ANTIMICROBIALS AND METHODS OF MAKING AND USING SAME

The present disclosure relates generally to the field of antimicrobial compounds and to methods of making and using them. These compounds are useful for treating, preventing, reducing the risk of, and delaying the onset of microbial infections in humans and animals. In some embodiments, the present disclosure provides a compound of Formula (I): or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application Ser. No. 62/681,498, filed Jun. 6, 2018; 62/660,782, filed Apr. 20, 2018; 62/660,747, filed Apr. 20, 2018; 62/633,554, filed Feb. 21, 2018; 62/633,454, filed Feb. 21, 2018; 62/593,445, filed Dec. 1, 2017; 62/522,574, filed Jun. 20, 2017; and 62/521,835, filed Jun. 19, 2017, each of which is herein incorporated by reference in its entirety.

TECHNICAL FIELD

This invention relates to antimicrobial compounds, and more particularly to pyrrolo[2,3-d]pyrimidin-2-ones useful for treating, preventing and reducing risk of microbial infections.

BACKGROUND

Since the discovery of penicillin in the 1920s and streptomycin in the 1940s, many new compounds have been discovered or specifically designed for use as antibiotic agents. It was once thought that infectious diseases could be completely controlled or eradicated with the use of such therapeutic agents. However, such views have been challenged because strains of cells or microorganisms resistant to currently effective therapeutic agents continue to evolve. Almost every antibiotic agent developed for clinical use has ultimately encountered problems with the emergence of resistant bacteria.

For example, resistant strains of Gram-positive bacteria such as methicillin-resistant staphylococci, penicillin-resistant streptococci, and vancomycin-resistant enterococci have developed. Resistant bacteria can cause serious and even fatal results for infected patients. See, e.g., Lowry, F. D. “Antimicrobial Resistance: The Example of Staphylococcus aureus,” J. Clin. Invest., vol. 111, no. 9, pp. 1265-1273 (2003); and Gold, H. S. and Moellering, R. C., Jr., “Antimicrobial-Drug Resistance,” N. Engl. J. Med., vol. 335, pp. 1445-53 (1996).

The discovery and development of new antibacterial agents have been for decades a major focus of many pharmaceutical companies. Nonetheless, in more recent years there has been an exodus from this area of research and drug development resulting in very few new antibiotics entering the market. This lack of new antibiotics is particularly disturbing, especially at a time when bacterial resistance to current therapies is increasing both in the hospital and community settings.

One approach to developing new antimicrobial compounds is to design modulators, for example, inhibitors, of bacterial ribosome function. By modulating or inhibiting bacterial ribosome function, antimicrobial compounds could interfere with essential processes such as RNA translation and protein synthesis, thereby providing an antimicrobial effect. In fact, some antibiotic compounds such as erythromycin, clindamycin, and linezolid are known to bind to the ribosome.

SUMMARY

The present disclosure relates generally to the field of antimicrobial compounds and to methods of making and using them. These compounds and tautomers thereof are useful for treating, preventing, reducing the risk of, or delaying the onset of microbial infections in humans and animals. The present disclosure also provides pharmaceutically acceptable salts of these compounds and tautomers.

In some embodiments, the present application provides a compound of Formula (AA):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

J is selected from

X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and the phenyl is optionally substituted with one or more RX;

Z is selected from

R1 is selected from H, C1-3 alkyl, and C2-4 alkenyl;

R2 is selected from H and C1-3 alkyl;

R3 is selected from H and C1-3 alkyl;

or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;

or R2 and R3 together with the nitrogen and sulfur atom to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;

R4 is selected from H and C1-3 alkyl;

R5 is selected from H and C1-6 alkyl;

R6 is selected from H, C1-6 alkyl, C2-6 alkenyl, and C3-6 cycloalkyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, and 3-6 membered heterocyclyl;

R7 is selected from H and C1-6 alkyl;

or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;

    • or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with OH;

Q is selected from C1-2 alkylene or —C(O)—;

R21 is selected from H, C1-6 alkyl optionally substituted with 1-3 halo;

R8 is selected from H and halogen;

each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;

or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;

each Ra is independently selected from H and C1-6 alkyl;

each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, OC(NH)NH2, and aryl;

each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and

each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

Also provided herein is a compound of Formula (A):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;

Z is selected from

R1 is selected from H and C1-3 alkyl;

R2 is selected from H and C1-3 alkyl;

R3 is selected from H and C1-3 alkyl;

or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;

R4 is selected from H and C1-3 alkyl;

R5 is selected from H and C1-6 alkyl;

R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, and 3-6 membered heterocyclyl;

R7 is selected from H and C1-6 alkyl;

or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;

    • or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with OH;

Q is selected from C1-2 alkylene or —C(O)—;

R21 is selected from H, C1-6 alkyl optionally substituted with 1-3 halo;

R8 is selected from H and halogen;

each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;

or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;

each Ra is independently selected from H and C1-6 alkyl;

each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;

each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and

each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

Also provided herein is a compound of Formula (I):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and the phenyl is optionally substituted with one or more RX;
  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
  • R4 is selected from H and C1-3 alkyl;
  • R5 is selected from H and C1-6 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, and 3-6 membered heterocyclyl;
  • R7 is selected from H and C1-6 alkyl;
  • or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

  • wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
  • or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

  • R8 is selected from H and halogen;
  • each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
  • or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
  • each Ra is independently selected from H and C1-6 alkyl;
  • each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
  • each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
  • each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

Also provided herein is a compound of Formula (II):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
  • R4 is selected from H and C1-3 alkyl;
  • R5 is selected from H and C1-6 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, or C3-6 heterocyclyl;
  • R7 is selected from H and C1-6 alkyl;
  • or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

  • wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
  • or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

  • R8 is selected from H and halogen;
  • R9 is selected from H and halogen;
  • R10 is selected from H, halogen, C1-4 alkyl, C1-4 haloalkyl, and C3-6 cycloalkyl;
  • R11 is selected from C1-3 alkyl, C2-4 alkenyl, and C3-6 cycloalkyl, wherein the C1-3 alkyl is optionally substituted with one or more Rb;
  • R17 is selected from H, halogen, and C1-6 alkyl;
  • each Ra is independently selected from H and C1-6 alkyl;
  • each Rb is independently selected from ORc, —C(O)ORc, and —(O)aryl, wherein the aryl is optionally substituted with one or more Rd;
  • each Rc is independently selected from hydrogen, C1-3 alkyl, OH, O(C1-3 alkyl), NO2, —C(O)aryl, and aryl; and
  • each Rd is C1-3 alkyl.

Also provided herein is a compound of Formula (III):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
  • R4 is selected from H and C1-3 alkyl;
  • R5 is selected from H and C1-6 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, and —SC(NH)NH2;
  • R7 is selected from H and C1-6 alkyl;
  • or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

  • wherein the ring is optionally substituted on a carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
  • or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

  • R8 is selected from H and halogen;
  • R12 is selected from H, halogen, and C1-6 alkyl;
  • R13 is selected from H, halogen, C1-6 alkyl, ORc, N(Rc)2, wherein C1-6 alkyl is optionally substituted with one or more of ORc and aryl;
  • R14 is selected from H and aryl;
  • or R13 and R14 together with the carbon atoms to which they are attached form a 5- or 6-membered cycloalkyl ring;
  • R15 is selected from H, C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more Rb.
  • each Ra is independently selected from H and C1-6 alkyl;
  • each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
  • each Rc is independently selected from hydrogen, C1-6 alkyl, aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
  • each Rd is independently selected from OH, O(C1-3 alkyl), NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments provided herein is a pharmaceutical composition comprising a compound of Formula (AA), Formula (A), Formula (I), Formula (II), or Formula (III), or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, and a pharmaceutically acceptable carrier.

In some embodiments provided herein is a method of treating a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of Formula (AA), Formula (A), Formula (I), Formula (II), or Formula (III), or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition as provided herein.

In some embodiments provided herein is a method of preventing a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of Formula (AA), Formula (A), Formula (I), Formula (II), or Formula (III), or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition as provided herein.

In some embodiments provided herein is a method of reducing the risk of a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of Formula (AA), Formula (A), Formula (I), Formula (II), or Formula (III), or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition as provided herein.

In some embodiments provided herein is a method of delaying the onset of a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of Formula (AA), Formula (A), Formula (I), Formula (II), or Formula (III), or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition as provided herein.

In some embodiments provided herein is a method of treating a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of Formula (AA), Formula (A), Formula (I), Formula (II), or Formula (III), or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition as provided herein.

In some embodiments provided herein is a use of a compound of Formula (AA), Formula (A), Formula (I), Formula (II), or Formula (III), or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, or reducing a microbial infection in a subject.

In some embodiments provided herein is a compound of Formula (AA), Formula (A), Formula (I), Formula (II), or Formula (III), or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, for use in treating, preventing, or reducing a microbial infection in a subject.

In addition, the disclosure provides methods of synthesizing the foregoing compounds and tautomers thereof, and pharmaceutically acceptable salts of the compounds and tautomers. Following synthesis, an effective amount of one or more of the compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers can be formulated with a pharmaceutically acceptable carrier for administration to a human or animal for use as antimicrobial agents, particularly as antibacterial agents. In certain embodiments, the compounds of the present disclosure are useful for treating, preventing, reducing the risk of, or delaying the onset of microbial infections or for the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of microbial infections.

Accordingly, the compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers or their formulations can be administered, for example, via oral, parenteral, intravenous, otic, ophthalmic, nasal, or topical routes, to provide an effective amount of the compound or tautomer thereof, or pharmaceutically acceptable salt of the compound or tautomer to the human or animal.

The foregoing and other aspects and embodiments of the disclosure can be more fully understood by reference to the following detailed description and claims.

DETAILED DESCRIPTION

The present disclosure utilizes a structure based drug design approach for discovering and developing new antimicrobial agents. This approach starts with a high resolution X-ray crystal of a ribosome to design new classes of antimicrobial compounds having specific chemical structures, ribosome binding characteristics, and antimicrobial activity. This structure based drug discovery approach is described in the following publication: Franceschi, F. and Duffy, E. M., “Structure-based drug design meets the ribosome”, Biochemical Pharmacology, vol. 71, pp. 1016-1025 (2006).

Based on this structure based drug design approach, the present disclosure describes new chemical classes of antimicrobial compounds useful for treating bacterial infections in humans and animals. Without being limited by theories, these compounds are believed to inhibit bacterial ribosome function by binding to the ribosome. By taking advantage of these ribosome binding sites, the antimicrobial compounds of the present disclosure can provide better activity, especially against resistant strains of bacteria, than currently available antibiotic compounds.

The present disclosure therefore fills an important ongoing need for new antimicrobial agents, particularly for antimicrobial agents, having activity against resistant pathogenic bacterial organisms.

The present disclosure provides a family of compounds or tautomers thereof, that can be used as antimicrobial agents, more particularly as antibacterial agents.

The present disclosure also includes pharmaceutically acceptable salts of the compounds and tautomers.

The compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers disclosed herein can have asymmetric centers. Compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure containing an asymmetrically substituted atom can be isolated in optically active or racemic forms. Optically active forms of compounds can be prepared, for example, by resolution of racemic forms or by synthesis from optically active starting materials. Many geometric isomers of olefins, C═N double bonds, and the like can also be present in the compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers disclosed herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure are described and can be isolated as a mixture of isomers or as separate isomeric forms. All chiral, diastereomeric, racemic, and geometric isomeric forms of a structure are intended, unless specific stereochemistry or isomeric form is specifically indicated. All processes used to prepare compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure and intermediates made herein are considered to be part of the present disclosure. All tautomers of shown or described compounds are also considered to be part of the present disclosure. Furthermore, the disclosure also includes metabolites of the compounds disclosed herein.

The disclosure also provides for isotopically-labeled compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers, which are identical to those recited in formulae of the disclosure, but for the replacement of one or more atoms by an atom having an atomic mass or mass number different from the atomic mass or mass number most commonly found in nature. Examples of isotopes that can be incorporated into compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers of the disclosure include isotopes of hydrogen, carbon, nitrogen, and fluorine, such as 3H, 11C, 14C, and 18F.

The compounds of the present disclosure or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers that contain the aforementioned isotopes and/or isotopes of other atoms are within the scope of the present disclosure. Isotopically-labeled compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure, for example, those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritium, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred due to their ease of preparation and detectability. 11C and 18F isotopes are particularly useful in PET (positron emission tomography). PET is useful in brain imaging. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, i.e., increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labeled compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers having a formula of the disclosed herein can generally be prepared as described in the procedures, Schemes and/or in the Examples disclosed herein, by substituting a non-isotopically labeled reagent with a readily available isotopically labeled reagent. In one embodiment, the compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers disclosed herein are not isotopically labeled.

When any variable (e.g., R) occurs more than one time in any constituent or formulae of the disclosed herein, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with one or more R moieties, then R at each occurrence is selected independently from the definition of R. Also, combinations of substituents and/or variables are permissible, but only if such combinations result in stable compounds within a designated atom's normal valence.

When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent can be bonded to any atom on the ring. When a substituent is listed without indicating the atom via which such substituent is bonded to the rest of the compound of a given formula, then such substituent can be bonded via any atom in such substituent. Combinations of substituents and/or variables are permissible, but only if such combinations result in stable compounds.

In cases wherein compounds of the present disclosure, or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers thereof, contain nitrogen atoms, these, where appropriate, can be converted to N-oxides by treatment with an oxidizing agent (e.g., meta-chloroperoxybenzoic acid (mCPBA) and/or hydrogen peroxides). Thus, shown and claimed nitrogen atoms are considered to cover both the shown nitrogen and its N-oxide (N→O) derivative, as appropriate. In some embodiments, the present disclosure relates to N-oxides of the compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers disclosed herein.

One approach to developing improved anti-proliferative and anti-infective agents is to provide modulators (for example, inhibitors) of ribosome function.

Ribosomes are ribonucleoproteins, which are present in both prokaryotes and eukaryotes. Ribosomes are the cellular organelles responsible for protein synthesis. During gene expression, ribosomes translate the genetic information encoded in a messenger RNA into protein (Garrett et al. (2000) “The Ribosome: Structure, Function, Antibiotics and Cellular Interactions,” American Society for Microbiology, Washington, D.C.).

Ribosomes comprise two nonequivalent ribonucleoprotein subunits. The larger subunit (also known as the “large ribosomal subunit”) is about twice the size of the smaller subunit (also known as the “small ribosomal subunit”). The small ribosomal subunit binds messenger RNA (mRNA) and mediates the interactions between mRNA and transfer RNA (tRNA) anticodons on which the fidelity of translation depends. The large ribosomal subunit catalyzes peptide bond formation, i.e., the peptidyl-transferase reaction of protein synthesis, and includes, at least, three different tRNA binding sites known as the aminoacyl, peptidyl, and exit sites. The aminoacyl site or A-site accommodates the incoming aminoacyl-tRNA that is to contribute its amino acid to the growing peptide chain. Also, the A space of the A-site is important. The peptidyl site or P-site accommodates the peptidyl-tRNA complex, i.e., the tRNA with its amino acid that is part of the growing peptide chain. The exit or E-site accommodates the deacylated tRNA after it has donated its amino acid to the growing polypeptide chain.

1. DEFINITIONS

“Isomerism” means compounds that have identical molecular formulae but that differ in the nature or the sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers”. Stereoisomers that are not mirror images of one another are termed “diastereoisomers”, and stereoisomers that are non-superimposable mirror images are termed “enantiomers”, or sometimes optical isomers. A carbon atom bonded to four nonidentical substituents is termed a “chiral center”.

“Chiral isomer” means a compound with at least one chiral center. A compound with one chiral center has two enantiomeric forms of opposite chirality and may exist either as an individual enantiomer or as a mixture of enantiomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a “racemic mixture”. A compound that has more than one chiral center has 2n-1 enantiomeric pairs, where n is the number of chiral centers. Compounds with more than one chiral center may exist as either an individual diastereomer or as a mixture of diastereomers, termed a “diastereomeric mixture”. When one chiral center is present, a stereoisomer may be characterized by the absolute configuration (R or S) of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al, Angew. Chem. Inter. Edit. 1966, 5, 385; errata 511; Cahn et al., Angew. Chem. 1966, 78, 413; Cahn and Ingold, J. Chem. Soc. 1951 (London), 612; Cahn et al., Experientia 1956, 12, 81; Cahn, J., Chem. Educ. 1964, 41, 116).

“Geometric Isomers” means the diastereomers that owe their existence to hindered rotation about double bonds. These configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold-Prelog rules.

Further, the compounds discussed in this application include all atropic isomers thereof. “Atropic isomers” are a type of stereoisomer in which the atoms of two isomers are arranged differently in space. Atropic isomers owe their existence to a restricted rotation caused by hindrance of rotation of large groups about a central bond. Such atropic isomers typically exist as a mixture, however, as a result of recent advances in chromatography techniques, it has been possible to separate mixtures of two atropic isomers in select cases.

Some compounds of the present disclosure can exist in a tautomeric form which is also intended to be encompassed within the scope of the present disclosure. “Tautomers” refers to compounds whose structures differ markedly in the arrangement of atoms, but which exist in easy and rapid equilibrium. It is to be understood that compounds of the present disclosure may be depicted as different tautomers. It should also be understood that when compounds have tautomeric forms, all tautomeric forms are intended to be within the scope of the disclosure, and the naming of the compounds does not exclude any tautomeric form.

The compounds and pharmaceutically acceptable salts of the present disclosure can exist in one or more tautomeric forms, including the enol and imine form and the keto and enamine form, and geometric isomers and mixtures thereof. All such tautomeric forms are included within the scope of the present disclosure. Tautomers exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer predominates. Even though one tautomer may be described, the present disclosure includes all tautomers of the compounds disclosed herein.

A tautomer is one of two or more structural isomers that exist in equilibrium and are readily converted from one isomeric form to another. This reaction results in the formal migration of a hydrogen atom accompanied by a shift of adjacent conjugated double bonds. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers can be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. The concept of tautomers that are interconvertible by tautomerizations is called tautomerism.

Of the various types of tautomerism that are possible, two are commonly observed. In keto-enol tautomerism, a simultaneous shift of electrons and a hydrogen atom occurs. Ring-chain tautomerism, exhibited by glucose and other sugars, arises as a result of the aldehyde group (—CHO) in a sugar chain molecule reacting with one of the hydroxy groups (—OH) in the same molecule to give it a cyclic (ring-shaped) form.

Tautomerizations are catalyzed by: Base: 1. deprotonation; 2. formation of a delocalized anion (e.g., an enolate); 3. protonation at a different position of the anion; Acid: 1. protonation; 2. formation of a delocalized cation; 3. deprotonation at a different position adjacent to the cation.

Common tautomeric pairs include: ketone—enol, amide—nitrile, lactam—lactim, amide—imidic acid tautomerism in heterocyclic rings (e.g., in the nucleobases guanine, thymine, and cytosine), amine—enamine and enamine—enamine. Examples below are included for illustrative purposes, and the present disclosure is not limited to the examples:

The term “substituted,” as used herein, means that any one or more hydrogens on the designated atom, usually a carbon, oxygen, or nitrogen atom, is replaced with a selection from the indicated group, provided that the designated atom's normal valency is not exceeded, and that the substitution results in a stable compound. When a substituent is keto or oxo (i.e., ═O), then 2 hydrogens on the atom are replaced. Ring double bonds, as used herein, are double bonds that are formed between two adjacent ring atoms (e.g., C═C, C═N, N═N, etc.).

As used herein, “alkyl” is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. For example, C1-4 is intended to include C1, C2, C3, and C4. C1-6 alkyl is intended to include C1, C2, C3, C4, C5, and C6 alkyl groups and C1-8 is intended to include C1, C2, C3, C4, C5, C6, C7, and C8. Some examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, s-pentyl, n-hexyl, n-heptyl, and n-octyl.

As used herein, “alkenyl” is intended to include hydrocarbon chains of either straight or branched configuration and one or more unsaturated carbon-carbon bonds that can occur in any stable point along the chain, such as ethenyl and propenyl. For example, C2-6 alkenyl is intended to include C2, C3, C4, C5, and C6 alkenyl groups and C2-8 alkenyl is intended to include C2, C3, C4, C5, C6, C7, and C8.

As used herein, “alkylene” is intended to include moieties which are diradicals, i.e., having two points of attachment. A non-limiting example of such alkylene moiety that is a diradical is —CH2CH2—, i.e., a C2 alkyl group that is covalently bonded via each terminal carbon atom to the remainder of the molecule. The alkylene diradicals are also known as “alkylenyl” radicals. Alkylene groups can be saturated or unsaturated (e.g., containing —CH═CH— or —C≡C— subunits), at one or several positions. In some embodiments, alkylene groups include 1 to 9 carbon atoms (for example, 1 to 6 carbon atoms, 1 to 4 carbon atoms, or 1 to 2 carbon atoms). Some examples of alkylene groups include, but not limited to, methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, sec-butylene, tert-butylene, n-pentylene, iso-pentylene, sec-pentylene and neo-pentylene.

As used herein, “cycloalkyl” is intended to include saturated or unsaturated nonaromatic ring groups, such as cyclopropyl, cyclobutyl, or cyclopentyl. C3-8 cycloalkyl is intended to include C3, C4, C5, C6, C7, and C8 cycloalkyl groups. Cycloalkyls may include multiple spiro- or fused rings.

As used herein, the term “heterocycloalkyl” refers to a saturated or unsaturated nonaromatic 3-8 membered monocyclic, 7-12 membered bicyclic (fused, bridged, or spiro rings), or 11-14 membered tricyclic ring system (fused, bridged, or spiro rings) having one or more heteroatoms (such as O, N, S, or Se), unless specified otherwise. A heterocycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring. In some embodiments, the heterocycloalkyl is a monocyclic 4-6 membered heterocycloalkyl having 1 or 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur and having one or more oxidized ring members. In some embodiments, the heterocycloalkyl is a monocyclic or bicyclic 4-10 membered heterocycloalkyl having 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur and having one or more oxidized ring members. Examples of heterocycloalkyl groups include, but are not limited to, piperidinyl, piperazinyl, pyrrolidinyl, dioxanyl, tetrahydrofuranyl, isoindolinyl, indolinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, oxiranyl, azetidinyl, oxetanyl, thietanyl, 1,2,3,6-tetrahydropyridinyl, tetrahydropyranyl, dihydropyranyl, pyranyl, morpholinyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, 1,4-dioxa-8-azaspiro[4.5]decanyl and the like.

As used herein, “amine” or “amino” refers to unsubstituted —NH2 unless otherwise specified.

As used herein, “halo” or “halogen” refers to fluoro, chloro, bromo, and iodo substituents.

As used herein, “haloalkyl” is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with one or more halogen (for example —CvFwH2v−w+1 wherein v=1 to 3 and w=1 to (2v+1)). Examples of haloalkyl include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.

The term “haloalkoxy” as used herein refers to an alkoxy group, as defined herein, which is substituted one or more halogen. Examples of haloalkoxy groups include, but are not limited to, trifluoromethoxy, difluoromethoxy, pentafluoroethoxy, trichloromethoxy, etc.

As used herein, “alkoxyl” or “alkoxy” refers to an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. C1-6 alkoxy, is intended to include C1, C2, C3, C4, C5, and C6 alkoxy groups. C1-8 alkoxy, is intended to include C1, C2, C3, C4, C5, C6, C7, and C8 alkoxy groups. Examples of alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, s-butoxy, t-butoxy, n-pentoxy, s-pentoxy, n-heptoxy, and n-octoxy.

As used herein, “aryl” includes groups with aromaticity, including “conjugated,” or multicyclic systems with at least one aromatic ring and do not contain any heteroatom in the ring structure. Aryl may be monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings). The term “Cn-m aryl” refers to an aryl group having from n to m ring carbon atoms. In some embodiments, aryl groups have from 6 to 10 carbon atoms. In some embodiments, the aryl group is phenyl or naphthyl.

As used herein, the term “aromatic heterocycle”, “aromatic heterocyclic” or “heteroaryl” ring is intended to mean a stable 5, 6, 7, 8, 9, 10, 11, or 12-membered monocyclic or bicyclic aromatic ring which consists of carbon atoms and one or more heteroatoms, e.g., 1 or 1-2 or 1-3 or 1-4 or 1-5 or 1-6 heteroatoms, independently selected from nitrogen, oxygen, and sulfur. In the case of bicyclic aromatic heterocyclic or heterocycle or heteroaryl rings, only one of the two rings needs to be aromatic (e.g., 2,3-dihydroindole), though both can be (e.g., quinoline). The second ring can also be fused or bridged as defined above for heterocycles. The nitrogen atom can be substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, as defined). The nitrogen and sulfur heteroatoms can optionally be oxidized (i.e., N→O and S(O)p, wherein p=1 or 2). In certain compounds, the total number of S and O atoms in the aromatic heterocycle is not more than 1.

Examples of aromatic heterocycles, aromatic heterocyclics or heteroaryls include, but are not limited to, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, benzooxadiazoly, carbazolyl, 4aH-carbazolyl, carbolinyl, cinnolinyl, furazanyl, imidazolyl, imidazolonyl, 1H-indazolyl, indolizinyl, indolyl, 3H-indolyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylbenztriazolyl, methylfuranyl, methylimidazolyl, methylthiazolyl, naphthyridinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyridinonyl, pyridyl, pyrimidinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, triazolopyrimidinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, and 1,3,4-triazolyl. The term “hydroxyalkyl” means an alkyl group as defined above, where the alkyl group is substituted with one or more OH groups. Examples of hydroxyalkyl groups include HO—CH2—, HO—CH2—CH2— and CH3—CH(OH)—.

The term “cyano” as used herein means a substituent having a carbon atom joined to a nitrogen atom by a triple bond, i.e., C≡∂N.

As used herein, “oxo” is means a “═O” group.

As used herein, the phrase “pharmaceutically acceptable” refers to those compounds or tautomers thereof, or salts thereof, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

As used herein, “pharmaceutically acceptable salts” refer to derivatives of the disclosed compounds or tautomers thereof, wherein the parent compound or a tautomer thereof, is modified by making of the acid or base salts thereof of the parent compound or a tautomer thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound, or a tautomer thereof, formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include, but are not limited to, those derived from inorganic and organic acids selected from 2-acetoxybenzoic, 2-hydroxyethane sulfonic, acetic, ascorbic, benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edetic, ethane disulfonic, ethane sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, glycollyarsanilic, hexylresorcinic, hydrabamic, hydrobromic, hydrochloric, hydroiodide, hydroxymaleic, hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic, malic, mandelic, methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic, phenylacetic, phosphoric, polygalacturonic, propionic, salicylic, stearic, subacetic, succinic, sulfamic, sulfanilic, sulfuric, tannic, tartaric, and toluene sulfonic.

The pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound or a tautomer thereof, that contains a basic or acidic moiety by conventional chemical methods. Generally, such pharmaceutically acceptable salts can be prepared by reacting the free acid or base forms of these compounds or tautomers thereof with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, Pa., USA, p. 1445 (1990).

As used herein, “stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.

As used herein, the term “treating” means to provide a therapeutic intervention to cure or ameliorate an infection. In some embodiments, “treating” refers to administering a compound or pharmaceutical composition as provided herein for therapeutic purposes. The term “therapeutic treatment” refers to administering treatment to a patient already suffering from a disease thus causing a therapeutically beneficial effect, such as ameliorating existing symptoms, ameliorating the underlying metabolic causes of symptoms, postponing or preventing the further development of a disorder, and/or reducing the severity of symptoms that will or are expected to develop.

