Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
RNA VIRUS INHIBITOR COMPOUNDS WITH IMPROVED METABOLIC STABILITY AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2023/235970
Kind Code:
A1
Abstract:
The present disclosure provides compounds with increased metabolic stability for inhibiting a virus infection, such as a Baltimore Group IV RNA virus infection, such as rhinovirus, coxsackievirus, norovirus and coronavirus. Aspects of the present disclosure also include methods of treating the virus infection in a subject with compounds with increased metabolic stability.

Inventors:
NIEMAN JAMES A (CA)
LEMIEUX M JOANNE (CA)
ARUTYUNOVA ELENA (CA)
JOYCE MICHAEL A (CA)
TYRRELL D LORNE (CA)
SAFFRAN HOLLY (CA)
BAI BING (CA)
HENA MOSTOFA (CA)
KANDADAI APPAN SRINIVAS (CA)
Application Number:
PCT/CA2023/050778
Publication Date:
December 14, 2023
Filing Date:
June 07, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV ALBERTA (CA)
International Classes:
C07K5/08; A61K38/06; A61P31/14; C07K5/097
Domestic Patent References:
WO2005113580A12005-12-01
WO2022020711A12022-01-27
Other References:
ROBERT L. HOFFMAN ET AL.: "Discovery of Ketone-Based Covalent Inhibitors of Coronavirus 3CL Proteases for the Potential Therapeutic Treatment of COVID-19.", J. MED. CHEM., vol. 63, 15 October 2020 (2020-10-15), pages 12725 - 12747, XP055803394, DOI: 10.1021/acs.jmedchem.0c01063
BAI BING, BELOVODSKIY ALEXANDR, HENA MOSTOFA, KANDADAI APPAN SRINIVAS, JOYCE MICHAEL A, SAFFRAN HOLLY A, SHIELDS JUSTIN A, KHAN MU: "Peptidomimetic α-Acyloxymethylketone Warheads with Six-Membered Lactam P1 Glutamine Mimic: SARS-CoV-2 3CL Protease Inhibition, Coronavirus Antiviral Activity, and in Vitro Biological Stability", JOURNAL OF MEDICINAL CHEMISTRY, 9 July 2021 (2021-07-09), XP055826782, Retrieved from the Internet [retrieved on 20210722], DOI: 10.1021/acs.jmedchem.1c00616
Attorney, Agent or Firm:
SMART & BIGGAR LP (CA)
Download PDF:
Claims:
CLAIMS WHAT IS CLAIMED IS: 1. A compound of formula (I): wherein: each R1 is independently selected from –H, –F, –Cl and –CH3; R2 is selected from –Cl and –F; R3 is selected from –CH2CH3, –CH(CH3)2, –C(CH3)3, –CF(CH3)2, –CF2CH3, –CH(CF3)CH3, –CH2CCl2H, –CH2CF3, –CH(CF3)2, cyclopropyl and cyclohexyl; R4 is selected from –H, –P(=O)(OH)2, –C(=O)CH(NH2)CH(CH3)2 and – C(=O)CH2NH2; X is selected from –CH2–, –CDH– and –CD2–; and Y is –CH2– or is absent; or a pharmaceutically acceptable salt, solvate, or hydrate thereof. 2. The compound of Claim 1, wherein each R1 is independently selected from –H and –F. 3. The compound of Claim 1 or 2, wherein R2 is –F. 4. The compound of Claim 1 or 2, wherein R2 is –Cl. 5. The compound of any one of Claims 1 to 4, wherein R3 is selected from –CH(CH3)2, –C(CH3)3, –CF(CH3)2, –CF2CH3, –CH(CF3)CH3,–CH2CF3, and cyclopropyl. 6. The compound of Claim 5, wherein R3 is –CH(CH3)2.

