

ttgttagatg gcaataatag attaactgga ataattgatt ttggagattc tggaattata 1200

gatgaatatt gtgattttat atacttactt gaagatagtg aagaagaaat aggaacaaat 1260

tttggagaag atatattaag aatgtatgga aatatagata ttgagaaagc aaaagaatat 1320

caagatatag ttgaagaata ttatcctatt gaaactattg tttatggaat taaaaatatt 1380

aaacaggaat ttatcgaaaa tggtagaaaa gaaatttata aaaggactta taaagattga 1440

<210> 5

<211> 777

<212> DNA

<213> synthetic AAC (3) -IV Gene

<400> 5

gtgcaatacg aatggcgaaa agccgagctc atcggtcagc ttctcaacct tggggttacc 60

cccggcggtg tgctgctggt ccacagctcc ttccgtagcg tccggcccct cgaagatggg 120

ccacttggac tgatcgaggc cctgcgtgct gcgctgggtc cgggagggac gctcgtcatg 180

ccctcgtggt caggtctgga cgacgagccg ttcgatcctg ccacgtcgcc cgttacaccg 240

gaccttggag ttgtctctga cacattctgg cgcctgccaa atgtaaagcg cagcgcccat 300

ccatttgcct ttgcggcagc ggggccacag gcagagcaga tcatctctga tccattgccc 360

ctgccacctc actcgcctgc aagcccggtc gcccgtgtcc atgaactcga tgggcaggta 420

cttctcctcg gcgtgggaca cgatgccaac acgacgctgc atcttgccga gttgatggca 480

aaggttccct atggggtgcc gagacactgc accattcttc aggatggcaa gttggtacgc 540

gtcgattatc tcgagaatga ccactgctgt gagcgctttg ccttggcgga caggtggctc 600

aaggagaaga gccttcagaa ggaaggtcca gtcggtcatg cctttgctcg gttgatccgc 660

tcccgcgaca ttgtggcgac agccctgggt caactgggcc gagatccgtt gatcttcctg 720

catccgccag aggcgggatg cgaagaatgc gatgccgctc gccagtcgat tggctga 777

<210> 6

<211> 633

<212> DNA

<213> synthetic AAC (6') Ib Gene

<400> 6

atgttacgca gcagcaacga tgttacgcag cagggcagtc gccctaaaac aaagttaggc 60

atcacaaagt acagcatcgt gaccaacagc accgattccg tcacactgcg cctcatgact 120

gagcatgacc ttgcgatgct ctatgagtgg ctaaatcgat ctcatatcgt cgagtggtgg 180

ggcggagaag aagcacgccc gacacttgct gacgtacagg aacagtactt gccaagcgtt 240

ttagcgcaag agtccgtcac tccatacatt gcaatgctga atggagagcc gattgggtat 300

gcccagtcgt acgttgctct tggaagcggg gacggatggt gggaagaaga aaccgatcca 360

ggagtacgcg gaatagacca gttactggcg aatgcatcac aactgggcaa aggcttggga 420

accaagctgg ttcgagctct ggttgagttg ctgttcaatg atcccgaggt caccaagatc 480

caaacggacc cgtcgccgag caacttgcga gcgatccgat gctacgagaa agcggggttt 540

gagaggcaag gtaccgtaac caccccagat ggtccagccg tgtacatggt tcaaacacgc 600

caggcattcg agcgaacacg cagtgatgcc taa 633

165页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:PPAR-δ激动剂在治疗线粒体肌病中的用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类