As used herein, the term “preventing”, as used herein means, to completely or almost completely stop an infection from occurring, for example when the patient or subject is predisposed to an infection or at risk of contracting an infection. Preventing can also include inhibiting, i.e., arresting the development, of an infection.

As used herein, the term “reducing the risk of”, as used herein, means to lower the likelihood or probability of an infection occurring, for example when the patient or subject is predisposed to an infection or at risk of contracting an infection.

As used herein, “unsaturated” refers to compounds having at least one degree of unsaturation (e.g., at least one multiple bond) and includes partially and fully unsaturated compounds.

As used herein, the term “effective amount” or “therapeutically effective amount” refers to an amount of a compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer (including combinations of compounds and/or tautomers thereof, and/or pharmaceutically acceptable salts of the compound or tautomer) of the present disclosure that is sufficient to elicit biological activity when administered alone or in combination as an antimicrobial agent. For example, an effective amount refers to an amount of the compound or tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer that is present in a composition, a formulation or on a medical device given to a recipient patient or subject sufficient to elicit biological activity, for example, anti-infective activity, such as e.g., anti-microbial activity, anti-bacterial activity, anti-fungal activity, anti-viral activity, or anti-parasitic activity.

The term “prophylactically effective amount” means an amount of a compound or a tautomer of the compound, or a pharmaceutically acceptable salt of the compound or tautomer (including combinations of compounds and/or tautomers thereof, and/or pharmaceutically acceptable salts thereof), of the present disclosure that is sufficient to prevent or reduce the risk of an infection when administered alone or in combination as an antimicrobial agent. For example, a prophylactically effective amount refers to an amount of the compound or tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer that is present in a composition, a formulation, or on a medical device given to a recipient patient or subject sufficient to prevent or reduce the risk of an infection due to a surgical procedure or an invasive medical procedure.

As used herein, the terms “expose,” “exposure,” or “exposed” means that a subject comes in contact in any way with a bacterium or any component thereof (e.g., bacterial cell wall, bacterial cell membrane, a bacterial nucleic acid, a bacterial polynucleotide, a bacterial protein, a bacterial polypeptide, a bacterial spore, and a bacterial toxin). For example, a subject can be exposed to a bacterium or any component thereof by ingesting, inhaling, or touching anything which contains the bacterium or any component thereof. Bacterium, as well as components of a bacterium (e.g., bacterial cell wall, bacterial cell membrane, a bacterial nucleic acid, a bacterial polynucleotide, a bacterial protein, a bacterial polypeptide, a bacterial spore, and a bacterial toxin), can cause an infection or symptoms of an infection in a subject. An example of a bacterial component that can cause an infection is a bacterial spore.

“Suspected exposure,” as used herein, means that there is certain possibility, although it is not known, that a subject has been exposed to a microorganism, for example, a bacterium, and thus is at the risk of a microbial (bacterial) infection, such as a bacterial infection. In some embodiments, “suspected exposure” means that there is greater than a 50% possibility that a subject has been exposed to a microorganism, for example, a bacterium.

As used herein, a “symptom” of a microbial infection, for example, a bacterial infection, can be any indication that the subject exposed or suspected of being exposed to the bacterium is not normal, well, or comfortable, regardless of the subject's subjective perception or feeling. “Symptom” includes, but is not limited to, headache, stomachache, abdominal cramps, abdominal pain, muscle pain, fever, diarrhea, vomiting, coughing, weakness, tiredness, soreness, rash or bumps on skin, wounds in any parts of the body (skin, head, eye, ear, nose, mouth, torso, limbs, arm, hand, leg, foot, etc.), and an abnormality in any tissue or organ (skin, bone, blood, lymph, intestine, stomach, pancreas, brain, heart, lung, liver, spleen, kidney, bladder, ovary, etc.).

A bacterium is “easily produced or disseminated” if the bacterium can be produced or disseminated by routine methods, processes, or techniques and with common materials, reagents, and equipment available in the art, or by methods, processes, or techniques and with materials, reagents, and equipment which are accessible to and can be operated or used by a lay person having little or no training in the art.

The term “moderate morbidity” refers to morbidity of no less than 10%, no less than 15%, no less than 20%, no less than 25%, no less than 30%, no less than 35%, no less than 40%, or no less than 45%. The term “high morbidity” refers to morbidity of no less than 50%, no less than 55%, no less than 60%, no less than 65%, no less than 70%, no less than 75%, no less than 80%, no less than 85%, no less than 90%, or no less than 95%.

The term “moderate mortality” refers to mortality of no less than 10%, no less than 15%, no less than 20%, no less than 25%, no less than 30%, no less than 35%, no less than 40%, or no less than 45%. The term “high mortality” refers to mortality of no less than 50%, no less than 55%, no less than 60%, no less than 65%, no less than 70%, no less than 75%, no less than 80%, no less than 85%, no less than 90%, or no less than 95%.

The terms “resistance” or “resistant” refer to the antibiotic/organism standards as defined by the Clinical and Laboratories Standards Institute (CLSI) and/or the Food and Drug Administration (FDA).

As used herewith, the terms “multi-drug resistance,” “multi-drug resistant,” or “MDR” refer to acquired non-susceptibility to at least two antimicrobial agents, e.g., resistance to one agent in three or more antimicrobial categories. The terms “extremely-drug resistant,” “extensive drug resistance,” or “XDR,” as used herein, refer to acquired non-susceptibility to at least one agent in all but two or fewer antimicrobial categories. For example, bacterial isolates remain susceptible to only one or two categories. Accordingly, an XDR bacterial isolate is always an MDR bacterial isolate, but an MDR bacterial isolate is not necessarily an XDR bacterial isolate. For example, an XDR microorganism is a Pseudomonas aeruginosa isolate that is susceptible to only one or two antimicrobial categories, such as a Pseudomonas aeruginosa isolate that is only susceptible to polymyxins (for example, colistin) or only susceptible to a pyrrolocytosine compound described herein. See, for example, Magiorakos et al., Clin. Microbial Infect. 2012; 18: 268-281, the content of which is hereby incorporated by reference in its entirety.

The term “subject” includes animals which either have or are susceptible or are suspected to have acquired a microbial infection (e.g., a bacterial infection). Examples of subjects include animals such as farm animals (e.g., cows, pigs, horses, goats, rabbits, sheep, chickens, etc.), lab animals (mice, rats, monkeys, chimpanzees, etc.), pets (e.g., dogs, cats, ferrets, hamsters, etc.), birds (e.g., chickens, turkeys, ducks, geese, crows, ravens, sparrows, etc.), primates (e.g., monkeys, gorillas, chimpanzees, bonobos, and humans), and other animals (e.g., squirrels, raccoons, mice, rats, etc.). In some embodiments, the subject is a mouse or rat. In yet another embodiment, the subject is a cow, a pig, or a chicken. In another embodiment, the subject is a human.

As used herein, the term ESBL is extended spectrum beta-lactamase. The term KPC is Klebsiella pneumoniae carbapenemase.

As used herein, the term acute bacterial skin and skin structure infection (ABSSSI) encompasses complicated skin and skin structure infections (cSSSI) and complication skin and soft tissue infections (cSSTI), which have been used interchangeably. The terms uncomplicated skin and skin structure infections (uCSSSI) and uncomplicated skin and soft tissue infections (uCSSTI) have been used interchangeably.

As used herein, the term “spp.” is the abbreviation for species.

As used herein, the term “formulae of the disclosure” or “formulae disclosed herein” includes one or more of the Formulae: (AA), (A), (I), (II), and (III).

As used herein, the term “compound of the disclosure” or “compound disclosed herein” includes one or more compounds of the formulae of the disclosure or a compound explicitly disclosed herein.

The term “about,” when used in conjunction with a numerical range, modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 10%. Thus, “about 10” means 9 to 11.

All percentages and ratios used herein, unless otherwise indicated, are by weight.

Throughout the description, where compositions are described as having, including, or comprising specific components, or where processes are described as having, including, or comprising specific process steps, it is contemplated that compositions of the present disclosure also consist essentially of, or consist of, the recited components, and that the processes of the present disclosure also consist essentially of, or consist of, the recited processing steps. Further, it should be understood that the order of steps or order for performing certain actions are immaterial so long as the invention remains operable. Moreover, two or more steps or actions can be conducted simultaneously.

2. COMPOUNDS OF THE DISCLOSURE

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

J is selected from

X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and the phenyl is optionally substituted with one or more RX;

Z is selected from

R1 is selected from H, C1-3 alkyl, and C2-4 alkenyl;

R2 is selected from H and C1-3 alkyl;

R3 is selected from H and C1-3 alkyl;

or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;

or R1 and R3 together with the nitrogen and sulfur atom to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;

R4 is selected from H and C1-3 alkyl;

R5 is selected from H and C1-6 alkyl;

R6 is selected from H, C1-6 alkyl, C2-6 alkenyl, and C3-6 cycloalkyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, and 3-6 membered heterocyclyl;

R7 is selected from H and C1-6 alkyl;

or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas

wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;

or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with OH;

Q is selected from C1-2 alkylene or —C(O)—;

R21 is selected from H, C1-6 alkyl optionally substituted with 1-3 halo;

R8 is selected from H and halogen;

each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;

or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;

each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, OC(NH)NH2, and aryl;

each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and

each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments of Formula (AA), J is

In some embodiments of Formula (AA), X is phenyl optionally substituted with one or more RX. For example, X is phenyl substituted with one or more halogens. In some embodiments, X is phenyl substituted with two halogens. For example, X is phenyl substituted with fluoro and chloro.

In some embodiments of Formula (AA), Z is

In some embodiments of Formula (AA), R1 is selected from H, C1-3 alkyl, and C2-4 alkenyl; R2 is selected from H and C1-3 alkyl; R3 is selected from H and C1-3 alkyl; or R1 and R3 together with the nitrogen and sulfur atom to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring.

In some embodiments of Formula (AA), R4 is H.

In some embodiments of Formula (AA), R5 is H.

In some embodiments of Formula (AA), R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having the formula:

In some embodiments of Formula (AA), R8 is selected from H and halogen. For example, R8 is H.

In some embodiments of Formula (AA), each RX is independently selected from halogen, C1-6 alkyl, and C1-4 haloalkyl. For example, each RX is halogen (e.g., fluoro or chloro).

In some embodiments of Formula (AA), J is

X is phenyl substituted with one or more halogens (e.g., X is phenyl substituted with fluoro and chloro); Z is

R1 is selected from C1-3 alkyl and C2-4 alkenyl; R2 is H; R3 is selected from H and C1-3 alkyl; or R1 and R3 together with the nitrogen and sulfur atom to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring; R4 is H; R5 is H; R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having the formula:

and R8 is selected from H and halogen (e.g., R8 is H).

In some embodiments, the present application provides a compound of Formula (A):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and the phenyl is optionally substituted with one or more RX;

Z is selected from

R1 is selected from H and C1-3 alkyl;

R2 is selected from H and C1-3 alkyl;

R3 is selected from H and C1-3 alkyl;

or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;

R4 is selected from H and C1-3 alkyl;

R5 is selected from H and C1-6 alkyl;

R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, and 3-6 membered heterocyclyl;

R7 is selected from H and C1-6 alkyl;

or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;

or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with OH;

Q is selected from C1-2 alkylene or —C(O)—;

R21 is selected from H, C1-6 alkyl optionally substituted with 1-3 halo;

R8 is selected from H and halogen;

each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;

or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;

each Ra is independently selected from H and C1-6 alkyl;

each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;

each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and

each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments, the present application provides a compound of Formula (I).

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;
  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
  • R4 is selected from H and C1-3 alkyl;
  • R5 is selected from H and C1-6 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, and 3-6 membered heterocyclyl;
  • R7 is selected from H and C1-6 alkyl;
  • or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

  • wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
  • or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

  • R8 is selected from H and halogen;
  • each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
  • or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
  • each Ra is independently selected from H and C1-6 alkyl;
  • each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
  • each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
  • each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments of Formula (I), each of R1, R2, and R3 is H. In some embodiments, two of R1, R2, and R3 are H, and the other is C1-3 alkyl. For example, two of R1, R2, and R3 are H, and the other is methyl. In some embodiments, one of R1, R2, and R3 is H, and the other two are C1-3 alkyl. For example, one of R1, R2, and R3 is H, and the other two are methyl. In some embodiments, R1 is H; and R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring. For example, R1 is H; and R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form an imidazoline.

In some embodiments of Formula (I), R4 is H. In some embodiments, R4 is C1-3 alkyl.

In some embodiments of Formula (I), one of R5 and R7 is H and the other is C1-6 alkyl. For example, R5 is H and R7 is C1-6 alkyl. In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

wherein the ring is optionally substituted on a carbon atom of the ring with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments of Formula (I), R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

wherein the ring is optionally substituted on a carbon atom of the ring with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments of Formula (I), R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments, R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments of Formula (I), R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments of Formula (I), R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments, R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments of Formula (I), R6 is selected from H and C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa. In some embodiments, R6 is H. In some embodiments, R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa. For example, R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of fluorine, bromine and OH.

In some embodiments of Formula (I), R6 is C1-6 alkyl optionally substituted with ORa. In some embodiments, Ra is H. In some embodiments, Ra is C1-6 alkyl.

In some embodiments of Formula (I), R6 is C1-6 alkyl substituted with —SC(NH)NH2. In some embodiments, R6 is C1-6 alkyl optionally substituted with halogen. For example, R6 is C1-6 alkyl optionally substituted with fluoro.

In some embodiments of Formula (I), X is selected from a 5-membered heterocyclyl ring and phenyl, wherein each of the 5-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX. For example, X is a 5-membered heterocyclyl ring optionally substituted with one or more RX. In some embodiments, X is a pyrrolidinyl optionally substituted with one or more RX. In some embodiments, X is a 2- or 3-pyrrolidinyl optionally substituted with one or more RX. In some embodiments, X is phenyl optionally substituted with one or more RX. For example, X is phenyl optionally substituted with three RX.

In some embodiments of Formula (I), RX is independently selected from halogen, C1-6 alkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb. In some embodiments, RX is independently selected from halogen and C1-6 alkyl optionally substituted with one or more Rb. In some such embodiments, Rb is selected from C2-6 alkenyl, ORc, N(Rc)2, and C3-6 cycloalkyl. For example, Rb is selected from C2-6 alkenyl, ORc, NH2, and cyclopropyl. In some such embodiments, Rc is selected from H and C1-6 alkyl. In some embodiments, Rb is selected from vinyl, OH, and NH2. In some embodiments, two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring.

In some embodiments of Formula (I), the compound is a compound of Formula (IA):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;
  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • R4 is selected from H and C1-3 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, or C3-6 heterocyclyl;
  • each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
  • or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
  • each Ra is independently selected from H and C1-6 alkyl;
  • each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
  • each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
  • each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments of Formula (IA), R1, R2, and R3 is H. In some embodiments, two of R1, R2, and R3 are H, and the other is C1-3 alkyl. For example, two of R1, R2, and R3 are H, and the other is methyl. In some embodiments, one of R1, R2, and R3 is H, and the other two are C1-3 alkyl. For example, one of R1, R2, and R3 is H, and the other two are methyl. In some embodiments, R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring. For example, R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form an imidazoline.

In some embodiments of Formula (IA), R4 is H. In some embodiments, R4 is C1-3 alkyl.

In some embodiments of Formula (IA), R6 is selected from H and C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa. In some embodiments, R6 is H. In some embodiments, R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa. For example, R6 is C1-6 alkyl optionally substituted with ORa. In some such embodiments, Ra is H. In other such embodiments, Ra is C1-6 alkyl. In some embodiments, R6 is C1-6 alkyl substituted with —SC(NH)NH2. In some embodiments, R6 is C1-6 alkyl optionally substituted with halogen. For example, R6 is C1-6 alkyl optionally substituted with fluoro.

In some embodiments of Formula (IA), X is selected from a 5-membered heterocyclyl ring and phenyl, wherein each of the 5-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX. In some embodiments, X is a 5-membered heterocyclyl ring optionally substituted with one or more RX. For example, X is a pyrrolidinyl optionally substituted with one or more RX. In some embodiments, X is a 2- or 3-pyrrolidinyl optionally substituted with one or more RX. In some embodiments, X is phenyl optionally substituted with one or more RX. For example, X is phenyl optionally substituted with three RX.

In some embodiments of Formula (IA), RX is independently selected from halogen, C1-6 alkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb. For example, RX is independently selected from halogen and C1-6 alkyl optionally substituted with one or more Rb. In some such embodiments, Rb is selected from C2-6 alkenyl, ORc, N(Rc)2, and C3-6 cycloalkyl. For example, Rb is selected from C2-6 alkenyl, ORc, NH2, and cyclopropyl. In some such embodiments, Rc is selected from H and C1-6 alkyl. In some embodiments, Rb is selected from vinyl, OH, and NH2.

In some embodiments of Formula (IA), two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring.

In some embodiments of Formula (I), the compound is a compound of Formula (IB).

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;
  • each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
  • or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
  • each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
  • each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
  • each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments of Formula (IB), X is selected from a 5-membered heterocyclyl ring and phenyl, wherein each of the 5-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX. In some embodiments, X is a 5-membered heterocyclyl ring optionally substituted with one or more RX. For example, X is a pyrrolidinyl optionally substituted with one or more RX. In some embodiments, X is a 2- or 3-pyrrolidinyl optionally substituted with one or more RX. In some embodiments, X is phenyl optionally substituted with one or more RX. For example, X is phenyl optionally substituted with three RX. In some embodiments, RX is independently selected from halogen, C1-6 alkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb. In some embodiments, RX is independently selected from halogen and C1-6 alkyl optionally substituted with one or more Rb. In some embodiments, Rb is selected from C2-6 alkenyl, ORc, N(Rc)2, and C3-6 cycloalkyl. For example, Rb is selected from C2-6 alkenyl, ORc, NH2, and cyclopropyl. In some such embodiments, Rc is selected from H and C1-6 alkyl. In some embodiments, Rb is selected from vinyl, OH, and NH2.

In some embodiments of Formula (IB), two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring.

Also provided herein is a compound of Formula (II):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
  • R4 is selected from H and C1-3 alkyl;
  • R5 is selected from H and C1-6 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, or C3-6 heterocyclyl;
  • R7 is selected from H and C1-6 alkyl;
  • or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

  • wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
  • or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

  • R8 is selected from H and halogen;
  • R9 is selected from H and halogen;
  • R10 is selected from H, halogen, C1-4 alkyl, C1-4 haloalkyl, and C3-6 cycloalkyl;
  • R11 is selected from C1-3 alkyl, C2-4 alkenyl, and C3-6 cycloalkyl, wherein the C1-3 alkyl is optionally substituted with one or more Rb;
  • R17 is selected from H, halogen, and C1-6 alkyl;
  • each Ra is independently selected from H and C1-6 alkyl;
  • each Rb is independently selected from ORc, —C(O)ORc, and —(O)aryl, wherein the aryl is optionally substituted with one or more Rd;
  • each Rc is independently selected from hydrogen, C1-3 alkyl, OH, O(C1-3 alkyl), NO2, —C(O)aryl, and aryl; and
  • each Rd is C1-3 alkyl.

In some embodiments of Formula (II), each of R1, R2, and R3 is H. In some embodiments, two of R1, R2, and R3 are H, and the other is C1-3 alkyl. For example, two of R1, R2, and R3 are H, and the other is methyl. In some embodiments, one of R1, R2, and R3 is H, and the other two are C1-3 alkyl. For example, one of R1, R2, and R3 is H, and the other two are methyl. In some embodiments, R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring. For example, R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form an imidazoline.

In some embodiments of Formula (II), R4 is H. In some embodiments, R4 is C1-3 alkyl.

In some embodiments of Formula (II), one of R5 and R7 is H and the other is C1-6 alkyl. For example, R5 is H and R7 is C1-6 alkyl. In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

wherein the ring is optionally substituted on a carbon atom of the ring with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments of Formula (II), R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

wherein the ring is optionally substituted on a carbon atom of the ring with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments of Formula (II), R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments, R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments of Formula (II), R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring-having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments, R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments, R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments of Formula (II), R6 is selected from H and C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa. In some embodiments, R6 is H. In some embodiments, R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa. In some embodiments, R6 is C1-6 alkyl optionally substituted with OR. In some such embodiments, Ra is H. In other such embodiments, Ra is C1-6 alkyl. In some embodiments, R6 is C1-6 alkyl substituted with —SC(NH)NH2. In some embodiments, R6 is C1-6 alkyl optionally substituted with halogen. For example, R6 is C1-6 alkyl optionally substituted with fluoro.

In some embodiments of Formula (II), R8 is H. In some embodiments, R8 is halogen. For example, R8 is F.

In some embodiments of Formula (II), one of R9 and R10 is halogen and the other is H. In some embodiments, each of R9 and R10 is halogen. For example, R9 is fluoro and R10 is chloro. In some embodiments, R9 is H and R10 is C1-4 alkyl. In some embodiments, R9 is halogen and R10 is C1-4 alkyl. For example, R9 is chloro and R10 is C1-4 alkyl. In some embodiments, R9 is fluoro and R10 is C1-4 alkyl. In some such embodiments, R10 is ethyl or R10 is isopropyl. In some embodiments, R9 is H and R10 is C3-6 cycloalkyl. For example, R10 is cyclopropyl or R10 is cyclopentyl.

In some embodiments of Formula (II), R11 is selected from C1-3 alkyl optionally substituted with one or more Rb, and C2-4 alkenyl. In some embodiments, R11 is selected from C1-3 alkyl optionally substituted with one or more R, and C3-6 cycloalkyl. In some embodiments, R11 is C1-3 alkyl optionally substituted with one or more Rb. In some such embodiments, Rb is ORc, wherein Rc is selected from H and C1-3 alkyl. For example, Rc is H. In some embodiments, Rc is C1-3 alkyl. In some embodiments, R11 is C1-3 alkyl. For example, R11 is methyl. In some embodiments, R11 is C3-6 cycloalkyl. For example, R11 is cyclopropyl.

In some embodiments of Formula (II), the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (R). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

In some embodiments, R17 is H. In some embodiments, R17 is halogen.

In some embodiments of Formula (II), each of R9 and R10 is halogen, and R11 is C1-3 alkyl optionally substituted with one or more Rb. For example, R9 is fluoro, R10 is chloro, and R11 is C1-3 alkyl optionally substituted with one or more Rb. In some embodiments, R9 is fluoro, R10 is chloro, and R11 is methyl. In some embodiments, R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

In some embodiments of Formula (II), each of R1, R2, and R3 is H; R4 is H; R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa; R9 is fluoro, R10 is chloro, and R11 is methyl. In some embodiments, each of R1, R2, and R3 is H; R4 is H; R6 is H or C1-6 alkyl; R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S). In some embodiments, each of R1, R2, and R3 is H; R4 is H; R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

R9 is fluoro; R10 is chloro; and R11 is methyl. In some embodiments, each of R1, R2, and R3 is H; R4 is H; R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

R6 is H or C1-6 alkyl; R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

In some embodiments of Formula (II), the compound is a compound of Formula (IIA):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • R4 is selected from H and C1-3 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen and ORa;
  • R9 is selected from H and halogen;
  • R10 is selected from H, halogen, C1-4 alkyl, C1-4 haloalkyl, and C3-6 cycloalkyl
  • R11 is selected from C1-3 alkyl, C2-4 alkenyl, and C3-6 cycloalkyl, wherein the C1-3 alkyl is optionally substituted with one or more Rb;
    • each Ra is independently selected from H and C1-6 alkyl;
    • each Rb is independently selected from ORc, —C(O)ORc, and —(O)aryl, wherein the aryl is optionally substituted with one or more Rd;
    • each Rc is independently selected from hydrogen, C1-3 alkyl, OH, O(C1-3 alkyl), NO2, —C(O)aryl, and aryl; and each Rd is C1-3 alkyl.

In some embodiments of Formula (IIA), each of R1, R2, and R3 is H. In some embodiments, two of R1, R2, and R3 are H, and the other is C1-3 alkyl. For example, two of R1, R2, and R3 are H, and the other is methyl. In some embodiments, one of R1, R2, and R3 is H, and the other two are C1-3 alkyl. For example, one of R1, R2, and R3 is H, and the other two are methyl. In some embodiments, R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring. For example, R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form an imidazoline.

In some embodiments of Formula (IIA), R4 is H.

In some embodiments of Formula (IIA), R6 is selected from H and C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa. In some embodiments, R6 is H. In some embodiments, R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa. For example, R6 is C1-6 alkyl optionally substituted with ORa. In some such embodiments, Ra is H. In other such embodiments, Ra is C1-6 alkyl. In some embodiments, R6 is C1-6 alkyl optionally substituted with halogen. For example, R6 is C1-6 alkyl optionally substituted with fluoro.

In some embodiments of Formula (IIA), one of R9 and R10 is halogen and the other is H. In some embodiments, each of R9 and R10 is halogen. For example, R9 is fluoro and R10 is chloro. In some embodiments, R9 is H and R10 is C1-4 alkyl. In some embodiments, R9 is halogen and R10 is C1-4 alkyl. For example, R9 is chloro and R10 is C1-4 alkyl. In some embodiments, R9 is fluoro and R10 is C1-4 alkyl. For example, R10 is ethyl or R10 is isopropyl. In some embodiments, R9 is H and R10 is C3-6 cycloalkyl. For example, R10 is cyclopropyl or R10 is cyclopentyl.

In some embodiments of Formula (IIA), R11 is selected from C1-3 alkyl optionally substituted with one or more R, and C2-4 alkenyl. In some embodiments, R11 is selected from C1-3 alkyl optionally substituted with one or more R, and C3-6 cycloalkyl. In some embodiments, R11 is C1-3 alkyl optionally substituted with one or more Rb. In some such embodiments, Rb is ORc, and wherein Rc is selected from H and C1-3 alkyl. For example, Rc is H. In some embodiments, Rc is C1-3 alkyl. In some embodiments, R11 is C1-3 alkyl. For example, R11 is methyl. In some embodiments, R11 is C3-6 cycloalkyl. For example, R11 is cyclopropyl.

In some embodiments of Formula (IIA), the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (R). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

In some embodiments of Formula (IIA), each of R9 and R10 is halogen, and R11 is C1-3 alkyl optionally substituted with one or more Rb. For example, R9 is fluoro, R10 is chloro, and R11 is C1-3 alkyl optionally substituted with one or more Rb. In some embodiments, R9 is fluoro, R10 is chloro, and R11 is methyl. In some embodiments, R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

In some embodiments of Formula (IIA), each of R1, R2, and R3 is H; R4 is H; R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa; R9 is fluoro, R10 is chloro, and R11 is methyl. In some embodiments, each of R1, R2, and R3 is H; R4 is H; R6 is H or C1-6 alkyl; R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

In some embodiments of Formula (II), the compound is a compound of Formula (IIB):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • R9 is selected from H and halogen;
  • R10 is selected from H, halogen, C1-4 alkyl, C1-4 haloalkyl, and C3-6 cycloalkyl;
  • R11 is selected from C1-3 alkyl, C2-4 alkenyl, and C3-6 cycloalkyl, wherein the C1-3 alkyl is optionally substituted with one or more Rb;
  • each Rb is independently selected from ORc, —C(O)ORc, and —(O)aryl, wherein the aryl is optionally substituted with one or more Rd;
  • each Rc is independently selected from hydrogen, C1-3 alkyl, OH, O(C1-3 alkyl), NO2, —C(O)aryl, and aryl; and
  • each Rd is C1-3 alkyl.

In some embodiments of Formula (IIB), one of R9 and R10 is halogen and the other is H. In some embodiments, each of R9 and R10 is halogen. For example, R9 is fluoro and R10 is chloro. In some embodiments, R9 is H and R10 is C1-4 alkyl. In some embodiments, R9 is halogen and R10 is C1-4 alkyl. For example, R9 is chloro and R10 is C1-4 alkyl. In some embodiments, R9 is fluoro and R10 is C1-4 alkyl. For example, R10 is ethyl or R10 is isopropyl. In some embodiments, R9 is H and R10 is C3-6 cycloalkyl. For example, R10 is cyclopropyl or R10 is cyclopentyl.

In some embodiments of Formula (IIB), R11 is selected from C1-3 alkyl optionally substituted with one or more Rb, and C2-4 alkenyl. In some embodiments, R11 is selected from C1-3 alkyl optionally substituted with one or more R, and C3-6 cycloalkyl. In some embodiments, R11 is C1-3 alkyl optionally substituted with one or more Rb. In some such embodiments, Rb is ORc, and Rc is selected from H and C1-3 alkyl. For example, Rc is H. In some embodiments, Rc is C1-3 alkyl. In some embodiments, R11 is C1-3 alkyl. For example, R11 is methyl. In some embodiments, R11 is C3-6 cycloalkyl. For example, R11 is cyclopropyl.

In some embodiments of Formula (IIB), the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (R). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

In some embodiments of Formula (IIB), each of R9 and R10 is halogen, and R11 is C1-3 alkyl optionally substituted with one or more Rb. For example, R9 is fluoro, R10 is chloro, and R11 is C1-3 alkyl optionally substituted with one or more Rb. In some embodiments, R9 is fluoro, R10 is chloro, and R11 is methyl. In some embodiments, R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

Also provided herein is a compound of Formula (III).

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
  • R4 is selected from H and C1-3 alkyl;
  • R5 is selected from H and C1-6 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, and —SC(NH)NH2;
  • R7 is selected from H and C1-6 alkyl;
  • or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

  • wherein the ring is optionally substituted on a carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
  • or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

  • R8 is selected from H and halogen;
  • R12 is selected from H, halogen, and C1-6 alkyl;
  • R13 is selected from H, halogen, C1-6 alkyl, ORc, N(Rc)2, wherein C1-6 alkyl is optionally substituted with one or more of OR and aryl;
  • R14 is selected from H and aryl;
  • or R13 and R14 together with the carbon atoms to which they are attached form a 5- or 6-membered cycloalkyl ring;
  • R15 is selected from H, C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more Rb.
  • each Ra is independently selected from H and C1-6 alkyl;
  • each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
  • each Rc is independently selected from hydrogen, C1-6 alkyl, aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
  • each Rd is independently selected from OH, O(C1-3 alkyl), NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments of Formula (III), each of R1, R2, and R3 is H. In some embodiments, two of R1, R2, and R3 are H, and the other is C1-3 alkyl. For example, two of R1, R2, and R3 are H, and the other is methyl. In some embodiments, one of R1, R2, and R3 is H, and the other two are C1-3 alkyl. For example, one of R1, R2, and R3 is H, and the other two are methyl.

In some embodiments of Formula (III), R4 is H.

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

wherein the ring is optionally substituted on a carbon atom of the ring with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments of Formula (III), R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

wherein the ring is optionally substituted on a carbon atom of the ring with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments, R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

In some embodiments of Formula (III), R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments, R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments of Formula (III), R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:

wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH. In some embodiments, R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments, R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

In some embodiments of Formula (III), R6 is selected from H or C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, and —SC(NH)NH2. In some embodiments, R6 is H.

In some embodiments of Formula (III), R8 is H. In some embodiments, R8 is halogen. For example, R8 is F.

In some embodiments of Formula (III), each of R12 and R13 is H. In some embodiments, each of R12 and R13 is not H. In some embodiments, one of R12 and R13 is H, and the other is not H. For example, R12 is H, and R13 is C1-6 alkyl optionally substituted with one or more of ORc or aryl. In some embodiments, R12 is H, and R13 is C1-6 alkyl optionally substituted with ORc. In some embodiments, R12 is H, and R13 is C1-6 alkyl. In some embodiments, R12 is H, and R13 is halogen. In some embodiments, R12 is H, and R13 is fluoro. In some embodiments, each of R12 and R13 is C1-6 alkyl. In some embodiments, each of R12 and R13 is halogen. For example, each of R12 and R13 is fluoro. In some embodiments, R12 is H, and R13 is ORc. In some embodiments, R12 is H, and R13 is N(Rc)2. In some such embodiments, each Rc is selected from H or C1-6 alkyl. In some other such embodiments, one Rc is H and the other Rc is C1-6 alkyl.

In some embodiments of Formula (III), each Rc is selected from H or C1-6 alkyl. In some embodiments, each Rc is selected from C1-6 alkyl or —(CH2)aryl. In some embodiments, each Rc is H. In some embodiments, each Rc is C1-6 alkyl. For example, each Rc is methyl. In some embodiments, each Rc is —(CH2)aryl.

In some embodiments of Formula (III), the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (R). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (S).

In some embodiments of Formula (III), R14 is H. In some embodiments, R14 is aryl. In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R14 is bonded to is (R). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R14 is bonded to is (S).

In some embodiments of Formula (III), R15 is H. In some embodiments, R15 is C1-6 alkyl optionally substituted with one or more Rb. For example, R15 is C1-6 alkyl optionally substituted with one Rb. In some embodiments, R15 is C4-5 alkyl optionally substituted with one Rb.

In some embodiments of Formula (III), Rb is selected from ORc, N(Rc)2, and cyclopropyl. For example, Rb is N(Rc)2. In some embodiments, Rb is NH2.

In some embodiments of Formula (III), the compound is a compound of Formula (IIIA):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • R1 is selected from H and C1-3 alkyl;
  • R2 is selected from H and C1-3 alkyl;
  • R3 is selected from H and C1-3 alkyl;
  • R4 is selected from H and C1-3 alkyl;
  • R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, and —SC(NH)NH2;
  • R8 is selected from H and halogen;
  • R12 is selected from H, halogen, and C1-6 alkyl;
  • R13 is selected from H, halogen, C1-6 alkyl, ORc, N(Rc)2, wherein C1-6 alkyl is optionally substituted with one or more of ORc and aryl; R14 is selected from H and aryl;
  • or R13 and R14 together with the carbon atoms to which they are attached form a 5- or 6-membered cycloalkyl ring;
  • R16 is C1-6 alkyl;
  • each Ra is independently selected from H and C1-6 alkyl;
  • each Rc is independently selected from hydrogen, C1-6 alkyl, aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
  • each Rd is independently selected from OH, O(C1-3 alkyl), NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments of Formula (IIIA), each of R1, R2, and R3 is H. In some embodiments, two of R1, R2, and R3 are H, and the other is C1-3 alkyl. For example, two of R1, R2, and R3 are H, and the other is methyl. In some embodiments, one of R1, R2, and R3 is H, and the other two are C1-3 alkyl. For example, one of R1, R2, and R3 is H, and the other two are methyl.

In some embodiments of Formula (IIIA), R4 is H.

In some embodiments of Formula (IIIA), R6 is selected from H or C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, or —SC(NH)NH2. In some embodiments, R6 is H.

In some embodiments of Formula (IIIA), R8 is H. In some embodiments, R8 is halogen. For example, R8 is F.

In some embodiments, of Formula (IIIA), each of R12 and R13 is H. In some embodiments, each of R12 and R13 is not H. In some embodiments, one of R12 and R13 is H, and the other is not H. For example, R12 is H, and R13 is C1-6 alkyl optionally substituted with one or more of ORc or aryl. In some embodiments, R12 is H, and R13 is C1-6 alkyl optionally substituted with OR. In some embodiments, R12 is H, and R13 is C1-6 alkyl. In some embodiments, R12 is H, and R13 is halogen. For example, R12 is H, and R13 is fluoro. In some embodiments, each of R12 and R13 is C1-6 alkyl. In some embodiments, each of R12 and R13 is halogen. For example, each of R12 and R13 is fluoro. In some embodiments, R12 is H, and R13 is OR. In some embodiments, R12 is H, and R13 is N(Rc)2. In some such embodiments, each Rc is selected from H or C1-6 alkyl. In other such embodiments, one Rc is H and the other Rc is C1-6 alkyl.

In some embodiments of Formula (IIIA), each Rc is selected from H or C1-6 alkyl. In some embodiments, Rc is selected from C1-6 alkyl or —(CH2)aryl. In some embodiments, Rc is H. In some embodiments, Rc is C1-6 alkyl. For example, Rc is methyl. In some embodiments, Rc is —(CH2)aryl. For example, Rc is —(CH2)phenyl.

In some embodiments of Formula (IIIA), the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (R). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (S).

In some embodiments of Formula (IIIA), R14 is H. In some embodiments, R14 is aryl. In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R14 is bonded to is (R). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R14 is bonded to is (S).

In some embodiments of Formula (IIIA), R16 is C1-6 alkyl. For example, R16 is methyl. In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (S). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (R).

In some embodiments of Formula (III), the compound is a compound of Formula (IIIB):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:

  • R12 is selected from H, halogen, and C1-6 alkyl;
  • R13 is selected from H, halogen, C1-6 alkyl, ORc, N(Rc)2, wherein C1-6 alkyl is optionally substituted with one or more of ORc and aryl;
  • R16 is C1-6 alkyl;
  • each Rc is independently selected from hydrogen, C1-6 alkyl, aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
  • each Rd is independently selected from OH, O(C1-3 alkyl), NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

In some embodiments of Formula (IIIB), each of R12 and R13 is H. In some embodiments, each of R12 and R13 is not H. In some embodiments, one of R12 and R13 is H, and the other is not H. In some embodiments, R12 is H, and R13 is C1-6 alkyl optionally substituted with one or more of ORc or aryl. In some embodiments, R12 is H, and R13 is C1-6 alkyl optionally substituted with ORc. In some embodiments, R12 is H, and R13 is C1-6 alkyl. In some embodiments, R12 is H, and R13 is halogen. For example, R12 is H, and R13 is fluoro. In some embodiments, each of R12 and R13 is C1-6 alkyl. In some embodiments, each of R12 and R13 is halogen. For example, each of R12 and R13 is fluoro. In some embodiments, R12 is H, and R13 is ORc. In some embodiments, R12 is H, and R13 is N(Rc)2.

In some embodiments of Formula (IIIB), each Rc is selected from H or C1-6 alkyl. In some embodiments, each Rc is selected from C1-6 alkyl or —(CH2)aryl. In some embodiments, each Rc is H. In some embodiments, each Rc is C1-6 alkyl. For example, each Rc is methyl. In some embodiments, each Rc is —(CH2)aryl.

In some embodiments of Formula (IIIB), the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (R). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (S).

In some embodiments of Formula (IIIB), R16 is C1-6 alkyl. For example, R16 is methyl. In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (S). In some embodiments, the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (R).

In some embodiments, one of R12 and R13 is H, and the other of R12 and R13 is not H; and R16 is methyl, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (S). In some embodiments, R12 is H, and R13 is ORc; Rc is selected from H or C1-6 alkyl; and R16 is methyl, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (S).

In some embodiments of Formula (I), Formula (II), or Formula (III), the present disclosure provides any one of compounds listed in Table 1, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer.

TABLE 1 ESI, MW m/z # Structure (g/mol) [M + H]+ 1 636.24 636.7 2 636.24 636.3 3 662.27 662.3 4 670.27 670.5 5 696.31 696.5 6 668.28 668.5 7 694.32 694.5 8 626.20 626.4 9 584.16 584.4 10 730.30 730.5 11 624.23 624.5 12 624.23 624.5 13 650.26 650.5 14 622.21 622.4 15 648.25 648.5 16 608.18 608.5 17 668.28 668.5 18 694.32 694.5 19 726.39 726.5 20 752.42 752.5 21 628.19 628.4 22 644.19 644.4 23 582.14 582 24 582.14 582 25 654.23 654.4 26 598.19 598.5 27 610.20 610.5 28 611.18 611.5 29 596.17 596 30 612.17 612 31 550.77 551 32 550.77 551 33 564.80 565 34 610.20 610.3 35 702.30 702.5 36 640.20 640.4 37 596.17 596 38 612.17 612 39 732.32 732.5 40 716.32 716.4 41 624.23 624.5 42 624.23 624.5 43 638.25 638.4 44 638.25 638.3 45 622.21 622.3 46 650.26 650.4 47 552.74 553 48 747.29 747.4 49 571.75 572 50 636.24 636.5 51 519.71 520 52 624.23 624.5 53 604.86 605.5 54 568.12 568 55 568.12 568 56 652.28 652.5 57 580.80 581 58 562.78 563 59 622.21 622.5 60 552.74 553 61 479.65 480 62 604.86 605 63 656.90 657 64 566.78 567 65 565.79 566 66 579.81 580 67 564.80 565 68 564.80 565 69 568.76 569 70 568.76 569 71 586.75 587 72 586.75 587 73 562.78 563 74 578.83 579 75 548.76 549 76 640.90 641 77 626.87 627 78 624.23 624.5 79 624.23 624.5 80 640.22 640.2 81 580.80 581 82 620.26 620.4 83 494.66 495 84 617.83 618.5 85 603.81 604.5 86 613.87 614.5 87 585.82 586.3 88 480.64 481 89 621.80 622 90 666.31 666 91 684.34 684 92 599.80 600 93 611.85 612.4 94 664.29 664 95 745.20 746 96 682.26 682 97 668.28 668 98 656.24 656 99 597.83 598.5 100 625.87 626 101 682.29 682 102 722.36 722 103 678.31 678 104 621.79 622 105 682.29 682 106 640.22 640.5 107 625.88 626.0 108 682.30 682.0 109 722.37 722.0 110 678.31 678.0 111 624.22 624.5 112 621.8 622.0 113 682.30 682.0 114 596.17 596.4 115 624.18 624.5 116 596.17 596.4 117 656.24 656.8 118 638.25 638.0 119 652.27 652.0 120 624.22 624.0 121 642.21 642.5 122 650.26 650.4 123 636.23 636.4

In Table 1 above, a bond indicated by a squiggle bond indicates a stereoisomer where the identity of the specific orientation of the bond has not been specified.

In some embodiments, the present disclosure relates to a compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer that binds the ribosome. In some embodiments, the ribosome is a bacterial ribosome.

In some embodiments, the present disclosure relates to a pharmaceutical composition comprising a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, and a pharmaceutically acceptable carrier. In some embodiments, the present disclosure relates to a compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer disclosed herein and a means for delivery.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of or delaying the onset of a disease state in a human or animal comprising administering to the human or animal in need thereof an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, the present disclosure relates to use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of, a microbial infection in a human or animal.

In another aspect, the present disclosure relates to a compound for use in the manufacture of a medicament for treating a microbial infection in a subject, wherein the compound is selected from a compound of the present disclosure, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, the present disclosure relates to a compound for use in the manufacture of a medicament for preventing a microbial infection in a subject, wherein the compound is selected from a compound of the present disclosure, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, the present disclosure relates to a compound for use in the manufacture of a medicament for reducing the risk of a microbial infection in a subject, wherein the compound is selected from a compound of the present disclosure, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, the present disclosure relates to a compound for use in the manufacture of a medicament for delaying the onset of a microbial infection in a subject, wherein the compound is selected from a compound of the present disclosure, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, the present disclosure relates to a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, for use in treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal.

In some embodiments, the present disclosure relates to a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, for use in treating a microbial infection in a human or animal.

In some embodiments, the present disclosure relates to a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, for use in preventing a microbial infection in a human or animal.

In some embodiments, the present disclosure relates to a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, for use in reducing the risk of a microbial infection in a human or animal.

In some embodiments, the present disclosure relates to a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, for use in delaying the onset of a microbial infection in a human or animal.

In some embodiments, a microbial infection as described herein is caused by one or more microoganisms selected from the group consisting of: Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter species, and Escherichia coli. This group of microoganisms can be referred to generally as the ESKAPE pathogens. In some embodiments, the microbial infection is caused by a microorganism which is resistant to at least one antibacterial. For example, the microorganism can be classified as multi-drug resistant or extremely-drug resistant. In some embodiments, the compounds provided herein have in vitro activity across the ESKAPE pathogens. For example, one or more of the compounds provided herein exhibit individual MICs and/or MIC90s of ≤4 mg/L. In some embodiments, one or more of the compounds provided herein exhibit individual MICs and/or MIC90s of ≤2 mg/L. For example, one or more of the compounds provided herein exhibit individual MICs and/or MIC90s of ≤1 mg/L. In some embodiments, one or more of the compounds provided herein exhibit individual MICs and/or MIC90s of ≤0.5 mg/L. For example, one or more of the compounds provided herein exhibit individual MICs and/or MIC90s of ≤0.25 mg/L. In some embodiments, one or more of the compounds provided herein exhibit individual MICs and/or MIC90s of ≤0.125 mg/L. For example, one or more of the compounds provided herein exhibit individual MICs and/or MIC90s of ≤0.05 mg/L.

In some embodiments, the compounds provided herein lack cross-resistance to current therapies, with demonstrated activity against one or more multidrug-resistant strains of E. faecium and MRSA; Enterobacteriaceae featuring cephalosporinases (ESBLs and AmpCs) and carbapenemases (classes A, B and D); P. aeruginosa strains with normal and raised efflux; and A. baumannii. In some embodiments, the compounds provided herein demonstrate one or more of low rate (E-10) and extent of resistance development in E. coli; activity in exemplary burden models of infection in the neutropenic thigh, ascending kidney and lung as well as in peritonitis models; and safety scorecard highlighted by 14-day dose-range-finding toxicology studies in rat and monkey, at multiples the exposures observed for efficacy, with minimal histopathological findings.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal comprising administering to the human or animal an effective amount of a compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, wherein the microbial infection is caused by one or more of the following microorganisms: Acinetobacter spp. (Acinetobacter baumanni), Bacteroides distasonis, Bacteroides fragilis, Bacteroides ovatus, Bacteroides thetaiotaomicron, Bacteroides uniformis, Bacteroides vulgatus, Citrobacter freundii, Citrobacter koser, Chlamydia trachomatis, Chlamydia psittaci, Chlamydia pneumoniae, Chlamydia pecorum, Chlamydia suis, Chlaymdia muridarum, Chlamydophila psittaci, Chlamydophila pneumoniae, Chlamydophila pecorum, Clostridioides spp. (e.g., Clostridioides difficile and Clostridioides mangenotii), Clostridium clostridioforme, Clostridium perfringens, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus spp. (vancomycin susceptible and resistant isolates), Escherichia coli (including ESBL and KPC producing isolates), Eubacterium lentum, Fusobacterium spp., Haemophilus influenzae (including beta-lactamase positive isolates), Haemophilus parainfluenzae, Klebsiella pneumoniae (including ESBL and KPC producing isolates), Klebsiella oxytoca (including ESBL and KPC producing isolates), Legionella pneumophilia Moraxella catarrhalis, Morganella morganii, Mycoplasma spp., Neisseria gonorrhoeae (including Neisseria gonorrhoeae ATCC49266, Neisseria gonorrhoeae 255123, Neisseria gonorrhoeae 255124, Neisseria gonorrhoeae 255125, Neisseria gonorrhoeae 255126, Neisseria gonorrhoeae 255127, Neisseria gonorrhoeae J9104300210, Neisseria gonorrhoeae J9107400107, Neisseria gonorrhoeae J9109510210, Neisseria gonorrhoeae J9108110210), Peptostreptococcus spp., Porphyromonas asaccharolytica, Prevotella bivia, Proteus mirabilis, Proteus vulgaris, Providencia rettgeri, Providencia stuartii, Pseudomonas aeruginosa, Salmonella enteritidis, Serratia marcescens, Streptococcus anginosus, Staphylococcus aureus (methicillin susceptible and resistant isolates), Staphylococcus epidermidis (methicillin susceptible and resistant isolates), Stenotrophomonas maltophilia, Streptococcus agalactiae, Streptococcus constellatus, Streptococcus pneumoniae (penicillin susceptible and resistant isolates), Streptococcus pyogenes, or Streptococcus pyogenes.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal comprising administering to the human or animal an effective amount of a compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, wherein the infection is caused by or involves one or more microorganisms selected from: Acinetobacter spp. (Acinetobacter baumanni), Bacteroides distasonis, Bacteroides fragilis, Bacteroides ovatus, Bacteroides thetaiotaomicron, Bacteroides unformis, Bacteroides vulgatus, Citrobacter freundii, Citrobacter koser, Chlamydia trachomatis, Chlamydia psittaci, Chlamydia pneumoniae, Chlamydia pecorum, hlamydia suis, Chlaymdia muridarum, Chlamydophila psittaci, Chlamydophila pneumoniae, Chlamydophila pecorum, Clostridioides spp. (e.g., Clostridioides difficile and Clostridioides mangenotii), Clostridium clostridioforme, Clostridium perfringens, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus spp., Escherichia coli, Eubacterium lentum, Fusobacterium spp., Haemophilus influenzae, Haemophilus parainfluenzae, Klebsiella pneumoniae, Klebsiella oxytoca, Legionella pneumophilia, Moraxella catarrhalis, Morganella morganii, Mycoplasma spp., Neisseria gonorrhoeae, Peptostreptococcus spp., Porphyromonas asaccharolytica, Prevotella bivia, Proteus mirabilis, Proteus vulgaris, Providencia rettgeri, Providencia stuartii, Pseudomonas aeruginosa, Salmonella enteritidis, Serratia marcescens, Streptococcus anginosus, Staphylococcus aureus, Staphylococcus epidermidis, Stenotrophomonas maltophilia, Streptococcus agalactiae, Streptococcus constellatus, Streptococcus pneumoniae, Streptococcus pyogenes, and Streptococcus pyogenes.

In some embodiments, the present disclosure relates to a method wherein the infection is caused by or involves one or more of aerobic and facultative gram-positive microorganisms selected from: Staphylococcus aureus, Streptococcus pneumoniae, Enterococcus spp., Streptococcus agalactiae, Streptococcus pyogenes, and Staphylococcus epidermidis.

In some embodiments, the present disclosure relates to a method wherein the infection is caused by or involves one or more of aerobic and facultative gram-negative microorganisms selected from: Escherichia coli, Haemophilus influenzae, Klebsiella pneumoniae, Citrobacter freundii, Chlamydia trachomatis, Chlamydia psittaci, Chlamydia pneumoniae, Chlamydia pecorum, hlamydia suis, Chlaymdia muridarum, Chlamydophila psittaci, Chlamydophila pneumoniae, Chlamydophila pecorum, Enterobacter aerogenes, Enterobacter cloacae, Morganella morganii, Neisseria gonorrhoeae, Salmonella enteritidis, Serratia marcescens, Pseudomonas aeruginosa, Acinetobacter baumanni, Moraxella catarrhalis, Proteus mirabilis, Citrobacter koseri, Haemophilus parainfluenzae, Klebsiella oxytoca, Proteus vulgaris, Providencia rettgeri, and Providencia stuarti.

In some embodiments, the present disclosure relates to a method wherein the infection is caused by or involves one or more anaerobic microorganisms: Bacteroides fragilis, Bacteroides distasonis, Bacteroides ovatus, Bacteroides thetaiotaomicron, Bacteroides uniformis, Clostridioides spp. (e.g., Clostridioides difficile and Clostridioides mangenotii), Clostridium clostridioforme, Eubacterium lentum, Peptostreptococcus spp., Porphyromonas asaccharolytica, Prevotella bivia, Bacteroides vulgatus, Clostridium perfringens, and Fusobacterium spp.

In some embodiments, the present disclosure relates to a method, wherein the microorganism Enterococcus spp. is selected from vancomycin susceptible isolate and vancomycin resistant isolate. For example, vancomycin-resistant Enterococci.

In some embodiments, the present disclosure relates to a method wherein the microorganism Escherichia coli is selected from extended spectrum beta-lactamase (ESBL) producing isolate and Klebsiella pneumoniae carbapenemase (KPC) producing isolate.

In some embodiments, the present disclosure relates to a method wherein the microorganism Haemophilus influenzae is a beta-lactamase positive isolate.

In some embodiments, the present disclosure relates to a method wherein, the microorganism Klebsiella pneumoniae is selected from extended spectrum beta-lactamase (ESBL) producing isolate and Klebsiella pneumoniae carbapenemase (KPC) producing isolate.

In some embodiments, the present disclosure relates to a method wherein the microorganism Klebsiella oxytoca selected from extended spectrum beta-lactamase (ESBL) producing isolate and Klebsiella pneumoniae carbapenemase (KPC) producing isolate.

In some embodiments, the present disclosure relates to a method wherein the microorganism Staphylococcus aureus is selected from methicillin susceptible isolate and methicillin resistant isolate.

In some embodiments, the present disclosure relates to a method wherein the microorganism Staphylococcus epidermidis is selected from methicillin susceptible isolate and methicillin resistant isolate.

In some embodiments, the present disclosure relates to a method wherein the microorganism Streptococcuspneumoniae is selected from penicillin susceptible isolate and penicillin resistant isolate.

In some embodiments, the present disclosure relates to a method wherein the microorganism Neisseria gonorrhoeae is selected from susceptible and resistant isolates, including, for example, ceftriaxone-resistant, ciprofloxacin-resistant and azithromycin-resistant isolates.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal comprising administering to the human or animal an effective amount of a compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, wherein the microbial infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons, e.g., wherein the one or more microorganisms are selected from Bacillus anthracis and Multi Drug Resistant (MDR) anthracis, Franciscella tularensis, Yersinia pestis, Burkholderia mallei, and Burkholderia pseudomallei.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal comprising administering to the human or animal an effective amount of a compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, wherein the microbial infection is caused by one or more of the following microorganisms: Bacillus anthracis and Multi Drug Resistant (MDR) anthracis, Franciscella tularensis, Yersinia pestis, Burkholderia mallei, and Burkholderia pseudomallei.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal, wherein the microbial infection is selected from the group consisting of: a skin infection, a Gram positive infection, a Gram negative infection, nosocomial pneumonia, community acquired pneumonia, post-viral pneumonia, hospital acquired pneumonia/ventilator associated pneumonia, a respiratory tract infection such as chronic respiratory tract infection (CRTI), acute pelvic infection, a complicated skin and skin structure infection, a skin and soft tissue infection (SSTI) including uncomplicated skin and soft tissue infections (uSSTI)s and complicated skin and soft tissue infections, an abdominal infection, a complicated intra-abdominal infection, a urinary tract infection, bacteremia, septicemia, endocarditis, an atrio-ventricular shunt infection, a vascular access infection, meningitis, surgical prophylaxis, a peritoneal infection, a bone infection, a joint infection, a methicillin-resistant Staphylococcus aureus infection, a vancomycin-resistant Enterococci infection, a ciprofloxacin-resistant Neisseria gonorrhoeae infection, a carbapenem-resistant Enterobacteriaceae infection, a linezolid-resistant organism infection, gonorrhea, chlamydia, and tuberculosis.

The compounds of the present disclosure can be used, for example for the treatment of patients with moderate to severe infections, which may be caused by susceptible isolates of the indicated microorganisms.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a complicated intra-abdominal infection in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a complicated intra-abdominal infection in a human or animal.

In some embodiments, the complicated intra-abdominal infection is selected from polymicrobial infections such as abscess due to Escherichia coli, Clostridium clostridioforme, Eubacterium lentum, Peptostreptococcus spp., Bacteroides fragilis, Bacteroides distasonis, Bacteroides ovatus, Bacteroides thetaiotaomicron, Bacteroides unformis, Streptococcus anginosus, Streptococcus constellatus, Enterococcus faecalis, Proteus mirabilis, or Clostridium perfringens.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a complicated skin and skin structure infection (cSSSI, also known as acute bacterial skin and skin structure infections or ABSSSI) in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a complicated skin and skin structure infection.

In some embodiments, the complicated skin and skin structure infection is selected from diabetic foot infections without osteomyelitis due to Staphylococcus aureus (methicillin susceptible and resistant isolates), Streptococcus agalactiae, Streptococcus pyogenes, Escherichia coi, Klebsiella pneumoniae, Proteus mirabilis, Bacteroides fragilis, Peptostreptococcus species, Porphyromonas asaccharolytica, or Prevotella bivia.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a community acquired pneumonia (CAP) in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of community acquired pneumonia.

In some embodiment, the community acquired pneumonia is due to Streptococcus pneumoniae (penicillin susceptible and resistant isolates) including cases with concurrent bacteremia, Haemophilus influenzae (including beta-lactamase positive isolates), Moraxella catarrhalis, or atypical bacteria like Mycoplasma spp.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a complicated urinary tract infection (cUTI) in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a complicated urinary tract infection.

In some embodiment, the complicated urinary tract infection is selected from pyelonephritis due to Escherichia coli, concurrent bacteremia, or Klebsiella pneumoniae.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of an acute pelvic infection in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of an acute pelvic infection.

In some embodiments, the acute pelvic infection is selected from postpartum endomyometritis, septic abortion and post-surgical gynecologic infections and the infection is due to a microorganism selected from Streptococcus agalactiae, Escherichia coli, Bacteroides fragilis, Porphyromonas asaccharolytica, Peptostreptococcus spp., and Prevotella bivia.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a hospital acquired pneumonia (HAP)/ventilator associated pneumonia (VAP) in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of hospital acquired pneumonia/ventilator associated pneumonia.

In some embodiments, the hospital acquired pneumonia/ventilator associated pneumonia is due to a microorganism selected from Streptococcus pneumoniae (penicillin susceptible and resistant isolates), Staphylococcus aureus (methicillin susceptible and resistant isolates), Klebsiella pneumoniae, Pseudomonas aeruginosa, Acinetobacter spp., Stenotrophomonas maltophilia, Haemophilus influenzae (including beta-lactamase positive isolates), and Legionella pneumophilia.

The compounds or tautomers or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure may also be useful for the prevention, prophylaxis, or reduction of surgical site infections. In some embodiments, the compounds or tautomers or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure are useful following elective colorectal surgery.

Appropriate specimens for bacteriological examination should be obtained in order to isolate and identify the causative organisms and to determine their susceptibility to the compounds of the present disclosure. Therapy with the compounds or tautomers or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure may be initiated empirically before results of these tests are known; once results become available, antimicrobial therapy should be adjusted accordingly.

To reduce the development of drug-resistant bacteria and maintain the effectiveness of the compounds or tautomers or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure and other antibacterial drugs, the compounds or tautomers or pharmaceutically acceptable salts of the compounds or tautomers should be used only to treat or prevent infections that are proven or strongly suspected to be caused by susceptible bacteria. When culture and susceptibility information are available, they should be considered in selecting or modifying antibacterial therapy. In the absence of such data, local epidemiology and susceptibility patterns may contribute to the empiric selection of therapy.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection due to an aerobic or facultative gram-positive microorganism in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a microbial infection due to an aerobic or facultative gram-positive microorganism.

In some embodiments, the aerobic or facultative gram-positive microorganism is selected from: Staphylococcus aureus (methicillin susceptible and resistant isolates), Streptococcus pneumoniae (penicillin susceptible and resistant isolates), Enterococcus spp. (vancomycin susceptible and resistant isolates), Streptococcus agalactiae, Streptococcus pyogenes, and Staphylococcus epidermidis (methicillin susceptible and resistant isolates).

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection due to an aerobic and facultative gram-negative microorganism in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a microbial infection due to an aerobic or facultative gram-positive microorganism.

In some embodiments, the aerobic and facultative gram-negative microorganism is selected from: Escherichia coli [including extended spectrum beta-lactamase (ESBL) and Klebsiella pneumoniae (KPC) producing isolates), Haemophilus influenzae (including Beta-lactamase positive isolates), Klebsiella pneumoniae (including ESBL and KPC producing isolates), Citrobacter freundii, Enterobacter aerogenes, Enterobacter cloacae, Morganella morganii, Salmonella enteritidis, Serratia marcescens, Pseudomonas aeruginosa, Acinetobacter baumanni, Moraxella catarrhalis, Proteus mirabilis, Citrobacter koseri, Haemophilus parainfluenzae, Klebsiella oxytoca (including ESBL and KPC producing isolates), Proteus vulgaris, Providencia rettgeri, and Providencia stuartii.

In some embodiments, the present disclosure relates to a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection due to an anaerobic microorganism in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a microbial infection due to an anaerobic microorganism.

In some embodiments, the anaerobic microorganism is selected from: Bacteroides fragilis, Bacteroides distasonis, Bacteroides ovatus, Bacteroides thetaiotaomicron, Bacteroides uniformis, Clostridioides spp. (e.g., Clostridioides difficile and Clostridioides mangenotii), Clostridium clostridioforme, Eubacterium lentum, Peptostreptococcus species, Porphyromonas asaccharolytica, Prevotella bivia, Bacteroides vulgates, Clostridium perfringens, and Fusobacterium spp.

In some embodiments, the present disclosure relates to a method of treating or reducing the risk of a microbial infection in a human or animal comprising administering to the human or animal an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, or to the use of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a microbial infection.

In some embodiments, the microorganism is Legionella pneumophilia.

In some embodiments, the microorganism Enterococcus spp. is selected from vancomycin susceptible isolate and vancomycin resistant isolate. In some embodiments, the microorganism Escherichia coli is selected from extended spectrum beta-lactamase (ESBL) producing isolate and Klebsiella pneumoniae carbapenemase (KPC) producing isolate. In some embodiments, the microorganism Haemophilus influenzae is a beta-lactamase positive isolate. In some embodiments, the microorganism Klebsiella pneumoniae is selected from extended spectrum beta-lactamase (ESBL) producing isolate and Klebsiella pneumoniae carbapenemase (KPC) producing isolate. In some embodiments, the microorganism Klebsiella oxytoca selected from extended spectrum beta-lactamase (ESBL) producing isolate and Klebsiella pneumoniae carbapenemase (KPC) producing isolate. In some embodiments, the microorganism Staphylococcus aureus is selected from methicillin susceptible isolate and methicillin resistant isolate. In some embodiments, the microorganism Staphylococcus epidermidis is selected from methicillin susceptible isolate and methicillin resistant isolate. In some embodiments, the microorganism Streptococcus pneumoniae is selected from penicillin susceptible isolate and penicillin resistant isolate.

In some embodiments, the microorganism is colistin-resistant. For example, a microorganism that is colistin-resistant exhibits a minimum inhibitory concentration (MIC) for colistin of >2 g/ml). In some embodiments, the microorganism is be a gram negative bacteria such as a Pseudomonas (e.g., Pseudomonas aeruginosa), Escherichia (Escherichia coli), Acinetobacter (e.g., Acinetobacter baumannii), or Klebsiella (e.g., Klebsiella pneumoniae) species that is resistant to treatment with the antibacterial agent known as colistin (polymyxin E). For example, the colistin-resistant microorganism is selected from Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii. In some embodiments, the colistin-resistant microorganism is a Stenotrophomonas, Burkholderia, Proteus, Serratia, Morganella, or Providencia species (e.g., the specific species provided herein).

In some embodiments, the microorganism is ceftazidime-resistant. For example, a microorganism that is ceftazidime-resistant exhibits a minimum inhibitory concentration (MIC) for ceftazidime of >2 g/ml). In some embodiments, the microorganism is be a gram negative bacteria such as a Pseudomonas (e.g., Pseudomonas aeruginosa), Escherichia (Escherichia col), or Klebsiella (e.g., Klebsiella pneumoniae) species that is resistant to treatment with the antibacterial agent known as ceftazidime (Fortraz). For example, the ceftazidime-resistant microorganism is selected from Pseudomonas aeruginosa, Klebsiella pneumoniae, and Escherichia coli. In some embodiments, the microorganism is gentamicin-resistant. For example, a microorganism that is gentamicin-resistant exhibits a minimum inhibitory concentration (MIC) for gentamicin of >2 g/ml). In some embodiments, the microorganism is selected from Pseudomonas (e.g., Pseudomonas aeruginosa), Escherichia (Escherichia coli), Acinetobacter (e.g., Acinetobacter baumannii or Acinetobacter calcoaceticus var. anitratum), Proteus (e.g., Proteus mirabilis or Proteus vulgaris), Enterobacter (Enterobacter aerogenes or Enterobacter cloacae), Staphylococcus (e.g., Staphylococcus aureus or Staphylococcus epidermidis) or Klebsiella (e.g., Klebsiella pneumoniae) species that is resistant to treatment with the antibacterial agent known as gentamicin (Garamycin). In some embodiments, the microorganism is a gram negative bacteria species that is resistant to treatment with the antibacterial agent known as gentamicin.

In some embodiments, the microorganism is levofloxacin-resistant. For example, a microorganism that is levofloxacin-resistant exhibits a minimum inhibitory concentration (MIC) for colistin of >2 g/ml). In some embodiments, the microorganism is selected from Escherichia (Escherichia coli), or Streptococcus (Streptococcus pneumoniae, Streptococcus agalactiae, or Streptococcus pyogenes) species that is resistant to treatment with the antibacterial agent known as levofloxacin (Levaquin). In some embodiments, the microorganism is a gram negative bacteria that is resistant to treatment with the antibacterial agent known as levofloxacin (Levaquin).

In some embodiments, the microorganism is carbapenem-resistant. For example, a microorganism that is carbapenem-resistant exhibits a minimum inhibitory concentration (MIC) for carbapenem of >2 g/ml). In some embodiments, the microorganism is be a selected from a Escherichia (Escherichia col), Enterobacter (Enterobacter aerogenes or Enterobacter cloacae), or Klebsiella (e.g., Klebsiella pneumoniae) species that is resistant to treatment with the antibacterial agent from the class known as carbapenems.

For example, the carbapemen-resistant microorganism is selected from Escherichia coli, Enterobacter aerogenes, Enterobacter cloacae complex, Klebsiella pneumoniae, or Klebsiella oxytoca that is resistant to treatment with the antibacterial agent from the class known as carbapenems. In some embodiments, the microorganism is a gram negative bacteria that is resistant to treatment with the antibacterial agent from the class known as carbapenems.

In some embodiments, a method or use disclosed herein is a method or use to treat a subject that would be subjected to a surgical or invasive medical procedure. Such a subject can be considered to be in need of the methods of treating, reducing the risk of or preventing the infection due to a surgical procedure or an invasive medical procedure. Such a subject can also be considered to be in need of peri-operative prophylaxis.

In some embodiments, a method or use provided herein is a method for treating sepsis in a subject comprising administering to the subject a therapeutically effective amount of a compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound of tautomer thereof. In some such embodiments, the patient is a pediatric patient, a geriatric patient, or a patient having a weakened immune system related to another disease or disorder (e.g., cancer, diabetes, major trauma, or burns). In some embodiments, the sepsis is severe sepsis. In some embodiments, the sepsis is septic shock. In some embodiments, the treatment of sepsis further comprises administration to the subject one or more of intravenous fluids, compounds capable of raising blood pressure, mechanical ventilation, and dialysis.

In some embodiments, the present disclosure provides a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal, the method including administering to the human or animal in need thereof an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer. In some embodiments, the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are extremely-drug resistant Gram-positive or Gram-negative pathogens.

In some embodiments, provided is the use of one or more compounds disclosed herein, including stereoisomers, tautomers, and salts thereof, in the manufacture of a medicament for treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal. In some embodiments, the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are extremely-drug resistant Gram-positive or Gram-negative pathogens.

In some embodiments, provided are one or more compounds disclosed herein, including stereoisomers, tautomers, and salts thereof, for use in treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a human or animal. In some embodiments, the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are extremely-drug resistant Gram-positive or Gram-negative pathogens.

In one embodiment, provided is a method of treating a microbial infection in a subject, that includes administering to the subject an effective amount of one or more compounds of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, where the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are extremely-drug resistant Gram-positive or Gram-negative pathogens.

In one embodiment, provided is a method of preventing a microbial infection in a subject, that includes administering to the subject an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, where the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are extremely-drug resistant Gram-positive or Gram-negative pathogens.

In one embodiment, provided is a method of reducing the risk of a microbial infection in a subject, that includes administering to the subject an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, where the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are extremely-drug resistant Gram-positive or Gram-negative pathogens.

In one embodiment, provided is a method of delaying the onset of a microbial infection in a subject, that includes administering to the subject an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, where the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are capable of being used as biological weapons. In some embodiments, the infection is caused by or involves one or more microorganisms which are extremely-drug resistant Gram-positive or Gram-negative pathogens.

In some embodiments, a bacterium which can be used as a biological weapon possesses one or more characteristics that include, but are not limited to, being easily being produced or disseminated, being easily transmitted from person to person, having the potential for moderate or high morbidity, having the potential for moderate or high mortality, having the potential for causing public panic and social disruption, requiring special action for public health preparedness, and requiring specific enhancements for diagnosis and disease surveillance.

In another embodiment, a bacterium which can be used as a biological weapon is stable or viable, for example, the bacterium is capable of performing all or part of its normal biological functions, such as replicating, forming spores, and infecting a subject, under various conditions. In some embodiments, the bacterium is stable or viable in one or more conditions that include, but are not limited to, heat, cold, high pressure, low pressure, acidic or basic conditions, humidity, dryness, and radiation, including extreme conditions.

In one embodiment, a bacterium which can be used as a biological weapon is stable or viable at a temperature above about 25° C., such as above about 30° C., about 40° C., about 50° C., about 60° C., about 70° C., about 80° C., about 90° C., about 100° C., about 125° C., about 150° C., about 175° C., or above about 200° C. In another embodiment, a bacterium which can be used as a biological weapon is stable or viable at a temperature below about 25° C., such as below about 20° C., about 10° C., about 5° C., about 0° C., about −10° C., about −20° C., about −30° C., about −40° C., about −50° C., about −60° C., about −70° C., about −100° C., or below about −150° C.

In one embodiment, a bacterium which can be used as a biological weapon is capable of infecting a subject under various conditions, such as various pressures. In one embodiment, a bacterium which can be used as a biological weapon is stable or viable under pressure above about 5×105 Pa, such as above about 10×105 Pa, about 15×105 Pa, about 20×105 Pa, about 30×105 Pa, about 40×105 Pa, about 50×105 Pa, about 75×105 Pa, or about 100×105 Pa. In another embodiment, a bacterium which can be used as a biological weapon is stable or viable under pressure below about 0.5×105 Pa, such as below about 0.2×105 Pa, about 0.1×105 Pa, about 0.05×105 Pa, about 0.02×105 Pa, about 0.01×105 Pa, about 0.005×105 Pa, about 0.002×105 Pa, or about 0.001×105 Pa.

In one embodiment, a bacterium which can be used as a biological weapon is stable or viable at a pH above about 8.0, such as above about 8.5, about 9.0, about 9.5, about 10.0, about 10.5, about 11.0, about 11.5, about 12.0, about 12.5, about 13.0, about 13.5, or about 14.0. In another embodiment, a bacterium which can be used as a biological weapon is stable or viable at a pH below about 6.0, such as below about 5.5, about 5.0, about 4.5, about 4.0, about 3.5, about 3.0, about 2.5, about 2.0, about 1.5, about 1.0, about 0.5, or about 0.0.

In one embodiment, a bacterium which can be used as a biological weapon is stable or viable under a relative humidity of about 10%, such as about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 99%.

In another embodiment, a bacterium which can be used as a biological weapon is stable or viable under UV radiation, X-ray radiation, a radiation, p radiation, or y radiation.

In one embodiment, a bacterium which can be used as a biological weapon is able to form spores.

In some embodiments, a bacterium which can be used as a biological weapon can be dispersed in air or in liquid. In one embodiment, the bacterium is in aeorosol form, for example, the bacterium is formulated as an aerosol. In another embodiment, the bacterium is in powder form, for example, the bacterium is formulated as powder.

In one embodiment, a bacterium which can be used as a biological weapon includes a bacterium which is resistant to existing antibiotics. In some embodiments, the bacterium is resistant to tetracycline antibiotics, including, but not limited to, tetracycline, doxycycline, minocycline, sancycline, methacycline, chlortetracycline, and deoxytetracycline, and a combination thereof. In some embodiments, the bacterium is resistant to other antibiotics, including, but not limited to, aminoglycosides, such as gentamicin and kanamycin, colistin, methicillin, oxacillin, vancomycin, penicillin, linezolid, fluoroquinolones, such as ciprofloxacin, ceftazidime, and macrolides, such as azithromycin. In some embodiments, a bacterium which can be used as a biological weapon includes a bacterium which is resistant to gentamicin. In some embodiments, a bacterium which can be used as a biological weapon includes a bacterium which is resistant to colistin. In some embodiments, a bacterium which can be used as a biological weapon includes a bacterium which is resistant to gentamicin and colistin.

In some embodiments of the disclosed methods, the one or more microorganisms are biodefense category A or biodefense category B pathogens. Biodefense category A pathogens are those organisms or biological agents that pose the highest risk to national security and public health because they (1) can be easily disseminated or transmitted from person to person, (2) result in high mortality rates and have the potential for major public health impact, (3) might cause public panic and social disruption, and (4) require special action for public health preparedness. Examples of category A pathogens include, but are not limited to, Bacillus anthracis (anthrax), Francisella tularensis (tularemia), Yersinia pestis (plague), Ebola, Marburg, Ebola-like viruses such as Bundibugyo ebolavirus, Sudan ebolavirus, TaiForest ebolavirus, Zaire ebolavirus and Marburg-like viruses such as Marburg virus and Ravn virus. In some embodiments, the one or more microorganisms are selected from the group consisting of biodefense category A pathogens Bacillus anthracis (anthrax), Yersinia pestis (plague), and Francisella tularensis (tularemia).

Biodefense category B pathogens are the second highest priority organisms or biological agents. They are moderately easy to disseminate, result in moderate morbidity rates and low mortality rates, and require specific enhancements for diagnostic capacity and enhanced disease surveillance. Examples of category B pathogens include, but are not limited to, Burkholderia pseudomallei (melioidosis), Coxiella burnetii (Q fever), Brucella species (brucellosis), Burkhoderia mallei (glanders), Chlamydia psittaci (psittacosis), Rickettsia prowazekii (typhus fever), diarrheagenic E. coli, pathogenic Vibrios, Shigella species, Salmonella, Listeria monocytogenes, Campylobacter jejuni, Yersinia enterocolitica, Staphylococcus enterotoxin B, and Hepatitis A. In some embodiments, the one or more microorganisms are selected from the group consisting of biodefense category B pathogens Burkholderia pseudomallei (melioidosis), Coxiella burnetii (Q fever), Brucella species (brucellosis), Burkhoderia mallei (glanders), Chlamydia psittaci (psittacosis), Rickettsia prowazekii (typhus fever), diarrheagenic E. coli, pathogenic Vibrios, Shigella species, Salmonella, Listeria monocytogenes, Campylobacter jejuni, and Yersinia enterocolitica.

More examples of category A or B pathogens are provided by the National Institute of Allergy and Infectious Diseases (NIAID) at http://www.niai d.nih.gov/topics/bi odefenserelated/bi odefense/pages/cata.aspx#, the contents of which are hereby incorporated by reference in its entirety.

In some embodiments, a bacterium which can be used as a biological weapon includes, but is not limited to, a bacterium of the Bacillus cereus group. The Bacillus cereus group of bacteria includes Bacillus anthracis (the etiologic agent of anthrax), Bacillus cereus, Bacillus weihenstephanensis (a food borne pathogen), Bacillus thuringiensis (an insect pathogen), and Bacillus mycoides. In some embodiments, the bacterium is selected from Bacillus anthracis, multidrug-resistant (MDR) anthrax, Francisella tularensis, Clostridium botulinum, Yersinia pestis, Burkholderia mallei, Burkholderia pseudomallei, Brucella species, Shigella species, Coxiella burnetii, Chlamydia psittaci, Clostridium perfringens, Rickettsia prowazekii, diarrheagenic E. coli, pathogenic Vibrios, Salmonella, Campylobacter jejuni, Yersinia enterocolitica, and Listeria monocytogenes. In some embodiments, the microorganism (bacterium) is selected from Bacillus anthracis, Franciscella tularensis, Yersinia pestis, Burkholderia mallei, and Burkholderia pseudomallei. In some embodiments, the microorganism (bacterium) is selected from Burkholderia mallei and Burkholderia pseudomallei. In some embodiments, the microorganism (bacterium) is Burkholderia pseudomallei.

In some embodiments, a bacterium which can be used as a biological weapon is Bacillus anthracis or multidrug-resistant (MDR) anthrax.

In some embodiments, a bacterium which can be used as a biological weapon is Burkholderia pseudomallei.

In some embodiments, a bacterium which can be used as a biological weapon includes, but is not limited to, gram-positive pathogens, gram-negative pathogens, anaerobic pathogens, or atypical pathogens, or a combination thereof. In some embodiments, the bacterium includes methicillin-susceptible Staphylococcus aureus (MSSA), methicillin-resistant Staphylococcus aureus (MRSA), oxacillin-susceptible Staphylococcus aureus, oxacillin-resistant Staphylococcus aureus, oxacillin-resistant coagulase-negative Staphylococcus, Enterococcus faecalis, Enterococcus faecium, vancomycin-susceptible Enterococcus faecium, vancomycin-resistant Enterococcus faecium, vancomycin-susceptible Enterococcus faecalis, vancomycin-resistant Enterococcus faecalis, Streptococcus pneumoniae, penicillin-susceptible Streptococcus pneumonia, penicillin-resistant Streptococcus pneumoniae (PRSP), Streptococcus pyogenes, Streptococcus agalactiae, Haemophilus influenzae, Moraxella catarrhalis, Neisseria gonorrhoeae, Escherichia coli, Shigella spp., Salmonella spp., Klebsiella pneumoniae, Enterobacter aerogenes, Enterobacter cloacae, Serratia marcescens, Acinetobacter baumannii, Stenotrophomonas maltophilia, Bacteroides fragilis, Clostridium perfringens, Chlamydia pneumoniae, Legionella pneumophila, Proteus mirabilis, Pseudomonas aeruginosa, and Burkholderia cepacia.

In some embodiments, the one or more microorganisms are extremely-drug resistant Gram-positive or Gram-negative pathogens.

In some embodiments, provided is a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a subject that is caused by or involves one or more microorganisms which are capable of being used as biological weapons that includes administering a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, the one or more microorganisms are biodefense category A pathogens. In some embodiments, the one or more microorganisms are biodefense category A pathogens selected from Bacillus anthracis (anthrax), Yersiniapestis (plague), and Francisella tularensis (tularemia).

In some embodiments, the one or more microorganisms are biodefense category B pathogens. In some embodiments, the one or more microorganisms are biodefense category B pathogens Burkholderia pseudomallei (melioidosis), Coxiella burnetii (Q fever), Brucella species (brucellosis), Burkhoderia mallei (glanders), Chlamydia psittaci (psittacosis), Rickettsia prowazekii (typhus fever), diarrheagenic E. coli, pathogenic Vibrios, Shigella species, Salmonella, Listeria monocytogenes, Campylobacter jejuni, and Yersinia enterocolitica.

In some embodiments, the one or more microorganisms are selected from Bacillus anthracis, Franciscella tularensis, Yersinia pestis, Burkholderia mallei, and Burkholderia pseudomallei.

In some embodiments, the one or more microorganisms are selected from Burkholderia mallei and Burkholderia pseudomallei. In some embodiments, the one or more microorganisms are Burkholderia pseudomallei.

In some embodiments, provided is a method of treating, preventing, reducing the risk of, or delaying the onset of a microbial infection in a subject that is caused by or involves one or more microorganisms which are extremely-drug resistant Gram-positive or Gram-negative pathogens that includes administering a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, provided is a method of treating a microbial infection in a subject that includes administering a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, after the subject has been exposed to the microorganism, but before the subject develops any symptom of the microbial infection. In some embodiments, the microorganism is a bacterium. In some embodiments, the microbial infection is a bacterial infection. In some embodiments, a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, is administered about 10 min, about 20 min, about 30 min, about 40 min, about 50 min, about 1 hr, about 2 hrs, about 3 hrs, about 6 hrs, about 12 hrs, about 18 hrs, about 24 hrs, about 36 hrs, about 48 hrs, about 72 hrs, about 96 hrs, about 1 week, or about 2 weeks after the subject has been exposed to the microorganism, but before the subject develops any symptoms. In another embodiment, provided is a method of treating a microbial infection in a subject that includes administering a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, after the subject develops a symptom after the subject has been exposed to the microorganism. In some embodiments, the microorganism is a bacterium. In one embodiment, a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, is administered about 10 min, about 20 min, about 30 min, about 40 min, about 50 min, about 1 hr, about 2 hrs, about 3 hrs, about 6 hrs, about 12 hrs, about 18 hrs, about 24 hrs, about 36 hrs, about 48 hrs, about 72 hrs, about 96 hrs, about 1 week, or about 2 weeks after the subject develops a symptom.

In another embodiment, provided is a method of treating a microbial infection in a subject that includes administering a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, after the subject's suspected exposure to the microorganism, but before the subject develops any symptom of the microbial infection. In one embodiment, the compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, is administered about 10 min, about 20 min, about 30 min, about 40 min, about 50 min, about 1 hr, about 2 hrs, about 3 hrs, about 6 hrs, about 12 hrs, about 18 hrs, about 24 hrs, about 36 hrs, about 48 hrs, about 72 hrs, about 96 hrs, about 1 week, or about 2 weeks after the subject's suspected exposure to the microorganism, but before the subject develops any symptoms. In some embodiments, the microorganism is a bacterium.

In some embodiments, provided is a method of preventing a microbial infection in a subject that includes administering a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, before the subject has been exposed to the microorganism. In some embodiments, the microorganism is a bacterium. In some embodiments, the microbial infection is a bacterial infection. In some embodiments, the compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, is administered about 10 min, about 20 min, about 30 min, about 40 min, about 50 min, about 1 hr, about 2 hrs, about 3 hrs, about 6 hrs, about 12 hrs, about 18 hrs, about 24 hrs, about 36 hrs, about 48 hrs, about 72 hrs, about 96 hrs, about 1 week, or about 2 weeks before the subject has been exposed to the microorganism.

In another embodiment, provided is a method of preventing a microbial infection in a subject that includes administering a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, before or after an event which raises the risk of the subject being exposed to the microorganism. In some embodiments, the microorganism is a bacterium. The event includes, but is not limited to, an attack, for example, a terrorist attack, with a biological weapon and the subject's entry into a risky territory, such as a battlefield. In one embodiment, a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, is administered to the subject about 10 min, about 20 min, about 30 min, about 40 min, about 50 min, about 1 hr, about 2 hrs, about 3 hrs, about 6 hrs, about 12 hrs, about 18 hrs, about 24 hrs, about 36 hrs, about 48 hrs, about 72 hrs, about 96 hrs, about 1 week, or about 2 weeks before the event. In another embodiment, a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer is administered to the subject about 10 min, about 20 min, about 30 min, about 40 min, about 50 min, about 1 hr, about 2 hrs, about 3 hrs, about 6 hrs, about 12 hrs, about 18 hrs, about 24 hrs, about 36 hrs, about 48 hrs, about 72 hrs, about 96 hrs, about 1 week, or about 2 weeks after the event.

In another embodiment, the method of the present disclosure includes, before administering a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, identifying a subject at risk of being exposed to a microorganism which can be used as a biological weapon. In some embodiments, the microorganism is a bacterium. In some embodiments, the subject at risk of being exposed to a microorganism which can be used as a biological weapon includes, but is not limited to, a subject travelling to, entering, or being in a conflict region, for example, a battlefield or combat zone, including military personnel, intelligence personnel, and animals used in the military, a subject engaged or about to be engaged in a security operation, such as governmental authorities (for example, police, governmental investigators, and secret service members) and other personnel (for example, doctors, nurses, and rescue workers), and animals used in such an operation, and a subject in an geographical area that can be a target of a terrorist attack, for example, a metropolitan area, a city, an area where there is a large population (for example, above 100,000, above 200,000, above 500,000, above 1 million, above 2 million, above 5 million, or above 10 million), or a location or area to which damage is likely to cause a threat to national security or public health (for example, a nuclear power plant, a chemical plant, an airport, or a hospital).

In some embodiments, provided is a method of treating a bacterial infection in a subject, where the subject is exposed or suspected of being exposed to a bacterium or a component thereof, that includes administering to the subject an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer. In another embodiment, provided is a method a method of preventing a bacterial infection in a subject, where the subject is at a risk of being exposed to a bacterium or a component thereof, that includes administering to the subject an effective amount of a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer. In one embodiment, the bacterium or a component thereof is formulated as an aerosol or power. In another embodiment, the bacterial component is a bacterial spore. In some embodiments, the present disclosure relates to a method, use, or compound disclosed herein, wherein the amount of compound or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer comprises from 0.1 mg to 1500 mg. For example, a dose of active compound can range from about 0.1 mg to about 1250 mg; about 0.1 mg to about 1000 mg; about 0.1 mg to about 800 mg; about 0.1 mg to about 500 mg; about 0.1 mg to about 250 mg; about 0.1 mg to about 100 mg; about 0.1 mg to about 50 mg; about 0.1 mg to about 25 mg; about 0.1 mg to about 20 mg; about 0.1 mg to about 10 mg; about 0.1 mg to about 5 mg; about 0.1 mg to about 1 mg; about 0.1 mg to about 0.5 mg; about 0.5 mg to about 1500 mg; about 1 mg to about 1500 mg; about 2.5 mg to about 1500 mg; about 5 mg to about 1500 mg; about 10 mg to about 1500 mg; about 50 mg to about 1500 mg; about 100 mg to about 1500 mg; about 250 mg to about 1500 mg; about 500 mg to about 1500 mg; about 750 mg to about 1500 mg; about 1000 mg to about 1500 mg; about 1250 mg to about 1500 mg; about 0.25 mg to about 2.5 mg; about 0.5 mg to about 5 mg; about 1 mg to about 10 mg; about 5 to about 20 mg; about 10 mg to about 50 mg; about 25 mg to about 75 mg; about 20 mg to about 100 mg; about 50 mg to about 200 mg; about 100 mg to about 500 mg; about 250 mg to about 750 mg; about 200 mg to about 800 mg; about 500 mg to about 1000 mg; or about 750 mg to about 1250 mg.

In some embodiments, the present disclosure relates to a method, use, or compound disclosed herein wherein the compound, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer, is administered optically, ophthalmically, nasally, orally, parenterally, topically, or intravenously.

In some embodiments, the present disclosure relates to a method of synthesizing a compound disclosed herein, or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer.

In some embodiments, the present disclosure relates to a medical device containing a compound disclosed herein or a tautomer thereof, or a pharmaceutically acceptable salt of the compound or tautomer. In some embodiments, the device is a stent.

3. SYNTHESIS OF THE COMPOUNDS OF THE DISCLOSURE

The compounds of the present disclosure can be synthesized by using art recognized techniques, such as those described in US 2012-0220566, WO 2012/173689, or PCT/US2014/054869, the contents of each of which are incorporated herein by reference in their entireties. The compounds thus obtained can be further purified, for example, by flash column chromatography, high performance liquid chromatography, crystallization, or any known purification method.

In one embodiment, compounds of the present disclosure can be synthesized according to the exemplary method described below.

The specific approaches and compounds shown in the schemes above are not intended to be limiting. The chemical structures in the schemes herein depict variables that are hereby defined commensurately with chemical group definitions (moieties, atoms, etc.) of the corresponding position in the compound formulae herein, whether identified by the same variable name (i.e., R1, R2, R3, etc.) or not. The suitability of a chemical group in a compound structure for use in the synthesis of another compound is within the knowledge of one of ordinary skill in the art.

Additional methods of synthesizing compounds of the formulae herein and their synthetic precursors, including those within routes not explicitly shown in schemes herein, are within the means of chemists of ordinary skill in the art. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the applicable compounds are known in the art and include, for example, those described in Larock R, Comprehensive Organic Transformations, VCH Publishers (1989); Fieser L et al., Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and Paquette L, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995) and subsequent editions thereof.

4. CHARACTERIZATION OF COMPOUNDS OF THE DISCLOSURE

Compounds designed, selected and/or optimized by methods described above, once produced, can be characterized using a variety of assays known to those skilled in the art to determine whether the compounds have biological activity. For example, the molecules can be characterized by conventional assays, including but not limited to those assays described below, to determine whether they have a predicted activity, binding activity and/or binding specificity.

Furthermore, high-throughput screening can be used to speed up analysis using such assays. As a result, it can be possible to rapidly screen the molecules disclosed herein for activity, for example, as anti-cancer, anti-bacterial, anti-fungal, anti-parasitic or anti-viral agents. Also, it can be possible to assay how the compounds interact with a ribosome or ribosomal subunit and/or are effective as modulators (for example, inhibitors) of protein synthesis using techniques known in the art. General methodologies for performing high-throughput screening are described, for example, in Devlin (1998) High Throughput Screening, Marcel Dekker; and U.S. Pat. No. 5,763,263. High-throughput assays can use one or more different assay techniques including, but not limited to, those described below.

(1) Surface Binding Studies. A variety of binding assays can be useful in screening new molecules for their binding activity. One approach includes surface plasmon resonance (SPR) that can be used to evaluate the binding properties of molecules of interest with respect to a ribosome, ribosomal subunit or a fragment thereof.

SPR methodologies measure the interaction between two or more macromolecules in real-time through the generation of a quantum-mechanical surface plasmon. One device, (BIAcore Biosensor RTM from Pharmacia Biosensor, Piscataway, N.J.) provides a focused beam of polychromatic light to the interface between a gold film (provided as a disposable biosensor “chip”) and a buffer compartment that can be regulated by the user. A 100 nm thick “hydrogel” composed of carboxylated dextran that provides a matrix for the covalent immobilization of analytes of interest is attached to the gold film. When the focused light interacts with the free electron cloud of the gold film, plasmon resonance is enhanced. The resulting reflected light is spectrally depleted in wavelengths that optimally evolved the resonance. By separating the reflected polychromatic light into its component wavelengths (by means of a prism), and determining the frequencies that are depleted, the BIAcore establishes an optical interface which accurately reports the behavior of the generated surface plasmon resonance. When designed as above, the plasmon resonance (and thus the depletion spectrum) is sensitive to mass in the evanescent field (which corresponds roughly to the thickness of the hydrogel). If one component of an interacting pair is immobilized to the hydrogel, and the interacting partner is provided through the buffer compartment, the interaction between the two components can be measured in real time based on the accumulation of mass in the evanescent field and its corresponding effects of the plasmon resonance as measured by the depletion spectrum. This system permits rapid and sensitive real-time measurement of the molecular interactions without the need to label either component.

(2) Fluorescence Polarization. Fluorescence polarization (FP) is a measurement technique that can readily be applied to protein-protein, protein-ligand, or RNA-ligand interactions in order to derive IC50s and Kds of the association reaction between two molecules. In this technique one of the molecules of interest is conjugated with a fluorophore. This is generally the smaller molecule in the system (in this case, the compound of interest). The sample mixture, containing both the ligand-probe conjugate and the ribosome, ribosomal subunit or fragment thereof, is excited with vertically polarized light. Light is absorbed by the probe fluorophores, and re-emitted a short time later. The degree of polarization of the emitted light is measured. Polarization of the emitted light is dependent on several factors, but most importantly on viscosity of the solution and on the apparent molecular weight of the fluorophore. With proper controls, changes in the degree of polarization of the emitted light depends only on changes in the apparent molecular weight of the fluorophore, which in-turn depends on whether the probe-ligand conjugate is free in solution, or is bound to a receptor. Binding assays based on FP have a number of important advantages, including the measurement of IC50s and Kds under true homogenous equilibrium conditions, speed of analysis and amenity to automation, and ability to screen in cloudy suspensions and colored solutions.

(3) Protein Synthesis. It is contemplated that, in addition to characterization by the foregoing biochemical assays, the compound of interest can also be characterized as a modulator (for example, an inhibitor of protein synthesis) of the functional activity of the ribosome or ribosomal subunit.

Furthermore, more specific protein synthesis inhibition assays can be performed by administering the compound to a whole organism, tissue, organ, organelle, cell, a cellular or subcellular extract, or a purified ribosome preparation and observing its pharmacological and inhibitory properties by determining, for example, its inhibition constant (IC50) for inhibiting protein synthesis. Incorporation of 3H leucine or 35S methionine, or similar experiments can be performed to investigate protein synthesis activity. A change in the amount or the rate of protein synthesis in the cell in the presence of a molecule of interest indicates that the molecule is a modulator of protein synthesis. A decrease in the rate or the amount of protein synthesis indicates that the molecule is an inhibitor of protein synthesis.

(4) Antimicrobial assays and other evaluation. Furthermore, the compounds can be assayed for anti-proliferative or anti-infective properties on a cellular level. For example, where the target organism is a microorganism, the activity of compounds of interest can be assayed by growing the microorganisms of interest in media either containing or lacking the compound. Growth inhibition can be indicative that the molecule can be acting as a protein synthesis inhibitor. More specifically, the activity of the compounds of interest against bacterial pathogens can be demonstrated by the ability of the compound to inhibit growth of defined strains of human pathogens. For this purpose, a panel of bacterial strains can be assembled to include a variety of target pathogenic species, some containing resistance mechanisms that have been characterized. Use of such a panel of organisms permits the determination of structure-activity relationships not only in regards to potency and spectrum, but also with a view to obviating resistance mechanisms.

(5) The translation-only assay for ribosomal protein production uses purified 70S ribosomes, corresponding S100 extracts containing the biological molecules necessary to support protein translation, and mRNA encoding firefly luciferase or another protein reporter. The resulting luminescence signal is proportional to protein translation and is determined by a luminescence assay plate reader (i.e. Victor2V Multilabel Reader). This assay is performed with varying concentrations of potential translation inhibitors in the assay. The resulting data are used to calculate IC50 values of inhibition for the compounds using appropriate software (i.e. MDL Assay Explorer with a one-site competition model of binding).

The in vitro activity of the compounds of the present disclosure can be determined. Antimicrobial testing is typically performed to determine the minimum inhibitory concentration (MIC). Minimum inhibitory concentrations (MICs) are determined by the microdilution method in a final volume of 100 μl according to protocols outlined by The Clinical and Laboratory Standards Institute (CLSI). Performance standards for reference strains are assessed within the same experimental design to maintain quality control. See, for example, Clinical Laboratory Standards Institute: Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically M7-A8. Approved Standard-Eighth Edition. Wayne, Pa.: CLSI; December 2008; and Clinical Laboratory Standards Institute: Performance Standards for Antimicrobial Susceptibility Testing M100-S20; Approved Standard-Twentieth Edition. Wayne, Pa.: CLSI; June 2010.

For example, an agar-dilution MIC assay could be run using the following protocol. Pure cultures of isolates to be tested are grown on Chocolate Agar at 35° C. to 36.5° C. in a CO2 enriched (5%) atmosphere for 16-18 hours. Using a cotton applicator or a bacteriologic loop, isolated colonies (or cells from less dense areas of growth on the plate) are suspended in 5 mL saline. The density of the suspension is then adjusted to contain 108 colony forming units (CFU)/ml by comparison with a 0.5 McFarland BaSO4 turbidity standard. This suspension is then diluted in 1:10 in MH broth to give 107 CFU/ml. Using a multichannel pipettor, 0.002 mL spots of the bacterial suspension is dispensed onto the surface of the medium, i.e., 104 CFU. Each plate of the set of antibiotic containing media plus a plate of Chocolate Agar or GCS medium (as a control to determine that all isolates grew) is inoculated. The inoculated plates are air-dried at room temperature for approximately 15 minutes. The plates are then inverted and incubated at 35° C. to 36.5° C. in a CO2-enriched (5%) atmosphere for 24 hours. The plates are then examined for growth.

Another in vitro assay that can be performed is a time-kill kinetic assay. Using this assay, bactericidal activity can be determined by time-kill methodology as described by Clinical Laboratory Standards Institute. For example, the compounds to be tested are added to test flasks at concentrations of 2×-32× the MIC (determined, for example, using the assays described herein). Once dissolved, compounds are diluted in Giolitti Cantoni (GC) broth to a volume of 1 mL at the 25× desired final concentration; a flask containing 1 mL of GC broth without compound is prepared as a growth control. A 0.5 McFarland equivalent is prepared for the test organism, diluted 1:200 in pre-warmed GC broth, and incubated in 5% CO2-enriched atmosphere at 35° C. for 30 minutes prior to exposure to the test compound. After the 30-minute pre-incubation, 24 mL is removed and added to each test flask for a final volume of 25 mL. A sample is removed from the growth control flask, diluted in Phosphate Buffered Saline (PBS) and plated on Chocolate Agar (CA) to confirm an inoculum of approximately 5×105 CFU/mL. Samples are then removed from all flasks at 1, 2, 4, 6, 8, and 24 hours, diluted in PBS and plated on CA to determine the number of viable cells in each flask. Plate counts are incubated at 35° C. in 5% CO2-enriched atmosphere for 48 hours and colonies are counted. Plate counts are then graphed.

The antimicrobial and other drug properties of the compounds can further be evaluated in various in vivo mammalian assays, such as a mouse or rat peritonitis infectious models, skin and soft tissue models (often referred to as the thigh model), or a mouse pneumonia model. There are septicemia or organ infection models known to those skilled in the art. These efficacy models can be used as part of the evaluation process and can be used as a guide of potential efficacy in humans. Endpoints can vary from reduction in bacterial burden to lethality. For the latter endpoint, results are often expressed as a PD50 value, or the dose of drug that protects 50% of the animals from mortality.

To further assess a compound's drug-like properties, measurements of inhibition of cytochrome P450 enzymes and phase II metabolizing enzyme activity can also be measured either using recombinant human enzyme systems or more complex systems like human liver microsomes. Further, compounds can be assessed as substrates of these metabolic enzyme activities as well. These activities are useful in determining the potential of a compound to cause drug-drug interactions or generate metabolites that retain or have no useful antimicrobial activity.

To get an estimate of the potential of the compound to be orally bioavailable, one can also perform solubility and Caco-2 assays. The latter is a cell line from human epithelium that allows measurement of drug uptake and passage through a Caco-2 cell monolayer often growing within wells of a 24-well microtiter plate equipped with a 1 micron membrane. Free drug concentrations can be measured on the basolateral side of the monolayer, assessing the amount of drug that can pass through the intestinal monolayer. Appropriate controls to ensure monolayer integrity and tightness of gap junctions are needed. Using this same system one can get an estimate of P-glycoprotein mediated efflux. P-glycoprotein is a pump that localizes to the apical membrane of cells, forming polarized monolayers. This pump can abrogate the active or passive uptake across the Caco-2 cell membrane, resulting in less drug passing through the intestinal epithelial layer. These results are often done in conjunction with solubility measurements and both of these factors are known to contribute to oral bioavailability in mammals. Measurements of oral bioavailability in animals and ultimately in man using traditional pharmacokinetic experiments will determine the absolute oral bioavailability.

Experimental results can also be used to build models that help predict physical-chemical parameters that contribute to drug-like properties. When such a model is verified, experimental methodology can be reduced, with increased reliance on the model predictability.

(5) Animal Pharmacology and Toxicology. The compounds of the present disclosure can be evaluated for efficacy in well-known animal models. The following table provides representative animal models for various infection indications.

Target Infection Indication Animal Model of Efficacy HAP/VAP Efficacy in mouse and/or rat pneumoniae model vs. respiratory tract infection pathogens of interest (Streptococcus pneumoniae, including multi-drug resistant Streptococcus pneumoniae, H. influenzae, methicillin resistant Staphylococcus aureus (MRSA), and Pseudomonas. aeruginosa) cSSSI Efficacy in mouse model against pathogens of interest (MRSA, K. pneumoniae) Sepsis Efficacy in mouse peritonitis model vs. pathogens of interest (E. coli, K. pneumoniae, E. faecalis, MRSA) cUTI Efficacy in mouse model against E. coli, K. pneumoniae and/or MRSA) Febrile neutropenia Efficacy in mouse peritonitis model against S. aureus, S. epidermidis, S. pneumoniae, S. pyogenes, P. aeruginosa

Animal Model for Complicated Skin and Skin Structure Infections (cSSSI): Murine Skin and Soft Tissue Infection Model of Klebsiella pneumoniae 1705966 in Thighs of Neutropenic Female CD-1 Mice

This model is useful to assess the efficacy of compounds of the present disclosure in a Klebsiella pneumoniae 1705966 neutropenic mouse thigh infection model using female ICR (CD-1) mice.

Study Design:

Species: Female ICR (CD-1) Mice, 8 to 9 weeks old, weighting 25-29 g.

Inoculum: Klebsiella pneumoniae 17059663 was streaked from frozen stock onto Blood agar (Tryptic Soy Agar+5% Sheep Blood), BD, #221261) and incubated overnight at 35° C. After overnight incubation, enough bacteria (approx. 1 full loop) to measure OD625=0.990 was transferred from plate and diluted into 10 ml pre-warmed Mueller-Hinton broth. This culture was further diluted 1:1000 into pre-warmed MH broth and grown for approximately 2 hours at 35° C. with shaking. Each mouse was given 0.1 mL of 1:1000 dilution culture injected into both caudal thigh muscles under isoflurane inhalation anesthesia.

Dilution Initial O.D. Final O.D. (after~2 hr. incubation) 1:10 0.135 0.424 1:100 0.014 0.215 1:1000 0.001 0.035

Neutropenia is induced by intraperitoneal (I.P.) administration of Cyclophosphamide monohydrate on Day −4 (150 mg/kg) and Day −1 (100 mg/kg).

Vehicle: 0.9% sodium chloride

Dosing: Each mouse in the treated groups was given the appropriate dose of the compound to be tested in a volume of 0.2 ml, 2 and 8 hrs. post bacterial inoculation.

Time points:

Controls: 0, 2, 6, and 24 hrs.

Treated: 24 hrs.

Sampling: 2 or 3 mice/time point were euthanized via CO2, and their caudal thigh muscles excised and homogenized. The thigh muscles were placed in 5 ml sterile PBS in Stomacher Filter bag and homogenized with MicroBiomaster80 (Brinkmann) for 60 seconds, normal setting and 1:10 dilutions were made per standard protocol in a 96-well plate. Aliquots of 25 ul for each dilution, as well as the homogenate, were plated on blood agar plates and incubated at 35° C. to determine the CFU/mL over the time course. After overnight incubation, colonies were counted.

Animal Model for Sepsis:

Murine Peritonitis Model (E. coli, K. Pneumoniae, E. Faecalis, MRSA)

This model is used to evaluate the effect of subcutaneous (SC) treatment with compounds of the present disclosure on growth of Escherichia coli ATCC 25922 in a mouse peritonitis model using female Swiss Webster mice.

Controls:

Negative: Inoculum only

Inoculum Vehicle Intraperitoneal

Positive: Ciprofloxacin

Study Design:

Species: Female Swiss Webster Mice

Inoculation: Escherichia coli ATCC 25922 is made by adding 1 ml (Apr. 6, 2007) stock to 9 ml 0.25% Brewer's Yeast to make (1:10), then 1 ml of the (1:10) will be added to 9 ml 0.25% Brewer's Yeast to make (1:100), then 1 ml of the (1:100) will be added to 9 ml 0.25% Brewer's Yeast to make (1:1000), then 2.5 ml of the (1:1000) will be added to 122.5 ml 0.25% Brewer's Yeast to make (1:50,000), 1 ml/mouse will be inoculated intraperitoneally (IP).

Route of Administration: SC

Dosing: Vehicle for compounds of the present disclosure: Saline or 50 mM Sodium phosphate buffer in 10% Captisol in water, pH=7.2.

Dose Administration: Q3H×3 beginning at 30 min post bacterial inoculation

Study Duration: 24 hrs. 0.25% Brewer's Yeast Extract (BYE): Dilute 2% prepared on 11/12/09 (Lot.2158K, MP Biomedicals) 25 ml 2%+175 ml 1× PBS.

Outcome Measures: Colony Forming Unit's from peritoneal wash and spleen homogenate and drug levels from wash, spleen homogenate, and plasma.

Blood is collected via cardiac puncture while mouse is under CO2 narcosis. The whole blood sample is placed in heparinized eppendorf tubes and kept on wet ice until centrifuged (4 min @ 14,000 rpm). Plasma is transferred to 96 deep-well block on dry ice and stored at −20° C. Immediately following blood collection, 2 ml of sterile PBS (phosphate buffered saline) was injected into the peritoneal cavity with a 25G needle. The abdomen was gently massaged, and a small incision was made to allow access to the peritoneal cavity. The peritoneal wash fluid was collected using sterile technique, serially diluted 1:10, plated on blood agar plates, and incubated overnight at 35° C.

Spleens were harvested and placed in 1 ml sterile PBS in Stomacher bag and homogenized with MicroBiomaster80 (Brinkmann) for 60 seconds, normal setting and 1:10 dilutions were made. 25 μl of each dilution, as well as the homogenate, was plated on blood agar plates and incubated at 35° C. to determine the CFU/mL over the time course. After overnight incubation, colonies were counted.

Other Animal Models

Similarly, other animal infection models can be used for hospital acquired pneumonia (HAP)/ventilator acquired pneumonia (VAP), complicated urinary tract infections (cUTI), and febrile neutropenia.

5. FORMULATION AND ADMINISTRATION

The compositions and methods of the present disclosure can be practiced by delivering the compounds of the present disclosure using a means for delivery e.g., any suitable carrier. The dose of active compound, mode of administration and use of suitable carrier will depend upon the intended patient or subject and the targeted microorganism, e.g., the target bacterial organism. The formulations, both for human medical use and veterinary use, of compounds according to the present disclosure typically include such compounds in association with a pharmaceutically acceptable carrier.

The carrier(s) should be “acceptable” in the sense of being compatible with compounds of the present disclosure and not deleterious to the recipient. Pharmaceutically acceptable carriers, in this regard, are intended to include any and all solvents, dispersion media, coatings, absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds (identified or designed according to the disclosure and/or known in the art) also can be incorporated into the compositions. In some embodiments, formulations are prepared by bringing the compound into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.

A pharmaceutical composition of the disclosure should be formulated to be compatible with its intended route of administration. Solutions or suspensions can include the following components: a sterile diluent such as water, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.

Formulations for parenteral administration can also include glycocholate for buccal administration, methoxysalicylate for rectal administration, or citric acid for vaginal administration. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Suppositories for rectal administration also can be prepared by mixing the drug with a non-irritating excipient such as cocoa butter, other glycerides, or other compositions which are solid at room temperature and liquid at body temperatures. Formulations also can include, for example, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, and hydrogenated naphthalenes. Formulations for direct administration can include glycerol and other compositions of high viscosity. Other potentially useful parenteral carriers for these drugs include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation administration can contain as excipients, for example, lactose, or can be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or oily solutions for administration in the form of nasal drops, or as a gel to be applied intranasally. Retention enemas also can be used for rectal delivery.

Formulations of the present disclosure suitable for oral administration can be in the form of: discrete units such as capsules, gelatin capsules, sachets, tablets, troches, or lozenges, each containing a predetermined amount of the drug; a powder or granular composition; a solution or a suspension in an aqueous liquid or non-aqueous liquid; or an oil-in-water emulsion or a water-in-oil emulsion. The drug can also be administered in the form of a bolus, electuary or paste. A tablet can be made by compressing or molding the drug optionally with one or more accessory ingredients. Compressed tablets can be prepared by compressing, in a suitable machine, the drug in a free-flowing form such as a powder or granules, optionally mixed by a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets can be made by molding, in a suitable machine, a mixture of the powdered drug and suitable carrier moistened with an inert liquid diluent.

Oral compositions generally include an inert diluent or an edible carrier. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients. Oral compositions prepared using a fluid carrier for use as a mouthwash include the compound in the fluid carrier and are applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose; a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.

Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). It should be stable under the conditions of manufacture and storage and should be preserved 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), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.

Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation include vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.

Formulations suitable for intra-articular administration can be in the form of a sterile aqueous preparation of the drug that can be in microcrystalline form, for example, in the form of an aqueous microcrystalline suspension. Liposomal formulations or biodegradable polymer systems can also be used to present the drug for both intra-articular and ophthalmic administration.

Formulations suitable for topical administration, including eye treatment, include liquid or semi-liquid preparations such as liniments, lotions, gels, applicants, oil-in-water or water-in-oil emulsions such as creams, ointments or pastes; or solutions or suspensions such as drops. Formulations for topical administration to the skin surface can be prepared by dispersing the drug with a dermatologically acceptable carrier such as a lotion, cream, ointment or soap. Useful are carriers capable of forming a film or layer over the skin to localize application and inhibit removal. For topical administration to internal tissue surfaces, the agent can be dispersed in a liquid tissue adhesive or other substance known to enhance adsorption to a tissue surface. For example, hydroxypropylcellulose or fibrinogen/thrombin solutions can be used to advantage. Alternatively, tissue-coating solutions, such as pectin-containing formulations can be used.

For inhalation treatments, inhalation of powder (self-propelling or spray formulations) dispensed with a spray can, a nebulizer, or an atomizer can be used. Such formulations can be in the form of a fine powder for pulmonary administration from a powder inhalation device or self-propelling powder-dispensing formulations. In the case of self-propelling solution and spray formulations, the effect can be achieved either by choice of a valve having the desired spray characteristics (i.e., being capable of producing a spray having the desired particle size) or by incorporating the active ingredient as a suspended powder in controlled particle size. For administration by inhalation, the compounds also can be delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.

Systemic administration also can be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants can include, for example, for transmucosal administration, detergents and bile salts. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds typically are formulated into ointments, salves, gels, or creams.

The active compounds can be prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Liposomal suspensions can also be used as pharmaceutically acceptable carriers.

Oral or parenteral compositions can be formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the disclosure are dictated by and directly dependent on the unique characteristics of the active compound and the therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals. Furthermore, administration can be by periodic injections of a bolus, or can be made more continuous by intravenous, intramuscular or intraperitoneal administration from an external reservoir (e.g., an intravenous bag).

Where adhesion to a tissue surface is desired the composition can include the drug dispersed in a fibrinogen-thrombin composition or other bioadhesive. The compound then can be painted, sprayed or otherwise applied to the desired tissue surface. Alternatively, the drugs can be formulated for parenteral or oral administration to humans or other mammals, for example, in effective amounts, e.g., amounts that provide appropriate concentrations of the drug to target tissue for a time sufficient to induce the desired effect.

Where the active compound is to be used as part of a transplant procedure, it can be provided to the living tissue or organ to be transplanted prior to removal of tissue or organ from the donor. The compound can be provided to the donor host. Alternatively, or, in addition, once removed from the donor, the organ or living tissue can be placed in a preservation solution containing the active compound. In all cases, the active compound can be administered directly to the desired tissue, as by injection to the tissue, or it can be provided systemically, either by oral or parenteral administration, using any of the methods and formulations disclosed herein. Where the drug comprises part of a tissue or organ preservation solution, any commercially available preservation solution can be used to advantage. For example, useful solutions known in the art include Collins solution, Wisconsin solution, Belzer solution, Eurocollins solution and lactated Ringer's solution.

Generally, an effective amount of dosage of active compound will be in the range of from about 0.1 mg/kg to about 100 mg/kg of body weight/day, for example, from about 1.0 mg/kg to about 50 mg/kg of body weight/day. In some embodiments, the dosage of active compound is in the range of from about 0.1 mg/kg to about 1.0 mg/kg of body weight/day; from about 0.1 mg/kg to about 5 mg/kg of body weight/day; from about 0.1 mg/kg to about 10 mg/kg of body weight/day; from about 0.1 mg/kg to about 25 mg/kg of body weight/day; from about 0.1 mg/kg to about 50 mg/kg of body weight/day; from about 1.0 mg/kg to about 5.0 mg/kg of body weight/day; from about 1.0 mg/kg to about 10 mg/kg of body weight/day; from about 1.0 mg/kg to about 20 mg/kg of body weight/day; from about 1.0 mg/kg to about 25 mg/kg of body weight/day; from about 1.0 mg/kg to about 40 mg/kg of body weight/day; from about 1.0 mg/kg to about 100 mg/kg of body weight/day; from about 10 mg/kg to about 100 mg/kg of body weight/day; from about 25 mg/kg to about 100 mg/kg of body weight/day; from about 50 mg/kg to about 100 mg/kg of body weight/day; from about 5.0 mg/kg to about 50 mg/kg of body weight/day; from about 10 mg/kg to about 50 mg/kg of body weight/day; or from about 25 mg/kg to about 50 mg/kg of body weight/day.

The amount administered will also likely depend on such variables as the type of surgery or invasive medical procedure, the overall health status of the patient, the relative biological efficacy of the compound delivered, the formulation of the drug, the presence and types of excipients in the formulation, and the route of administration. Also, it is to be understood that the initial dosage administered can be increased beyond the above upper level in order to rapidly achieve the desired blood-level or tissue level, or the initial dosage can be smaller than the optimum.

Nonlimiting doses of active compound comprise from about 0.1 mg to about 1500 mg per dose. For example, a dose of active compound can range from about 0.1 mg to about 1250 mg; about 0.1 mg to about 1000 mg; about 0.1 mg to about 800 mg; about 0.1 mg to about 500 mg; about 0.1 mg to about 250 mg; about 0.1 mg to about 100 mg; about 0.1 mg to about 50 mg; about 0.1 mg to about 25 mg; about 0.1 mg to about 20 mg; about 0.1 mg to about 10 mg; about 0.1 mg to about 5 mg; about 0.1 mg to about 1 mg; about 0.1 mg to about 0.5 mg; about 0.5 mg to about 1500 mg; about 1 mg to about 1500 mg; about 2.5 mg to about 1500 mg; about 5 mg to about 1500 mg; about 10 mg to about 1500 mg; about 50 mg to about 1500 mg; about 100 mg to about 1500 mg; about 250 mg to about 1500 mg; about 500 mg to about 1500 mg; about 750 mg to about 1500 mg; about 1000 mg to about 1500 mg; about 1250 mg to about 1500 mg; about 0.25 mg to about 2.5 mg; about 0.5 mg to about 5 mg; about 1 mg to about 10 mg; about 5 to about 20 mg; about 10 mg to about 50 mg; about 25 mg to about 75 mg; about 20 mg to about 100 mg; about 50 mg to about 200 mg; about 100 mg to about 500 mg; about 250 mg to about 750 mg; about 200 mg to about 800 mg; about 500 mg to about 1000 mg; or about 750 mg to about 1250 mg.

As is understood by one of ordinary skill in the art, generally, when dosages are described for a pharmaceutical active, the dosage is given on the basis of the parent or active moiety. Therefore, if a salt, hydrate, or another form of the parent or active moiety is used, a corresponding adjustment in the weight of the compound is made, although the dose is still referred to on the basis of the parent or active moiety delivered. As a nonlimiting example, if the parent or active moiety of interest is a monocarboxylic acid having a molecular weight of 250, and if the monosodium salt of the acid is desired to be delivered to be delivered at the same dosage, then an adjustment is made recognizing that the monosodium salt would have a molecular weight of approximately 272 (i.e., minus 1H or 1.008 atomic mass units and plus 1 Na or 22.99 atomic mass units). Therefore, a 250 mg dosage of the parent or active compound would correspond to about 272 mg of the monosodium salt, which would also deliver 250 mg of the parent or active compound. The another way, about 272 mg of the monosodium salt would be equivalent to a 250 mg dosage of the parent or active compound.

In some embodiments, pyrrolocytosines, such as the compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers, as provided herein, can exhibit an acute clinical syndrome, which manifests as a Cmax-driven hemodynamic effect and is associated with immediate clinical signs such as labored breathing. Cmax is the peak concentration a molecule reaches in the plasma (e.g., directly following intravenous administration), and is expressed generally in micrograms/milliliter. The syndrome is dose-dependent, meaning that the higher the amount of drug given, the more severe are the effects. In some embodiments, this is the limiting toxicity for the class. In some embodiments, however, the efficacy for the pyrrolocytosines, including the compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers, as provided herein, is not driven by the Cmax but rather by the AUC (Area-Under-the-plasma-drug-concentration-time-Curve), which is an expression of the total body exposure to the drug and is expressed generally in micrograms*hour/milliliter. In rat studies with several pyrrolocytosines, including selected compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers as provided herein, it has been shown that increasing the length, or duration, of the intravenous administration results in one or more of effectively modulating or eliminating the clinical syndrome and depressing the concentration maximum. In some embodiments, these effects result in a short distribution half-life but still afford drug exposures necessary for efficacy.

FORMULATION EXAMPLES IA. Formulation for Intravenous Administration

Ingredients Amount Antimicrobial Compound 0.1-1500 total mg of the present disclosure Dextrose, USP 50 mg/ml Sodium citrate, USP 1.60-1.75 mg/ml Citric Acid, USP 0.80-0.90 mg/ml Water, USP q.s

This formulation for intravenous administration is formulated by heating water for injection to about 60° C. Next the sodium citrate, citric acid and dextrose are added and stirred until dissolved. A solution or aqueous slurry of the antimicrobial compound is added to the previous mixture and stirred until dissolved. The mixture is cooled to 25° C. with stirring. The pH is measured and adjusted if necessary. Lastly the mixture is brought to the desired volume, if necessary, with water for injection. The mixture is filtered, filled into the desired container (vial, syringe, infusion container, etc.), over wrapped and terminally moist heat sterilized.

This formulation is useful for intravenous administration, either bolus or infusion, to a patient for treating, preventing, reducing the risk of, or delaying the onset of infection.

IB. Formulation for Intravenous Administration

This formulation for intravenous administration utilizes 6.5 nM tartaric acid buffer in 5% Dextrose, and has a pH of 4.4. This formulation is useful for intravenous administration, either bolus or infusion, to a patient for treating, preventing, reducing the risk of, or delaying the onset of infection.

II. Lyophilisate for Reconstitution

Alternatively, the antimicrobial compound can be provided as a lyophilisate which can be reconstituted before intravenous or intramuscular administration.

Ingredient mg per injection vial Antimicrobial Compound 0.1-1500 of the present disclosure Cyclodextrin 1500

Reconstitution solution for a volume to be administered of 50 ml (infusion): 5% aqueous glucose solution.

Reconstitution solution for a volume to be administered of 15 ml (bolus): 3.3% aqueous glucose solution.

The foregoing lyophilisate is useful for reconstitution and intravenous administration, either bolus or infusion, to a patient for treating, preventing, reducing the risk of, or delaying the onset of infection.

III. Lyophilisate for Reconstitution

Ingredient mg per injection vial Antimicrobial Compound 0.1-1500 of the present disclosure soya lecithin 2250 Sodium cholate 1500

Reconstitution solution for a volume to be administered of 50 ml (infusion): 4% aqueous glucose solution.

Reconstitution solution for a volume to be administered of 15 ml (bolus): 2% aqueous glucose solution

The foregoing lyophilisate is useful for reconstitution and intravenous administration, either bolus or infusion, to a patient for treating, preventing, reducing the risk of, or delaying the onset of infection.

IV. Lyophilisate for Reconstitution

Ingredient mg per injection vial Antimicrobial Compound 0.1-1500 of the present disclosure soya lecithin 900 Sodium glycocholate 540

Reconstitution solution for a volume to be administered of 15 ml (bolus): 3.3% aqueous glucose solution.

The foregoing lyophilisate is useful for reconstitution and intravenous administration, either bolus or infusion, to a patient for treating, preventing, reducing the risk of, or delaying the onset of infection.

V. Tablet for Oral Administration

Ingredients Per Tablet Per 4000 Tablets Antimicrobial Compound 0.1-1500 mg 0.4-6000 g of the present disclosure Anhydrous Lactose, NF 110.45 mg 441.8 g Microcrystalline  80.0 mg 320.0 g Cellulose NF Magnesium Stearate  1.00 mg  4.0 g Impalpable Powder NF Croscarmellose Sodium  2.00 mg  8.0 g NF Type A

The antimicrobial compound (any of the compounds equivalent to the desired delivery strength, e.g., 50 to 1500 mg per tablet) is premixed with ⅓ of the microcrystalline cellulose NF and ½ of the anhydrous lactose NF in a ribbon blender for 5 minutes at 20 RPM. To the premix is added the remaining ⅔ of the microcrystalline cellulose NF and the remaining ½ of the anhydrous lactose NF. This is blended for 10 minutes at 20 RPM. Croscarmellose sodium is added to the blended powders and mixed for 5 minutes at 20 RPM. Finally, the magnesium stearate is added to the mixture by passing through a 90 mesh screen and blended for an additional 5 minutes at 20 RPM. The lubricated mixture is compressed to provide tablets of 500 mg active ingredient.

These tablets are useful for oral administration to a patient for treating, prevention, reducing the risk of, or delaying the onset of infection.

6. EXAMPLES

Nuclear magnetic resonance (NMR) spectra were obtained on a Bruker Avance 300 or Avance 500 spectrometer, or in some cases a GE-Nicolet 300 spectrometer. Common reaction solvents were either high performance liquid chromatography (HPLC) grade or American Chemical Society (ACS) grade, and anhydrous as obtained from the manufacturer unless otherwise noted. “Chromatography” or “purified by silica gel” refers to flash column chromatography using silica gel (EM Merck, Silica Gel 60, 230-400 mesh) unless otherwise noted.

The compounds or tautomers thereof, or pharmaceutically acceptable salts of the compounds or tautomers of the present disclosure can be prepared using known chemical transformations adapted to the particular situation at hand.

Some of the abbreviations used in the following experimental details of the synthesis of the examples are defined below: h or hr=hour(s); min=minute(s); mol=mole(s); mmol=millimole(s); M=molar; μM=micromolar; g=gram(s); μg=microgram(s); rt=room temperature; L=liter(s); mL=milliliter(s); Et2O=diethyl ether; THF=tetrahydrofuran; DMSO=dimethyl sulfoxide; EtOAc=ethyl acetate; Et3N=triethylamine; i-Pr2NEt or DIPEA=diisopropylethylamine; CH2Cl2=methylene chloride; CHCl3=chloroform; CDCl3=deuterated chloroform; CCl4=carbon tetrachloride; MeOH=methanol; CD3OD=deuterated methanol; EtOH=ethanol; DMF=dimethylformamide; BOC=t-butoxycarbonyl; CBZ=benzyloxycarbonyl; TBS=t-butyldimethylsilyl; TBSCl=t-butyldimethylsilyl chloride; TFA=trifluoroacetic acid; DBU=diazabicycloundecene; TBDPSCl=t-butyldiphenylchlorosilane; Hunig's Base=N,N-diisopropylethylamine; DMAP=4-dimethylaminopyridine; CuI=copper (I) iodide; MsCl=methanesulfonyl chloride; NaN3=sodium azide; Na2SO4=sodium sulfate; NaHCO3=sodium bicarbonate; NaOH=sodium hydroxide; MgSO4=magnesium sulfate; K2CO3=potassium carbonate; KOH=potassium hydroxide; NH4OH=ammonium hydroxide; NH4Cl=ammonium chloride; SiO2=silica; Pd—C=palladium on carbon; Pd(dppf)Cl2=dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium (II); Cs2CO3=cesium carbonate; Zn=zinc; LiCl=lithium chloride; DMF=N,N-dimethylformamide; 9-BBN=9-Borabicyclo[3.3.1]nonane; K3PO4=potassium phosphate; DMA=N,N-dimethylacetamide; DIBAL and DIBAL-H=diisobutylaluminum hydride; m-CPBA=meta-chloroperoxybenzoic acid; KOAc=potassium acetate; B2Pin2=bis(pinacolato)diboron; Cu(OAc)2=copper (II) acetate; TMEDA=tetramethylethylenediamine; Bz2O=benzoyl anhydride; DIPEA=N,N-diisopropylethylamine; Pd(PPh3)4=tetrakis(triphenylphosphine)palladium(0); TEA=triethylamine; MsCl=mesityl chloride; HBr=hydrogen bromide; AcOH=acetic acid; IPAC=isopropyl acetate; EDTA=ethylenediaminetetraacetic acid.

Exemplary compounds synthesized in accordance with the disclosure are listed in Tables 1. A bolded or dashed bond is shown to indicate a particular stereochemistry at a chiral center, whereas a wavy bond indicates that the substituent can be in either orientation or that the compound is a mixture thereof.

The compounds of the present disclosure can be prepared, formulated, and delivered as salts. For convenience, the compounds are generally shown without indicating a particular salt form.

The compounds of the present disclosure can be made using synthetic chemical techniques well known to those of skill in the art.

Example 1: Syntheses of Compound 34

Preparation of Intermediate 2a

4-Bromobenzaldehyde 1a (100.0 g, 540.5 mmol) was added in several portions to a solution of (S)-(−)-2-methyl-2-propanesulfinamide (65.51 g, 540.5 mmol) in CH2Cl2 (495 mL) at room temperature. The mixture was stirred under argon until all solids were dissolved, and then Cs2CO3 (176.1 g, 540.5 mmol) was added in several portions. The mixture was stirred and heated to gentle reflux (42-43° C.). After 16 h it was cooled to 0-5° C., and water (500 mL) was slowly added at 15° C. The mixture was stirred at 15-20° C. for 10 min, the phases were separated, and the organic phase was washed with water (250 mL). Afterwards, the organic layer was concentrated in vacuo to about 250 g, additional anhydrous CH2Cl2 (300 mL) was added, the solution was concentrated to a constant mass, then dried at room temperature to give sulfinylimine 2a as a pale yellow oil (156.4 g, 99%).

Preparation of Intermediate 3a

To a solution of sulfinylimine 2a (85.87 g, 297.9 mmol) in DMF (450 mL) at 35° C. was added lithium chloride (25.3 g, 595.8 mmol) over 2 min. Afterwards, the mixture was cooled to 25° C. and H2O (4.56 g, 0.85 mol equivalent) was added. The mixture was stirred at 20-25° C. for 5 min, and then freshly activated zinc powder (38.95 g; 595.8 mmol) was added. Immediately afterwards, allyl bromide (72.1 g; 595.8 mmol) was added dropwise to the mixture over 10 min at 55° C. The temperature was maintained at 45-60° C. for the next 20 minutes. Subsequently, the cooling bath was removed and the mixture was stirred to 30-45° C. over 40-60 min. After reaction completion, the mixture was cooled to 10-15° C. and IPAC (560 mL) was added, followed by the dropwise addition of H2O (400 mL) at 25° C. The resulting mixture was cooled to 15-20° C., and 1N HCl/H2O (550 mL, 6.4 Vol, 0.92 equiv./zinc) was added dropwise at 25° C. Afterwards, IPAC (200 mL) was added and the mixture was stirred for 20 min at at pH of 6. The phases were separated, the organic phase was washed with 5% EDTA solution (pH=7.5; 400 mL), and then with water (2×500 mL). The organic phase was concentrated in vacuo to a constant mass, affording compound 3a (98.02 g, by 1H-NMR contains 2.8 wt % IPAC; 95.27 g, 97% yield).

Preparation of Intermediate 5a

Compound 3a (85.0 g, 257.4 mmol) was dissolved in THE (210 mL). The solution was loaded under argon into a 3 L reactor, stirred and cooled using a water bath at 15-17° C. A solution of 9-BBN in THF (0.5 M; 927 mL, 463.3 mmol) was added dropwise at 17-20° C., over 20 min. The mixture was stirred at 20-22° C. for 50 min. Subsequently, a solution of K3PO4/H2O (2.0 M, in water; 258 mL) was added dropwise over 10 min at 22° C. The resulting mixture was stirred for 5 min, and a solution of t-butyl (Z)-3-iodoacrylate 4a (75.2 g, 296.0 mmol) in THF (50 mL, anhydrous) was added over 5 min. Pd(PPh3)4 (7.4 g, 6.43 mmol) was added and the mixture was stirred at 50-55° C. for 3 h. The mixture was cooled to room temperature, followed by a dropwise addition of water (250 mL) under argon. The phases were separated, the organic phase was concentrated and the residue was partitioned between IPAC (600 mL) and H2O (400 mL). The organic layer was washed with water (2×400 mL) and concentrated in vacuo to a thick oil (208 g). This material was purified on a plug of silica (230-400 mesh, 1.5 kg), eluting with a gradient of 30%-55% EtOAc/heptane (20 L), basing collection of fractions on HPLC analysis. This gave the acrylate 5a (103.0 g, 82.6%) as a light-brown, thick oil.

Preparation of Intermediate 6a

Compound 5a (101.0 g, 220.3 mmol) was dissolved in dimethylacetamide (605 mL). The solution was loaded under argon into a 3 L reactor, and Cs2CO3 (358.4 g, 1.10 mol) was added. The mixture was stirred at room temperature for 10 min, and then heated at 50-52° C. for 8 h, followed by stirring at room temperature for 14 h. Subsequently, the mixture was cooled to 5-10° C. and IPAC (600 mL) was added, followed by an addition of H2O (600 mL) at 30° C., then saturated aqueous NH4Cl (600 mL) over 5 min, resulting in a solution having a pH of 8.5. The mixture was stirred for 10 min at room temperature and the phases were separated. The organic phase was washed with water (2×500 mL) and concentrated in vacuo to give crude product 6a as a tan solid (111.0 g). This sample was dissolved at 60° C. in IPAC (200 mL). Heptane (250 mL) was added, the mixture was cooled to room temperature, seeded with compound 6a (0.2 g), then stirred at room temperature for 14 h. The product was filtered, washed with heptane, and dried at 40° C. to afford product 6a (40.94 g, white needles).

Preparation of Intermediate 7a

A solution of piperidine 6a (71.7 g, 156.4 mmol) in THE (360 mL) was placed under argon in a 3 L reactor. DIBAL-H/THF (1.0M; 469 mL, 469 mmol) was added dropwise at 23-28° C. over 40 min. Afterwards, the mixture was stirred at 22-27° C. for 3 h, and then it was cooled to 0-5° C. and IPAC (940 mL) was slowly added at 15° C. The mixture was stirred for 10 min, and then it was added slowly, at 20° C., to a 5 L reactor containing a solution of potassium-sodium tartrate tetrahydrate (460 g; 1.63 mol) in water (1.0 L) that had been previously to 5-10° C. After the addition, the cooling bath was removed, and the mixture was stirred at room temperature for 3 h. The phases were separated, the organic phase was concentrated in vacuo to 320 g, during which rapid precipitation occurred. The mixture was left standing at room temperature for 14 h, the solid was filtered, washed with IPAC (50 mL), and dried at 50° C., affording the alcohol 7a (53.5 g, 88%).

Preparation of Intermediate 8a

50 g (129 mmol) of 7a was dissolved in 1000 ml of dichloromethane, resulting in a dark brown solution with some undissolved solid. mCPBA (44.6 g, 258 mmol) was added in six portions over 30 minutes (it was observed that the dark brown color of the solution of 7a fades after addition of mCPBA). The resulting clear solution was heated to 35-40° C. in an oil bath for 3 h. The reaction mixture was cooled to room temperature, 2M K2CO3 solution (500 ml) was added, and the mixture was stirred for 15 minutes. The reaction mixture was transferred to a separatory funnel and the layers partitioned. The organic layer was concentrated to afford ˜53 g of an off-white solid which was purified using Combiflash chromatography (330 g column size) to afford 45.95 g (88%) 8a as a white solid.

Preparation of Intermediate 9a

Compound 8a (45.9 g, 113.6 mmol) was dissolved in DMSO (300 ml). To this solution was added bis(pinacolato)diboron (43.3 g, 170.4 mmol), KOAc (33.4 g, 340.8 mmol), and PdCl2(dppf)CH2Cl2 (4.7 g, 5.7 mmol) under argon. The resulting solution was heated in an oil bath at 80° C. under an argon atmosphere for 15 h. After completion of reaction as determined by LC-MS, the reaction mixture was cooled to rt, and diluted with cold water (300 ml) and EtOAc (300 ml). The resulting emulsion was filtered through celite to remove suspended particles, the layers were partitioned, and the aqueous layer was extracted with EtOAc. The combined organic layers were concentrated to obtain a black, viscous oil which was purified by flash chromatography over silica gel to afford 50.64 g (99%) of 9a as a foam.

Preparation of Intermediate 10a

Compound 9a (28.0 g, 62.1 mmol) was dissolved in 1 L of MeOH. 200 mL of water was added to the resulting solution slowly, then additional MeOH was added as needed to eliminate the turbidity that developed after the addition of water. To this solution 5-iodocytosine (17.7 g, 75.5 mmol) was added at RT to result in a first solution. In a separate flask, 100 ml MeOH:water (4:1) was added to Cu(OAc)2.H2O (13.7 g, 68.31 mmol), then TMEDA (37.4 ml, 248.4 mmol) was added to this mixture slowly, resulting in a deep blue second solution that was stirred at RT for 10 minutes. Afterwards, the second solution was added to the first solution. Air was bubbled slowly in the resulting reaction mixture. After stirring at RT for 7 hr, the reaction mixture was concentrated in vacuo to remove as much as MeOH as possible (˜1 L removed). NH4OH (300 ml) and CH2Cl2 (400 ml) was added and resulting solution was stirred at RT overnight (˜12 h). The reaction mixture was filtered to remove the precipitated solid, which was washed with 10% MeOH in dichloromethane. The solid was dried to afford 9.2 g (26%) of 10a as a white solid. The filtrate was extracted with 10 to 20% MeOH in dichloromethane, the washed with water and brine to afford 24.48 g (70%) of 10a as a green foam. Total yield 96%.

Preparation of Intermediates 11a and 12a

45.03 g (80.34 mmol) of crude 10a was dissolved in dry DMF (250 ml). To the solution was added benzoic anhydride (20.9 g, 92.5 mmol) in portions (alternatively, it can be added as a solution in anhydrous DMF). The resulting solution was heated at ˜50-55° C. in an oil bath under argon for 20 h. The reaction mixture was cooled to RT, water and saturated sodium bicarbonate solution was added, then the mixture was stirred for 15 min. The mixture was extracted with EtOAc, then the organic layer was concentrated and purified using Combiflash to afford 30.69 g (58%) of 11a and 12.45 g (17%) of 12a as a foam.

Preparation of Intermediate 14a

To a solution of iodobenzamide 11a (20.3 g, 30.57 mmol) in DMF (120 mL) was added DIPEA (16.0 mL, 91.71 mmol) and the reaction mixture was stirred for 10 min. Pd(PPh3)4 (1.76 g, 1.53 mmol) and CuI (0.58 g, 3.06 mmol) were added, followed by heating to 50° C. over 10 minutes. Then a solution of the alkyne (13a) in DMF (30 mL) was added and stirring was continued for 15 h at 70° C. When the LCMS showed the completion of reaction, it was cooled to ambient temperature and MeOH (200 mL) was added, stirring for 3 h at 70° C. When the LCMS showed the completion of debenzoylation and formation of 14a, it was cooled to room temperature, the methanol was evaporated, and 200 ml of water was added. The product was extracted using ethyl acetate (3×100 ml) and the combined organic layers were washed with water (100 ml), 14% ammonium hydroxide (2×100 ml), water (100 ml), and brine (100 ml). It was then dried over sodium sulfate, concentrated, and purified by flash column chromatography over silica gel eluting with CH2Cl2 and 90% CH2Cl2+9.8% methanol+0.2% NH4OH (i.e., “CMA”) to obtain 18.75 g (yield, 76%) of 14a as an orange solid.

Preparation of Intermediate 15a

To a solution of the alcohol 14a (18.75 g, 23.3 mmol) in MeOH (250 mL) were added silathiol (5 g) and activated charcoal (5 g). The mixture was stirred overnight at ambient temperature. The solution was filtered over a bed of celite (10 g) and was washed with additional MeOH (2×100 mL). The combined filtrates were concentrated and purified by flash column chromatography over silica gel eluting with CH2Cl2 and 90% CH2Cl2+9.8% methanol+0.2% NH4OH (i.e., “CMA”) to obtain (17.0 g, 21.1 mmol) metal-free 14a as a golden orange solid. To a solution of the solid in CH2Cl2 (200 mL) at −5° C. were added TEA (2.63 mL, 18.99 mmol) and MsCl (1.37 mL, 17.94 mmol), and stirring was continued for 15 minutes. When LCMS indicated near complete conversion of the alcohol to mesylate, it was diluted with ice water (50 mL) and brine (100 mL). The organic layer was separated and the aqueous layer was extracted with additional CH2Cl2 (2×150 mL). The combined organic layers were dried over sodium sulfate and concentrated to dryness. To this crude mesylate in EtOH (200 mL) was added thiourea (8.03 g, 105.5 mmol), and the mixture was stirred at 65° C. for 13 h. When LCMS indicated near complete conversion of mesylate to isothiourea, it was then concentrated and diluted with water (150 mL) and saturated NaHCO3 (50 mL). The resulting mixture was extracted with 10% MeOH in EtOAc (3×200 mL), and the combined organic layers were dried over sodium sulfate, concentrated in vacuo, and purified by flash column chromatography over silica gel eluting with CH2Cl2 and 90% CH2Cl2+9.8% methanol+0.2% NH4OH (i.e., “CMA”) to obtain 15.0 g (yield, 75.0%) of 15a as an orange solid.

Preparation of Compound 34

To a solution of 15a (15.0 g, 17.35 mmol) in anhydrous methanol (50 mL) under an argon atmosphere was added 120 mL of HBr solution (33% in acetic acid) at 0° C. This was stirred for 10 min after which the cooling bath was removed. The stirring was continued at room temperature overnight. The solution was concentrated in vacuo, then azeotroped with toluene and ethanol (50 mL each) for 2 times and purified by combiflash using C18 stationary phase (Column: Silicycle 300 g Catalog: FLH-R33230B-IS330). Desired fractions were combined and concentrated to dryness. TFA salt was fully exchanged with HCl (6N aq., 100 ml×1 with 100 ml of ethanol) and lyophilized to obtain 8.5 g (yield, 65%) of 34 as golden yellow solid. 1H NMR (300 MHz, D2O): δ 8.43 (s, 1H), 7.68 (d, J=8.4 Hz, 2H), 7.56 (d, J=8.4 Hz, 2H), 7.39 (d, J=6.5 Hz, 1H), 7.31 (d, J=5.4 Hz, 1H), 6.80 (s, 1H), 4.62-4.58 (m, 1H), 3.87-3.85 (m, 1H), 3.36-3.17 (m, 3H), 2.64-2.48 (m, 3H), 2.28-1.91 (m, 7H), 1.68-1.52 (m, 4H), 1.25 (d, J=6.6, 3H); MS (ESI) m/z [M+H]+; calcd for C31H37ClFN7OS; 610.2, found 610.5.

Example 2: Syntheses of Compound 41

To a solution of 14a (0.5 g, 0.62 mmol) in CH2Cl2 (5 mL) at 0° C. were added TEA (0.6 mL, 1.86 mmol) and MsCl (0.72 mL, 0.9 mmol). Stirring was continued for 3 h, then the reaction was warmed to room temperature and stirred for 1 h. It was then diluted with ice-cold water (10 mL) and brine (10 mL). The organic layer was separated and the aqueous layer was extracted with additional CH2Cl2 (2×10 mL). The combined organic layers were dried over sodium sulfate and concentrated to dryness to afford 0.40 g of the mesylate. To this crude mesylate in EtOH (5 mL) was added N-methyl thiourea (0.42 g, 4.5 mol) and stirred at 70° C. for 3 h. It was then concentrated in vacuo to afford 16a. Crude 16a was diluted with methanol (3 mL) and HBr solution (3 mL, 33% in acetic acid) at 0° C. The resulting mixture was stirred for 10 min and the cooling bath was removed. The stirring was continued at room temperature overnight. The solution was concentrated and azeotroped with toluene and ethanol (10 mL each) 3 times and purified by HPLC using a methanol/TFA solvent system. TFA was exchanged with HCl (6N aq., 10 ml×4) and lyophilized to obtain 0.17 g of compound 41.

Example 3: Syntheses of Compound 43

To a solution of 14a (0.5 g, 0.62 mmol) in CH2Cl2 (5 mL) at 0° C. were added TEA (0.6 mL, 1.86 mmol) and MsCl (0.724 mL, 0.9 mmol). The mixture was stirred for 3 h and then warmed up to room temperature and stirred for an additional 1 h. It was then diluted with ice-cold water (10 mL) and brine (10 mL). The organic layer was separated and the aqueous layer was extracted with additional CH2Cl2 (2×10 mL). The combined organic layers were dried over sodium sulfate and concentrated to dryness to 0.44 g of the mesylate. To this crude mesylate in EtOH (5 mL) was added N,N′dimethyl thiourea (0.53 g, 4.5 mol) and the mixture stirred at 70° C. for 3 h. It was then concentrated in vacuo to afford 17a. This compound was diluted with methanol (3 mL) and HBr solution (3 mL, 33% in acetic acid) at 0° C. This was stirred for 10 min and the cooling bath was removed. The stirring was continued at room temperature overnight. The solution was concentrated and azeotroped with toluene and ethanol (10 mL each) for 3 times and purified by HPLC using a methanol-TFA solvent system. TFA was fully exchanged with HCl (6N aq., 10 ml x 4) and lyophilized to obtain 0.15 g of compound 43.

Example 4: Syntheses of Compound 2

To a solution of 14a (0.8 g, 0.98 mmol) in CH2Cl2 (10 mL) at 0° C. were added TEA (0.6 mL, 1.86 mmol) and MsCl (1.4 mL, 1.2 mmol). The reaction was stirred for 3 h, then warmed to ambient temperature and stirred for an additional 1 h. It was then diluted with ice-cold water (20 mL) and brine (20 mL). The organic layer was separated and the aqueous layer was extracted with additional CH2Cl2 (2×20 mL). The combined organic layers were dried over sodium sulfate and concentrated to dryness to 0.88 g of the mesylate. To this crude mesylate in EtOH (10 mL) was added imidazolidine-2-thione (0.25 g, 2.5 mol) and stirred at 70° C. for 3 h. It was then concentrated in vacuo to afford 18a (0.3 g). This compound was diluted with methanol (3 mL) and HBr solution (3 mL, 33% in acetic acid) at 0° C. This was stirred for 10 min and the cooling bath was removed. The stirring was continued at room temperature overnight. The solution was concentrated and azeotroped with toluene and ethanol (10 mL each) 3 times and purified by HPLC using a methanol-TFA solvent system. TFA was fully exchanged with HCl (6N aq., 10 ml x 4) and lyophilized to obtain 0.103 g of compound 2.

Example 4: Syntheses of Compound 120

To a solution of 200 (800 mg, 935 umol, 1 eq) in tetrahydrofuran (10 mL) was added triethylamine (189 mg, 1.87 mmol, 2 eq) and compound 203 (200 mg, 1.12 mmol, 1.2 eq). The reaction was stirred at room temperature for 2 hours. Then ammonia (637 mg, 9.35 mmol, 624 uL, 25% purity, 10 eq) and methanol (3 mL) was added to the reaction mixture. The reaction was stirred at room temperature for 3 hours. On completion, the reaction mixture was concentrated in vacuum. The residue was purified by silica gel chromatography (Dichloromethane:Methanol=20:1) to give compound 201 (250 mg, 27.6% yield) as a yellow solid. LCMS: m/z (M+H)+=878.3

To a solution of compound 201 (0.4 g, 456 umol, 1 eq) in methanol (10 mL) was added methyl iodide (64 mg, 456 umol, 1 eq) at 0° C. The reaction mixture was stirred at room temperature for 16 hours. On completion, the reaction mixture was concentrated in vacuum and purified by silica gel chromatography (Dichloromethane:Methanol=50:1) to give compound 202 (0.2 g, 49% yield) as a yellow solid. LCMS: m/z (M+H)+=892.2

To a solution of compound 202 (0.15 g, 168 umol, 1 eq) in methanol (4.5 mL) was added hydrogen bromide (10.5 mL, 30% in acetic acid) at 0° C. The reaction was stirred at room temperature for 12 hours. On completion, the reaction mixture was concentrated under vacuum to remove the solvents and the solid was dissolved in methanol (5.00 mL) and then purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 7%-37%, 10 min). The solution was lyophilized to give compound 120 (120, combined two batches, 46.3% yield) as a yellow solid: LCMS: m/z (M+H)+=624.2.

Example 6—Antimicrobial Activity

The compounds of the present disclosure were tested for antimicrobial activity. These data are presented in Table 2. The Compounds 1-122 were run against Eschericia coli (E. coli) strain ATCC25922 and against Staphylococcus aureus (S. aureus) 11540 strain using a standard microdilution assay to determine minimum inhibitory concentrations (MICs). The data is presented whereby a “+” indicates that the compound has an MIC value of 16 micrograms/mL or less and a “−” indicates that the compound has an MIC value greater than 16 micrograms/mL. It will be recognized by one skilled in the art that the compounds can be assessed against other bacterial organisms and that the presentation of data for activity against Eschericia coli and Staphylococcus aureus are illustrative and in no way is intended to limit the scope of the present disclosure. The compounds of the present disclosure can be assayed against a range of other microorganisms depending upon the performance activity desired to be gathered. Furthermore, the “+” and “−” representation and the selection of a cutoff value of 16 micrograms/mL is also illustrative and in no way is intended to limit the scope of the present disclosure. For example, a “−” is not meant to indicate that the compound necessarily lacks activity or utility, but rather that its MIC value against the indicated microorganism is greater than 16 micrograms/mL.

TABLE 2 MIC MIC # S. aureus E.coli 1 + + 2 + + 3 + + 4 + + 5 + + 6 + + 7 + + 8 + + 9 + + 10 + + 11 + + 12 + + 13 + + 14 + + 15 + + 16 + + 17 + + 18 + + 19 + + 20 + + 21 + + 22 + + 23 + + 24 + + 25 + + 26 + + 27 + + 28 + + 29 + + 30 + + 31 + + 32 + + 33 + + 34 + + 35 + + 36 + + 37 + + 38 + + 39 + + 40 + + 41 + + 42 + + 43 + + 44 + + 45 + + 46 + + 47 + + 48 + + 49 + + 50 + + 51 + + 52 + + 53 + + 54 + + 55 + + 56 + + 57 + + 58 + + 59 + + 60 + + 61 + + 62 + + 63 + + 64 + + 65 + + 66 + + 67 + + 68 + + 69 + + 70 + + 71 + + 72 + + 73 + + 74 + + 75 + + 76 + + 77 + 78 + + 79 + + 80 + + 81 + + 82 + + 83 + + 84 + + 85 + + 86 + + 87 + + 88 + + 89 + + 90 + + 91 + + 92 + + 93 + + 94 + + 95 + + 96 + + 97 + 98 + + 99 + + 100 + + 101 + + 102 + + 103 + + 104 + + 105 + + 106 + + 107 + + 108 + + 109 + + 110 + + 111 + + 112 + + 113 + + 114 + + 115 + 116 + + 117 + + 118 + + 119 + + 120 + + 121 + + 122 + +

Example 7—Rat Infusion Study

Many pyrrolocytosines, including Compound No. 34, share an acute clinical syndrome, which manifests in as a Cmax-driven hemodynamic effect and is associated with immediate clinical signs such as labored breathing. Cmax is the peak concentration a molecule reaches in the plasma, in this case directly following intravenous administration, and is expressed generally in micrograms/milliliter. The syndrome is dose-dependent, meaning that the higher the amount of drug given, the more severe the effects. This is generally considered the limiting toxicity for the effected compounds of this class. Importantly, the efficacy for the pyrrolocytosines, including Compound No. 34, is not driven by the Cmax but rather by the AUC (Area-Under-the-plasma-drug-concentration-time-Curve), which is an expression of the total body exposure to the drug and is expressed generally in micrograms*hour/milliliter. In rat studies with several pyrrolocytosines, including Compound No. 34, it has been shown that increasing the length, or duration, of the intravenous administration effectively modulates or eliminates the clinical syndrome, depresses the concentration maximum, results in a short distribution half-life but still affords drug exposures necessary for efficacy.

As an example, a study conducted by PharmOptima under study number 2017-164 with Compound No. 34, compared the clinical signs and plasma drug concentration data across several infusion lengths: 10-minutes, which is the screening length at which the acute syndrome is observed, 1-hour, 2-hour, 3-hour and 24-hour. A total of 15 male Sprague-Dawley rats (275-300 grams) were used in the study, divided evenly across the 5 dosing regimens. The route of drug administration was via jugular venous infusion, using a syringe infusion pump. Compound No. 34 was formulated in phosphate-buffered mannitol, with a starting pH=6.5. Details of the infusions and the plasma sample timepoints are given in Table 3.

TABLE 3 Details of Rat Infusion Periods Plasma timepoints, Test Article Infusion Infusion Volume in hours Group (dose) Duration rate delivered (0 h = infusion end) 1 (n = 3) 10 min 3 mL/10 min  3 mL 0, 0.083, 0.5, 1, 2, 4, 6, 24, 48, 72, 96 2 (n = 3) Compound  1 h 1 mL/h  1 mL −0.25, 0, 0.083, 0.5, 1, 2, No. 34 (4 4, 6, 24, 48, 72, 96 3 (n = 3) mg/kg)  2 h  2 mL −0.25, 0, 0.083, 0.5, 1, 2, 4, 6, 24, 48, 72, 96 4 (n = 3)  3 h  3 mL −0.25, 0, 0.083, 0.5, 1, 2, 4, 6, 24, 48, 72, 96 5 (n = 3) 24 h 24 mL −22, −20, −18, −1, −0.25, 0, 0.083, 0.5, 1, 2, 4, 6, 24, 48, 72, 96

Table 4 shows the results. In the short 10-minute infusion, all three rats displayed signs of significant labored breathing; the 1 h, 2 h and 3 h infusion groups only showed minor signs of chewing and licking, and the 24 h infusion group showed no clinical signs. There was clear decrease in Cmax as infusion length was increased. The intermediate dosing groups still achieved significant AUC, or drug exposures for efficacy, and particularly with the 3 h infusion itis clear that the dose can be increased to move the AUC higher while not approaching the Cmax associated with the severe end of the acute tolerability. The 24 h infusion is particularly compelling. It indicates the concentration can be titrated to achieve steady-state drug levels for efficacy while remaining well below the Cmax associated with clinical signs. Furthermore, the alpha, or distribution-phase, half-life is short, which indicates that—if clinical signs are observed—the infusion can be stopped and the plasma levels drop quickly. In the end, this translates into a generous therapeutic window.

TABLE 4 IV Plasma Pharmacokinetics of Compound No. 34 in Male Sprague-Dawley Rats (N = 3/infusion-group) IV Infusion 10-min 1 h 2 h 3 h 24 h Clinical Signs significant labored chewing/licking none breathing Cmax (μg/mL) 23.60 8.89 8.12 3.76 0.56 T½ (h)-terminal 15.19 15.17 T½ (h)-alpha 1.01 0.91 1.02 0.99 1.01 AUCinf (μg/mL*h) 21.99 14.37 14.43 9.15 11.05

INCORPORATION BY REFERENCE

The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes.

EQUIVALENTS

The disclosure can be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the disclosure described herein. Scope of the disclosure is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims

1. A compound of Formula (AA):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
J is selected from
X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;
Z is selected from
R1 is selected from H, C1-3 alkyl, and C2-4 alkenyl;
R2 is selected from H and C1-3 alkyl;
R3 is selected from H and C1-3 alkyl;
or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
or R1 and R3 together with the nitrogen and sulfur atom to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
R4 is selected from H and C1-3 alkyl;
R5 is selected from H and C1-6 alkyl;
R6 is selected from H, C1-6 alkyl, C2-6 alkenyl, and C3-6 cycloalkyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, and 3-6 membered heterocyclyl;
R7 is selected from H and C1-6 alkyl;
or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:
wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:
wherein the ring is optionally substituted on a ring carbon atom with OH;
Q is selected from C1-2 alkylene or —C(O)—;
R21 is selected from H, C1-6 alkyl optionally substituted with 1-3 halo;
R8 is selected from H and halogen;
each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
each Ra is independently selected from H and C1-6 alkyl;
each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, OC(NH)NH2, and aryl;
each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2; or
a compound of Formula (A):
or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;
Z is selected from
R1 is selected from H and C1-3 alkyl;
R2 is selected from H and C1-3 alkyl;
R3 is selected from H and C1-3 alkyl;
or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
R4 is selected from H and C1-3 alkyl;
R5 is selected from H and C1-6 alkyl;
R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, and 3-6 membered heterocyclyl;
R7 is selected from H and C1-6 alkyl;
or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:
wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:
wherein the ring is optionally substituted on a ring carbon atom with OH;
Q is selected from C1-2 alkylene or —C(O)—;
R21 is selected from H, C1-6 alkyl optionally substituted with 1-3 halo;
R8 is selected from H and halogen;
each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
each Ra is independently selected from H and C1-6 alkyl;
each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2;
or a compound of Formula (I)
or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;
R1 is selected from Hand C1-3 alkyl;
R2 is selected from Hand C1-3alkyl;
R3 is selected from Hand C1-3alkyl;
or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
R4 is selected from Hand C1-3alkyl;
R5 is selected from Hand C1-6alkyl;
R6 is selected from H, C1-6alkyl, and C2-6 alkenyl, wherein the C1-6alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6cycloalkyl, and 3-6 membered heterocyclyl;
R7 is selected from H and C1-6alkyl;
or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:
wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:
wherein the ring is optionally substituted on a ring carbon atom with OH;
R8 is selected from H and halogen;
each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
each Ra is independently selected from H and C1-6 alkyl;
each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

2. The compound of claim 1, wherein each of R1, R2, and R3 is H.

3. The compound of claim 1, wherein two of R1, R2, and R3 are H, and the other is C1-3 alkyl.

4. The compound of any one of claims 1 and 2, wherein two of R1, R2, and R3 are H, and the other is methyl.

5. The compound of claim 1, wherein one of R1, R2, and R3 is H, and the other two are C1-3 alkyl.

6. The compound of any one of claims 1 and 5, wherein one of R1, R2, and R3 is H, and the other two are methyl.

7. The compound of claim 1, wherein R1 is H; and R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring.

8. The compound of claim 1, wherein R1 is H; and R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form an imidazoline.

9. The compound of any one of claims 1-8, wherein R4 is H.

10. The compound of any one of claims 1-8, wherein R4 is C1-3 alkyl.

11. The compound of any one of claims 1-10, wherein one of R5 and R7 is H and the other is C1-6 alkyl.

12. The compound of any one of claims 1-11, wherein R5 is H and R7 is C1-6 alkyl.

13. The compound of any one of claims 1-10, wherein R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

14. The compound of any one of claims 1-10, wherein R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form wherein the ring is optionally substituted with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH.

15. The compound of any one of claims 1-10, wherein R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

16. The compound of any one of claims 1-10, wherein R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

17. The compound of any one of claims 1-10, wherein R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

18. The compound of any one of claims 1-10, wherein R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

19. The compound of any one of claims 1-16, wherein R6 is selected from H and C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa.

20. The compound of any one of claims 1-19, wherein R6 is H.

21. The compound of any one of claims 1-19, wherein R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa.

22. The compound of any one of claims 1-19 and 21, wherein R6 is C1-6 alkyl optionally substituted with ORa.

23. The compound of any one of claims 1-19, 21, and 22, wherein Ra is H.

24. The compound of any one of claims 1-19, 21, and 22, wherein Ra is C1-6 alkyl.

25. The compound of any one of claims 1-19, 21, and 22, wherein R6 is C1-6 alkyl substituted with —SC(NH)NH2.

26. The compound of any one of claims 1-16, wherein R6 is C1-6 alkyl optionally substituted with halogen.

27. The compound of any one of claims 1-16 and 26, wherein R6 is C1-6 alkyl optionally substituted with fluoro.

28. The compound of any one of claims 1-27, wherein X is selected from a 5-membered heterocyclyl ring and phenyl, wherein each of the 5-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX.

29. The compound of any one of claims 1-28, wherein X is a 5-membered heterocyclyl ring optionally substituted with one or more RX.

30. The compound of any one of claims 1-29, wherein X is a pyrrolidinyl optionally substituted with one or more RX.

31. The compound of any one of claims 1-30, wherein X is a 2- or 3-pyrrolidinyl optionally substituted with one or more RX.

32. The compound of any one of claims 1-28, wherein X is phenyl optionally substituted with one or more RX.

33. The compound of any one of claims 1-28 and 32, wherein X is phenyl optionally substituted with three RX.

34. The compound of any one of claims 1-33, wherein RX is independently selected from halogen, C1-6 alkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb.

35. The compound of any one of claims 1-34, wherein RX is independently selected from halogen and C1-6 alkyl optionally substituted with one or more Rb.

36. The compound of any one of claims 1-35, wherein Rb is selected from C2-6 alkenyl, ORc, N(Rc)2, and C3-6 cycloalkyl.

37. The compound of any one of claims 1-36, wherein Rb is selected from C2-6 alkenyl, ORc, NH2, and cyclopropyl.

38. The compound of any one of claims 1-37, wherein Rc is selected from H and C1-6 alkyl.

39. The compound of any one of claims 1-37, wherein Rb is selected from vinyl, OH, and NH2.

40. The compound of any one of claims 1-39, wherein two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring.

41. A compound of Formula (IA):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;
R1 is selected from H and C1-3 alkyl;
R2 is selected from H and C1-3 alkyl;
R3 is selected from H and C1-3 alkyl;
R4 is selected from H and C1-3 alkyl;
R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, or C3-6 heterocyclyl;
each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
each Ra is independently selected from H and C1-6 alkyl;
each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

42. The compound of claim 41, wherein each of R1, R2, and R3 is H.

43. The compound of claim 41, wherein two of R1, R2, and R3 are H, and the other is C1-3 alkyl.

44. The compound of any one of claims 41 and 42, wherein two of R1, R2, and R3 are H, and the other is methyl.

45. The compound of claim 41, wherein one of R1, R2, and R3 is H, and the other two are C1-3 alkyl.

46. The compound of any one of claims 41 and 45, wherein one of R1, R2, and R3 is H, and the other two are methyl.

47. The compound of claim 41, wherein R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring.

48. The compound of claim 41, wherein R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form an imidazoline.

49. The compound of any one of claims 41-48, wherein R4 is H.

50. The compound of any one of claims 41-48, wherein R4 is C1-3 alkyl.

51. The compound of any one of claims 41-50, wherein R6 is selected from H and C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and OR.

52. The compound of any one of claims 41-51, wherein R6 is H.

53. The compound of any one of claims 41-51, wherein R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa.

54. The compound of any one of claims 41-51 and 53, wherein R6 is C1-6 alkyl optionally substituted with ORa.

55. The compound of any one of claims 41-51, 53, and 54, wherein Ra is H.

56. The compound of any one of claims 41-51, 53, and 54, wherein Ra is C1-6 alkyl.

57. The compound of any one of claims 41-50, wherein R6 is C1-6 alkyl substituted with —SC(NH)NH2.

58. The compound of any one of claims 41-50, wherein R6 is C1-6 alkyl optionally substituted with halogen.

59. The compound of any one of claims 41-50 and 58, wherein R6 is C1-6 alkyl optionally substituted with fluoro.

60. The compound of any one of claims 41-59, wherein X is selected from a 5-membered heterocyclyl ring and phenyl, wherein each of the 5-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX.

61. The compound of any one of claims 41-60, wherein X is a 5-membered heterocyclyl ring optionally substituted with one or more RX.

62. The compound of any one of claims 41-61, wherein X is a pyrrolidinyl optionally substituted with one or more RX.

63. The compound of any one of claims 41-62, wherein X is a 2- or 3-pyrrolidinyl optionally substituted with one or more RX.

64. The compound of any one of claims 41-60, wherein X is phenyl optionally substituted with one or more RX.

65. The compound of any one of claims 41-60 and 64, wherein X is phenyl optionally substituted with three RX.

66. The compound of any one of claims 41-65, wherein RX is independently selected from halogen, C1-6 alkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb.

67. The compound of any one of claims 41-66, wherein RX is independently selected from halogen and C1-6 alkyl optionally substituted with one or more Rb.

68. The compound of any one of claims 41-66, wherein Rb is selected from C2-6 alkenyl, ORc, N(Rc)2, and C3-6 cycloalkyl.

69. The compound of any one of claims 41-66 and 68, wherein Rb is selected from C2-6 alkenyl, ORc, NH2, and cyclopropyl.

70. The compound of claim 69, wherein Rc is selected from H and C1-6 alkyl.

71. The compound of claim 69, wherein Rb is selected from vinyl, OH, and NH2.

72. The compound of any one of claims 41-71, wherein two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring.

73. A compound of Formula (IB):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
X is selected from a 5- or 6-membered heterocyclyl ring and phenyl, wherein each of the 5- or 6-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX;
each RX is independently selected from halogen, C1-6 alkyl, C1-4 haloalkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
or two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring;
each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
each Rc is independently selected from H, C1-6 alkyl, aryl, —C(O)aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
each Rd is independently selected from C1-3 alkyl, OH, O(C1-3 alkyl), NO2, NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

74. The compound of claim 73, wherein X is selected from a 5-membered heterocyclyl ring and phenyl, wherein each of the 5-membered heterocyclyl ring and phenyl is optionally substituted with one or more RX.

75. The compound of any one of claims 73 and 74, wherein X is a 5-membered heterocyclyl ring optionally substituted with one or more RX.

76. The compound of any one of claims 73-75, wherein X is a pyrrolidinyl optionally substituted with one or more RX.

77. The compound of any one of claims 73-76, wherein X is a 2- or 3-pyrrolidinyl optionally substituted with one or more RX.

78. The compound of any one of claims 73 and 74, wherein X is phenyl optionally substituted with one or more RX.

79. The compound of any one of claims 73, 74, and 78, wherein X is phenyl optionally substituted with three RX.

80. The compound of any one of claims 73-79, wherein RX is independently selected from halogen, C1-6 alkyl, ORc, N(Rc)2, —C(O)ORc, —C(O)Rc, C3-6 cycloalkyl, and aryl, wherein the C1-6 alkyl is optionally substituted with one or more Rb.

81. The compound of any one of claims 73-79, wherein RX is independently selected from halogen and C1-6 alkyl optionally substituted with one or more Rb.

82. The compound of claim 81, wherein Rb is selected from C2-6 alkenyl, ORc, N(Rc)2, and C3-6 cycloalkyl.

83. The compound of any one of claims 81 and 82, wherein Rb is selected from C2-6 alkenyl, ORc, NH2, and cyclopropyl.

84. The compound of any one of claims 73-83, wherein Rc is selected from H and C1-6 alkyl.

85. The compound of any one of claims 81-83, wherein Rb is selected from vinyl, OH, and NH2.

86. The compound of any one of claims 73-85, wherein two adjacent RX come together with the atoms to which they are attached to form a 5- or 6-membered ring.

87. A compound of Formula (II):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
R1 is selected from H and C1-3 alkyl;
R2 is selected from H and C1-3 alkyl;
R3 is selected from H and C1-3 alkyl;
or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
R4 is selected from H and C1-3 alkyl;
R5 is selected from H and C1-6 alkyl;
R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, —SC(NH)NH2, C3-6 cycloalkyl, or C3-6 heterocyclyl;
R7 is selected from H and C1-6 alkyl;
or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:
wherein the ring is optionally substituted on a ring carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:
R8 is selected from H and halogen;
R9 is selected from H and halogen;
R10 is selected from H, halogen, C1-4 alkyl, C1-4 haloalkyl, and C3-6 cycloalkyl;
R11 is selected from C1-3 alkyl, C2-4 alkenyl, and C3-6 cycloalkyl, wherein the C1-3 alkyl is optionally substituted with one or more Rb;
R17 is selected from H, halogen, and C1-6 alkyl;
each Ra is independently selected from H and C1-6 alkyl;
each Rb is independently selected from ORc, —C(O)ORc, and —(O)aryl, wherein the aryl is optionally substituted with one or more Rd;
each Rc is independently selected from hydrogen, C1-3 alkyl, OH, O(C1-3 alkyl), NO2, —C(O)aryl, and aryl; and
each Rd is C1-3 alkyl.

88. The compound of claim 87, wherein each of R1, R2, and R3 is H.

89. The compound of claim 87, wherein two of R1, R2, and R3 are H, and the other is C1-3 alkyl.

90. The compound of any one of claims 87 and 89, wherein two of R1, R2, and R3 are H, and the other is methyl.

91. The compound of claim 87, wherein one of R1, R2, and R3 is H, and the other two are C1-3 alkyl.

92. The compound of any one of claims 87 and 91, wherein one of R1, R2, and R3 is H, and the other two are methyl.

93. The compound of claim 87, wherein R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring.

94. The compound of any one of claim 87 or 93, wherein R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form an imidazoline.

95. The compound of any one of claims 87-94, wherein R4 is H.

96. The compound of any one of claims 87-94, wherein R4 is C1-3 alkyl.

97. The compound of any one of claims 87-96, wherein one of R5 and R7 is H and the other is C1-6 alkyl.

98. The compound of any one of claims 87-97, wherein R5 is H and R7 is C1-6 alkyl.

99. The compound of any one of claims 87-96, wherein R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

100. The compound of any one of claims 87-96, wherein R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form wherein the ring is optionally substituted with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH.

101. The compound of any one of claims 87-96, wherein R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

102. The compound of any one of claims 87-96, wherein R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form

103. The compound of any one of claims 87-96, wherein R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

104. The compound of any one of claims 87-96, wherein R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula

105. The compound of any one of claims 87-102, wherein R6 is selected from H and C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and OR.

106. The compound of any one of claims 87-102 and 105, wherein R6 is H.

107. The compound of any one of claims 87-102 and 105, wherein R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa.

108. The compound of any one of claims 87-102, 105, and 107, wherein R6 is C1-6 alkyl optionally substituted with OR.

109. The compound of any one of claims 87-105, 107, and 108, wherein Ra is H.

110. The compound of any one of claims 87-105, 107, and 108, wherein Ra is C1-6 alkyl.

111. The compound of any one of claims 87-102, wherein R6 is C1-6 alkyl substituted with —SC(NH)NH2.

112. The compound of any one of claims 87-102, 105, and 107, wherein R6 is C1-6 alkyl optionally substituted with halogen.

113. The compound of any one of claims 87-102, 105, 107, and 112, wherein R6 is C1-6 alkyl optionally substituted with fluoro.

114. The compound of any one of claims 87-113, wherein R8 is H.

115. The compound of any one of claims 87-114, wherein one of R9 and R10 is halogen and the other is H.

116. The compound of any one of claims 87-114, wherein each of R9 and R10 is halogen.

117. The compound of any one of claims 87-114 and 116, wherein R9 is fluoro and R10 is chloro.

118. The compound of any one of claims 87-114, wherein R9 is H and R10 is C1-4 alkyl.

119. The compound of any one of claims 87-114, wherein R9 is halogen and R10 is C1-4 alkyl.

120. The compound of any one of claims 87-114 and 119, wherein R9 is chloro and R10 is C1-4 alkyl.

121. The compound of any one of claims 87-114 and 119, wherein R9 is fluoro and R10 is C1-4 alkyl.

122. The compound of any one of claims 87-114 and 118-121, wherein R10 is ethyl.

123. The compound of any one of claims 87-114 and 118-121, wherein R10 is isopropyl.

124. The compound of any one of claims 87-114, wherein R9 is H and R10 is C3-6 cycloalkyl.

125. The compound of any one of claims 87-114 and 124, wherein R10 is cyclopropyl.

126. The compound of any one of claims 87-114 and 124, wherein R10 is cyclopentyl.

127. The compound of any one of claims 87-126, wherein R11 is selected from C1-3 alkyl optionally substituted with one or more R, and C2-4 alkenyl.

128. The compound of any one of claims 87-126, wherein R11 is selected from C1-3 alkyl optionally substituted with one or more R, and C3-6 cycloalkyl.

129. The compound of any one of claims 87-128, wherein R11 is C1-3 alkyl optionally substituted with one or more Rb.

130. The compound of any one of claims 127-129, wherein Rb is ORc, and wherein Rc is selected from H and C1-3 alkyl.

131. The compound of claim 130, wherein Rc is H.

132. The compound of claim 130, wherein Rc is C1-3 alkyl.

133. The compound of any one of claims 87-129, wherein R11 is C1-3 alkyl.

134. The compound of any one of claims 87-129 and 133, wherein R11 is methyl.

135. The compound of any one of claims 87-126 and 128, wherein R11 is C3-6 cycloalkyl.

136. The compound of any one of claims 87-126, 128, and 135, wherein R11 is cyclopropyl.

137. The compound of any one of claims 87-136, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (R).

138. The compound of any one of claims 87-137, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

139. The compound of any one of claims 87-138, wherein R17 is H.

140. The compound of any one of claims 87-138, wherein R17 is halogen.

141. The compound of any one of claims 87-114, 116, 129, and 137-140, wherein each of R9 and R10 is halogen, and R11 is C1-3 alkyl optionally substituted with one or more Rb.

142. The compound of claim 141, wherein R9 is fluoro, R10 is chloro, and R11 is C1-3 alkyl optionally substituted with one or more Rb.

143. The compound of claim 142, wherein R9 is fluoro, R10 is chloro, and R11 is methyl.

144. The compound of claim 143, wherein R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

145. The compound of any one of claims 87, 97-102, 114, 139, and 140, wherein each of R1, R2, and R3 is H; R4 is H; R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa; R9 is fluoro, R10 is chloro, and R11 is methyl.

146. The compound of any one of claims 87, 97-102, 114, 139, and 140, wherein each of R1, R2, and R3 is H; R4 is H; R6 is H or C1-6 alkyl; R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

147. The compound of any one of claims 87, 114, 139, and 140, wherein each of R1, R2, and R3 is H; R4 is H; R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring of formula R9 is fluoro; R10 is chloro; and R11 is methyl.

148. The compound of any one of claims 87, 114, 139, and 140, wherein each of R1, R2, and R3 is H; R4 is H; R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form R6; R6 is H or C1-6 alkyl; R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

149. A compound of Formula (IIA):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
R1 is selected from H and C1-3 alkyl;
R2 is selected from H and C1-3 alkyl;
R3 is selected from H and C1-3 alkyl;
R4 is selected from H and C1-3 alkyl;
R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen and ORa;
R9 is selected from H and halogen;
R10 is selected from H, halogen, C1-4 alkyl, C1-4 haloalkyl, and C3-6 cycloalkyl
R11 is selected from C1-3 alkyl, C2-4 alkenyl, and C3-6 cycloalkyl, wherein the C1-3 alkyl is optionally substituted with one or more Rb;
each Ra is independently selected from H and C1-6 alkyl;
each Rb is independently selected from ORc, —C(O)ORc, and —(O)aryl, wherein the aryl is optionally substituted with one or more Rd;
each Rc is independently selected from hydrogen, C1-3 alkyl, OH, O(C1-3 alkyl), NO2, —C(O)aryl, and aryl; and
each Rd is C1-3 alkyl.

150. The compound of claim 149, wherein each of R1, R2, and R3 is H.

151. The compound of claim 149, wherein two of R1, R2, and R3 are H, and the other is C1-3 alkyl.

152. The compound of any one of claims 149 and 151, wherein two of R1, R2, and R3 are H, and the other is methyl.

153. The compound of claim 149, wherein one of R1, R2, and R3 is H, and the other two are C1-3 alkyl.

154. The compound of any one of claims 149 and 153, wherein one of R1, R2, and R3 is H, and the other two are methyl.

155. The compound of claim 149, wherein R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring.

156. The compound of any one of claims 149 and 155, wherein R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form an imidazoline.

157. The compound of any one of claims 149-156, wherein R4 is H.

158. The compound of any one of claims 149-156, wherein R6 is selected from H and C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and OR.

159. The compound of any one of claims 149-158, wherein R6 is H.

160. The compound of any one of claims 149-158, wherein R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa.

161. The compound of any one of claims 149-158 and 160, wherein R6 is C1-6 alkyl optionally substituted with ORa.

162. The compound of any one of claims 149-158 and 160-161, wherein Ra is H.

163. The compound of any one of claims 149-158 and 160-161, wherein Ra is C1-6 alkyl.

164. The compound of any one of claims 149-158 and 160, wherein R6 is C1-6 alkyl optionally substituted with halogen.

165. The compound of any one of claims 149-158, 160, and 164, wherein R6 is C1-6 alkyl optionally substituted with fluoro.

166. The compound of any one of claims 149-165, wherein one of R9 and R10 is halogen and the other is H.

167. The compound of any one of claims 149-165, wherein each of R9 and R10 is halogen.

168. The compound of any one of claims 149-165 and 167, wherein R9 is fluoro and R10 is chloro.

169. The compound of any one of claims 149-165, wherein R9 is H and R10 is C1-4 alkyl.

170. The compound of any one of claims 149-165, wherein R9 is halogen and R10 is C1-4 alkyl.

171. The compound of any one of claims 149-165 and 170, wherein R9 is chloro and R10 is C1-4 alkyl.

172. The compound of any one of claims 149-165 and 170, wherein R9 is fluoro and R10 is C1-4 alkyl.

173. The compound of any one of claims 149-165 and 169-172, wherein R10 is ethyl.

174. The compound of any one of claims 149-165 and 169-172, wherein R10 is isopropyl.

175. The compound of any one of claims 149-165, wherein R9 is H and R10 is C3-6 cycloalkyl.

176. The compound of any one of claims 149-165 and 175, wherein R10 is cyclopropyl.

177. The compound of any one of claims 149-165 and 175, wherein R10 is cyclopentyl.

178. The compound of any one of claims 149-177, wherein R11 is selected from C1-3 alkyl optionally substituted with one or more Rb, and C2-4 alkenyl.

179. The compound of any one of claims 149-177, wherein R11 is selected from C1-3 alkyl optionally substituted with one or more Rb, and C3-6 cycloalkyl.

180. The compound of any one of claims 149-179, wherein R11 is C1-3 alkyl optionally substituted with one or more Rb.

181. The compound of any one of claims 178-180, wherein Rb is ORc, and wherein Rc is selected from H and C1-3 alkyl.

182. The compound of claim 181, wherein Rc is H.

183. The compound of claim 181, wherein Rc is C1-3 alkyl.

184. The compound of any one of claims 149-179, wherein R11 is C1-3 alkyl.

185. The compound of any one of claims 149-179 and 184, wherein R11 is methyl.

186. The compound of any one of claims 149-177 and 179, wherein R11 is C3-6 cycloalkyl.

187. The compound of any one of claims 149-177, 179, and 186, wherein R11 is cyclopropyl.

188. The compound of any one of claims 149-187, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (R).

189. The compound of any one of claims 149-188, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

190. The compound of any one of claims 149-165, wherein each of R9 and R10 is halogen, and R11 is C1-3 alkyl optionally substituted with one or more Rb.

191. The compound of any one of claims 149-165 and 190, wherein R9 is fluoro, R10 is chloro, and R11 is C1-3 alkyl optionally substituted with one or more Rb.

192. The compound of any one of claims 149-165, 190, and 191, wherein R9 is fluoro, R10 is chloro, and R11 is methyl.

193. The compound of any one of claims 149-165 and 190-192, wherein R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

194. The compound of claim 149, wherein each of R1, R2, and R3 is H; R4 is H; R6 is C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen and ORa; R9 is fluoro, R10 is chloro, and R11 is methyl.

195. The compound of any one of claims 149 and 194, wherein each of R1, R2, and R3 is H; R4 is H; R6 is H or C1-6 alkyl; R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R is bonded to is (S).

196. A compound of Formula (IIB):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
R9 is selected from H and halogen;
R10 is selected from H, halogen, C1-4 alkyl, C1-4 haloalkyl, and C3-6 cycloalkyl;
R11 is selected from C1-3 alkyl, C2-4 alkenyl, and C3-6 cycloalkyl, wherein the C1-3 alkyl is optionally substituted with one or more Rb;
each Rb is independently selected from ORc, —C(O)ORc, and —(O)aryl, wherein the aryl is optionally substituted with one or more Rd;
each Rc is independently selected from hydrogen, C1-3 alkyl, OH, O(C1-3 alkyl), NO2, —C(O)aryl, and aryl; and
each Rd is C1-3 alkyl.

197. The compound of claim 196, wherein one of R9 and R10 is halogen and the other is H.

198. The compound of claim 196, wherein each of R9 and R10 is halogen.

199. The compound of any one of claims 196 and 198, wherein R9 is fluoro and R10 is chloro.

200. The compound of any one of claim 196, wherein R9 is H and R10 is C1-4 alkyl.

201. The compound of any one of claim 196, wherein R9 is halogen and R10 is C1-4 alkyl.

202. The compound of any one of claims 196 and 201, wherein R9 is chloro and R10 is C1-4 alkyl.

203. The compound of any one of claims 196 and 201, wherein R9 is fluoro and R10 is C1-4 alkyl.

204. The compound of any one of claims 196 and 200-203, wherein R10 is ethyl.

205. The compound of any one of claims 196 and 200-203, wherein R10 is isopropyl.

206. The compound of claim 196, wherein R9 is H and R10 is C3-6 cycloalkyl.

207. The compound of any one of claims 196 and 206, wherein R10 is cyclopropyl.

208. The compound of any one of claims 196 and 206, wherein R10 is cyclopentyl.

209. The compound of any one of claims 196-208, wherein R11 is selected from C1-3 alkyl optionally substituted with one or more Rb, and C2-4 alkenyl.

210. The compound of any one of claims 196-208, wherein R11 is selected from C1-3 alkyl optionally substituted with one or more Rb, and C3-6 cycloalkyl.

211. The compound of any one of claims 196-210, wherein R11 is C1-3 alkyl optionally substituted with one or more Rb.

212. The compound of any one of claims 196 and 209-211, wherein Rb is ORc, and wherein Rc is selected from H and C1-3 alkyl.

213. The compound of claim 212, wherein Rc is H.

214. The compound of claim 212, wherein Rc is C1-3 alkyl.

215. The compound of any one of claims 196-211, wherein R11 is C1-3 alkyl.

216. The compound of any one of claims 196-211 and 215, wherein R11 is methyl.

217. The compound of any one of claims 196-208 and 210, wherein R11 is C3-6 cycloalkyl.

218. The compound of any one of claims 196-208, 210, and 217, wherein R11 is cyclopropyl.

219. The compound of any one of claims 196-218, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (R).

220. The compound of any one of claims 196-219, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

221. The compound of claim 196, wherein each of R9 and R10 is halogen, and R11 is C1-3 alkyl optionally substituted with one or more Rb.

222. The compound of any one of claims 196 and 221, wherein R9 is fluoro, R10 is chloro, and R11 is C1-3 alkyl optionally substituted with one or more Rb.

223. The compound of any one of claims 196, 221, and 222, wherein R9 is fluoro, R10 is chloro, and R11 is methyl.

224. The compound of any one of claims 196 and 221-223, wherein R9 is fluoro, R10 is chloro, R11 is methyl, and wherein the Cahn-Ingold-Prelog configuration at the carbon atom R11 is bonded to is (S).

225. A compound of Formula (III):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
R1 is selected from H and C1-3 alkyl;
R2 is selected from H and C1-3 alkyl;
R3 is selected from H and C1-3 alkyl;
or R2 and R3 together with the nitrogen atoms to which they are attached and the carbon atom connecting the two nitrogen atoms form a 5- or 6-membered ring;
R4 is selected from H and C1-3 alkyl;
R5 is selected from H and C1-6 alkyl;
R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, and —SC(NH)NH2;
R7 is selected from H and C1-6 alkyl;
or R6 and R7 together with the carbon and nitrogen atoms to which they are attached form a ring having one of the formulas:
wherein the ring is optionally substituted on a carbon atom with C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more OH;
or R5 and R7 together with the carbon atoms to which they are attached and the nitrogen atom connecting the two carbon atoms form a ring having one of the formulas:
R8 is selected from H and halogen;
R12 is selected from H, halogen, and C1-6 alkyl;
R13 is selected from H, halogen, C1-6 alkyl, ORc, N(Rc)2, wherein C1-6 alkyl is optionally substituted with one or more of ORc and aryl;
R14 is selected from H and aryl;
or R13 and R14 together with the carbon atoms to which they are attached form a 5- or 6-membered cycloalkyl ring;
R15 is selected from H, C1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with one or more Rb;
each Ra is independently selected from H and C1-6 alkyl;
each Rb is independently selected from C2-6 alkenyl, ORc, N(Rc)2, —C(O)ORc, C3-6 cycloalkyl, and aryl;
each Rc is independently selected from hydrogen, C1-6 alkyl, aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
each Rd is independently selected from OH, O(C1-3 alkyl), NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

226. The compound of claim 225, wherein each of R1, R2, and R3 is H.

227. The compound of claim 225, wherein two of R1, R2, and R3 are H, and the other is C1-3 alkyl.

228. The compound of any one of claims 225 and 227, wherein two of R1, R2, and R3 are H, and the other is methyl.

229. The compound of claim 225, wherein one of R1, R2, and R3 is H, and the other two are C1-3 alkyl.

230. The compound of any one of claims 225 and 229, wherein one of R1, R2, and R3 is H, and the other two are methyl.

231. The compound of any one of claims 225-230, wherein R4 is H.

232. The compound of any one of claims 225-231, wherein R6 is selected from H or C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, or —SC(NH)NH2.

233. The compound of any one of claims 225-232, wherein R6 is H.

234. The compound of any one of claims 225-233, wherein R8 is H.

235. The compound of any one of claims 225-234, wherein each of R12 and R13 is H.

236. The compound of any one of claims 225-234, wherein each of R12 and R13 is not H.

237. The compound of any one of claims 225-234, wherein one of R12 and R13 is H, and the other is not H.

238. The compound of any one of claims 225-234 and 237, wherein R12 is H, and R13 is C1-6 alkyl optionally substituted with one or more of ORc or aryl.

239. The compound of any one of claims 225-234 and 237-238, wherein R12 is H, and R13 is C1-6 alkyl optionally substituted with OR.

240. The compound of any one of claims 225-234 and 237-239, wherein R12 is H, and R13 is C1-6 alkyl.

241. The compound of any one of claims 225-234 and 237-240, wherein R12 is H, and R13 is halogen.

242. The compound of any one of claims 225-234 and 237-241, wherein R12 is H, and R13 is fluoro.

243. The compound of any one of claims 225-234 and 236, wherein each of R12 and R13 is C1-6 alkyl.

244. The compound of any one of claims 225-234 and 236, wherein each of R12 and R13 is halogen.

245. The compound of any one of claims 225-234, 236, and 244, wherein each of R12 and R13 is fluoro.

246. The compound of any one of claims 225-234 and 237, wherein R12 is H, and R13 is OR.

247. The compound of any one of claims 225-234 and 237, wherein R12 is H, and R13 is N(Rc)2.

248. The compound of any one of claims 246 and 247, wherein each Rc is selected from H or C1-6 alkyl.

249. The compound of any one of claims 246 and 247, wherein each Rc is selected from C1-6 alkyl or —(CH2)aryl.

250. The compound of any one of claims 246, 247, and 248, wherein each Rc is H.

251. The compound of any one of claims 246 and 247-249, wherein each Rc is C1-6 alkyl.

252. The compound of any one of claims 246, 247-249, and 251, wherein each Rc is methyl.

253. The compound of any one of claims 246, 247, and 249, wherein each Rc is —(CH2)aryl.

254. The compound of any one of claim 247, wherein one Rc is H and the other Rc is C1-6 alkyl.

255. The compound of any one of claims 225-254, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (R).

256. The compound of any one of claims 225-254, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (S).

257. The compound of any one of claims 225-256, wherein R14 is H.

258. The compound of any one of claims 225-256, wherein R14 is aryl.

259. The compound of any one of claims 225-258, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R14 is bonded to is (R).

260. The compound of any one of claims 225-258, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R14 is bonded to is (S).

261. The compound of any one of claims 225-260, wherein R15 is H.

262. The compound of any one of claims 225-261, wherein R15 is C1-6 alkyl optionally substituted with one or more Rb.

263. The compound of any one of claims 225-261, wherein R15 is C1-6 alkyl optionally substituted with one Rb.

264. The compound of any one of claims 225-263, wherein R15 is C4-5 alkyl optionally substituted with one Rb.

265. The compound of any one of claims 262-264, wherein Rb is selected from ORc, N(Rc)2, and cyclopropyl.

266. The compound of any one of claims 262-265, wherein Rb is N(Rc)2.

267. The compound of any one of claims 262-266, wherein Rb is NH2.

268. A compound of Formula (IIIA):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
R1 is selected from H and C1-3 alkyl;
R2 is selected from H and C1-3 alkyl;
R3 is selected from H and C1-3 alkyl;
R4 is selected from H and C1-3 alkyl;
R6 is selected from H, C1-6 alkyl, and C2-6 alkenyl, wherein the C1-6 alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, and —SC(NH)NH2;
R8 is selected from H and halogen;
R12 is selected from H, halogen, and C1-6 alkyl;
R13 is selected from H, halogen, C1-6 alkyl, ORc, N(Rc)2, wherein C1-6 alkyl is optionally substituted with one or more of ORc and aryl;
R14 is selected from H and aryl;
or R13 and R14 together with the carbon atoms to which they are attached form a 5- or 6-membered cycloalkyl ring;
R16 is C1-6 alkyl;
each Ra is independently selected from H and C1-6 alkyl;
each Rc is independently selected from hydrogen, C1-6 alkyl, aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
each Rd is independently selected from OH, O(C1-3 alkyl), NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

269. The compound of claim 268, wherein each of R1, R2, and R3 is H.

270. The compound of claim 268, wherein two of R1, R2, and R3 are H, and the other is C1-3 alkyl.

271. The compound of any one of claims 268 and 270, wherein two of R1, R2, and R3 are H, and the other is methyl.

272. The compound of claim 268, wherein one of R1, R2, and R3 is H, and the other two are C1-3 alkyl.

273. The compound of any one of claims 268 and 272, wherein one of R1, R2, and R3 is H, and the other two are methyl.

274. The compound of any one of claims 268-273, wherein R4 is H.

275. The compound of any one of claims 268-274, wherein R6 is selected from H or C1-6 alkyl optionally substituted with one or more substituents selected from the group consisting of halogen, ORa, SRa, —C(O)ORa, or —SC(NH)NH2.

276. The compound of any one of claims 268-275, wherein R6 is H.

277. The compound of any one of claims 268-276, wherein R8 is H.

278. The compound of any one of claims 268-277, wherein each of R12 and R13 is H.

279. The compound of any one of claims 268-277, wherein each of R12 and R13 is not H.

280. The compound of any one of claims 268-277, wherein one of R12 and R13 is H, and the other is not H.

281. The compound of any one of claims 268-277, wherein R12 is H, and R13 is C1-6 alkyl optionally substituted with one or more of ORc or aryl.

282. The compound of any one of claims 268-277 and 281, wherein R12 is H, and R13 is C1-6 alkyl optionally substituted with ORc.

283. The compound of any one of claims 268-277, 281, and 282, wherein R12 is H, and R13 is C1-6 alkyl.

284. The compound of any one of claims 268-277 and 280, wherein R12 is H, and R13 is halogen.

285. The compound of any one of claims 268-277, 280, and 284, wherein R12 is H, and R13 is fluoro.

286. The compound of any one of claims 268-277 and 279, wherein each of R12 and R13 is C1-6 alkyl.

287. The compound of any one of claims 268-277 and 279, wherein each of R12 and R13 is halogen.

288. The compound of any one of claims 268-277 and 279, wherein each of R12 and R13 is fluoro.

289. The compound of any one of claims 268-277 and 280, wherein R12 is H, and R13 is OR.

290. The compound of any one of claims 268-277 and 280, wherein R12 is H, and R13 is N(Rc)2.

291. The compound of any one of claims 289 and 290, wherein each Rc is selected from H or C1-6 alkyl.

292. The compound of any one of claims 289 and 290, wherein Rc is selected from C1-6 alkyl or —(CH2)aryl.

293. The compound of any one of claims 289 and 290, wherein Rc is H.

294. The compound of any one of claims 289 and 290, wherein Rc is C1-6 alkyl.

295. The compound of any one of claims 289 and 290, wherein Rc is methyl.

296. The compound of any one of claims 289 and 290, wherein Rc is —(CH2)aryl.

297. The compound of claim 290, wherein one Rc is H and the other Rc is C1-6 alkyl.

298. The compound of any one of claims 268-297, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (R).

299. The compound of any one of claims 268-297, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (S).

300. The compound of any one of claims 268-299, wherein R14 is H.

301. The compound of any one of claims 268-299, wherein R14 is aryl.

302. The compound of any one of claims 268-301, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R14 is bonded to is (R).

303. The compound of any one of claims 268-301, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R14 is bonded to is (S).

304. The compound of any one of claims 268-303, wherein R16 is C1-6 alkyl.

305. The compound of any one of claims 268-304, wherein R16 is methyl.

306. The compound of any one of claims 268-305, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (S).

307. The compound of any one of claims 268-305, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (R).

308. A compound of Formula (IIIB):

or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, wherein:
R12 is selected from H, halogen, and C1-6 alkyl;
R13 is selected from H, halogen, C1-6 alkyl, ORc, N(Rc)2, wherein C1-6 alkyl is optionally substituted with one or more of ORc and aryl;
R16 is C1-6 alkyl;
each Rc is independently selected from hydrogen, C1-6 alkyl, aryl, and —(CH2)aryl, wherein the C1-6 alkyl and the aryl are each optionally substituted with one or more Rd; and
each Rd is independently selected from OH, O(C1-3 alkyl), NH2, NH(C1-3 alkyl), and N(C1-3 alkyl)2.

309. The compound of claim 308, wherein each of R12 and R13 is H.

310. The compound of claim 308, wherein each of R12 and R13 is not H.

311. The compound of claim 308, wherein one of R12 and R13 is H, and the other is not H.

312. The compound of any one of claims 308 and 311, wherein R12 is H, and R13 is C1-6 alkyl optionally substituted with one or more of ORc or aryl.

313. The compound of any one of claims 308 and 311-312, wherein R12 is H, and R13 is C1-6 alkyl optionally substituted with ORc.

314. The compound of any one of claims 308 and 311-313, wherein R12 is H, and R13 is C1-6 alkyl.

315. The compound of any one of claims 308 and 311, wherein R12 is H, and R13 is halogen.

316. The compound of any one of claims 308, 311, 315, wherein R12 is H, and R13 is fluoro.

317. The compound of any one of claims 308 and 310, wherein each of R12 and R13 is C1-6 alkyl.

318. The compound of any one of claims 308 and 310, wherein each of R12 and R13 is halogen.

319. The compound of any one of claims 308, 310, and 318, wherein each of R12 and R13 is fluoro.

320. The compound of any one of claims 308 and 311, wherein R12 is H, and R13 is ORc.

321. The compound of any one of claims 308 and 311, wherein R12 is H, and R13 is N(Rc)2.

322. The compound of any one of claims 320 and 321, wherein each Rc is selected from H or C1-6 alkyl.

323. The compound of any one of claims 320 and 321, wherein each Rc is selected from C1-6 alkyl or —(CH2)aryl.

324. The compound of any one of claims 320 and 321, wherein each Rc is H.

325. The compound of any one of claims 320 and 321, wherein each Rc is C1-6 alkyl.

326. The compound of any one of claims 320 and 321, wherein each Rc is methyl.

327. The compound of any one of claims 320 and 321, wherein each Rc is —(CH2)aryl.

328. The compound of claim 321, wherein one Rc is H and the other Rc is C1-6 alkyl.

329. The compound of any one of claims 308-328, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (R).

330. The compound of any one of claims 308-328, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R13 is bonded to is (S).

331. The compound of any one of claims 308-330, wherein R16 is C1-6 alkyl.

332. The compound of any one of claims 308-331, wherein R16 is methyl.

333. The compound of any one of claims 308-332, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (S).

334. The compound of any one of claims 308-332, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (R).

335. The compound of claim 308, wherein one of R12 and R13 is H, and the other of R12 and R13 is not H; and R16 is methyl, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (S).

336. The compound of any one of claims 308 and 335, wherein R12 is H, and R13 is ORc; Rc is selected from H or C1-6 alkyl; and R16 is methyl, wherein the Cahn-Ingold-Prelog configuration at the carbon atom R16 is bonded to is (S).

337. A compound selected from any one of the compounds listed in Table 1, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer.

338. A pharmaceutical composition comprising a compound of any one of claims 1-337, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, and a pharmaceutically acceptable carrier.

339. A method of treating a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1-337, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition of claim 338.

340. A method of preventing a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1-337, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition of claim 338.

341. A method of reducing the risk of a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1-337, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition of claim 338.

342. A method of delaying the onset of a microbial infection comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1-337, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, or a pharmaceutically acceptable composition of claim 338.

343. Use of a compound according to any one of claims 1-337, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, in the manufacture of a medicament for treating, preventing, or reducing a microbial infection in a subject.

344. A compound according to any one of claims 1-337, or a tautomer thereof or a pharmaceutically acceptable salt of the compound or tautomer, for use in treating, preventing, or reducing a microbial infection in a subject.

Patent History
Publication number: 20210147422
Type: Application
Filed: Jun 15, 2018
Publication Date: May 20, 2021
Inventors: Erin M. Duffy (Deep River, CT), Ashoke Bhattacharjee (Cheshire, CT), Zoltan F. Kanyo (North Haven, CT), Joseph A. Ippolito (Guilford, CT), Andrea Marra (New Haven, CT)
Application Number: 16/622,969
Classifications
International Classification: C07D 487/04 (20060101); A61P 31/04 (20060101);