7. The compound of Claim 5, wherein R3 is –CF2CH3. 8. The compound of Claim 5, wherein R3 is –CF(CH3)2. 9. The compound of Claim 5, wherein R3 is cyclopropyl. 10. The compound of any one of Claims 1 to 9, wherein R4 is selected from –H, – P(=O)(OH)2, and –C(=O)CH(NH2)CH(CH3)2. 11. The compound of Claim 10, wherein R4 is –C(=O)CH(NH2)CH(CH3)2. 12. The compound of Claim 10, wherein R4 is –H. 13. The compound of any one of Claims 1 to 12, wherein Y is absent. 14. The compound of any one of Claims 1 to 12, wherein Y is –CH2–. 15. The compound of any one of Claims 1 to 14, wherein X is –CH2– or –CD2–. 16. The compound of Claims 15, wherein X is –CH2–. 17. The compound of Claim 1, wherein the compound is selected from:

18. The compound of Claim 1, wherein the compound is selected from:

19. A method of inhibiting a Baltimore Group IV RNA virus in a cell infected with a Baltimore Group IV RNA virus, the method comprising contacting the cell with a compound of any one of Claims 1 to 18.

20. The method of Claim 19, wherein the Baltimore Group IV RNA virus is selected from the family of Picornaviridae, Calciviridae and Coronaviridae.

21. The method of Claim 20, wherein the Baltimore Group IV RNA virus is selected from rhinovirus, coxsackievirus, norovirus and coronavirus.

22. The method of Claim 21, wherein the Baltimore Group IV RNA virus is coronavirus.

23. The method of Claim 22, wherein the coronavirus is one that causes disease in mammals.

24. The method of Claim 23, wherein the coronavirus causes disease in companion animals or livestock.

25. The method of Claim 24, wherein the coronavirus is a feline coronavirus.

26. The method of Claim 25, wherein the coronavirus is feline infectious peritonitis.

27. The method of Claim 23, wherein the coronavirus is a human coronavirus.

28. The method of Claim 27, wherein the coronavirus is selected from Severe Acute

Respiratory Syndrome coronavirus 2 (SARS-CoV-2), Severe Acute Respiratory syndrome coronavirus 1 (SARS-CoV-1) and Middle Eastern Respiratory syndrome-related coronavirus (MERS-CoV).

29. A method of treating a Baltimore Group IV RNA virus infection in a mammal, the method comprising administering to the mammal an effective amount of a compound according to any one of Claims 1 to 18.

30. The method of Claim 29, wherein the mammal is selected from a companion animal and livestock.

31. The method of Claim 30, wherein the mammal is a feline.

32. The method of Claim 29, wherein the mammal is a human.

33. The method of Claim 29, wherein the Baltimore Group IV RNA virus is selected from rhinovirus, coxsackievirus, norovirus and coronavirus.

34. The method of Claim 33, wherein the Baltimore Group IV RNA virus is selected from norovirus, and coronavirus.

35. The method of Claim 34, wherein the Baltimore Group IV RNA virus is human norovirus.

36. The method of Claim 34, wherein the Baltimore Group IV RNA virus is a coronavirus that causes disease in mammals.

37. The method of Claim 36, wherein the coronavirus is a feline coronavirus.

38. The method of Claim 37, wherein the feline coronavirus is feline infectious peritonitis.

39. The method of Claim 36, wherein the coronavirus is a human coronavirus.

40. The method of Claim 39, wherein the human coronavirus is selected from Severe Acute

Respiratory Syndrome coronavirus 2 (SARS-CoV-2), Severe Acute Respiratory syndrome coronavirus 1 (SARS-CoV-1) and Middle Eastern Respiratory syndrome-related coronavirus (MERS-CoV).

Description:
RNA VIRUS INHIBITOR COMPOUNDS WITH IMPROVED METABOLIC STABILITY AND USES THEREOF CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of priority to U.S. Provisional Application No. 63/350,545, filed June 9, 2022, and U.S. Provisional Application No. 63/433,339, filed December 16, 2022, the disclosures of each of which are incorporated herein by reference. INTRODUCTION [0002] Ribonucleic acid (RNA) viruses have genomes made of RNA. RNA viruses may be categorized based on their genetic material by the Baltimore classification strategy. The groups include, for example, double-stranded RNA (dsRNA) viruses (Group III), positive sense single-stranded RNA viruses (+ssRNA) viruses (Group IV), and negative sense single-stranded RNA (-ssRNA) viruses (Group V). Single-stranded RNA (ssRNA) viruses cause many diseases in wildlife, domestic animals and humans. These viruses are genetically and antigenically diverse, exhibiting broad tissue tropisms and a wide pathogenic potential. The incubation periods of some of the most pathogenic viruses, e.g. the caliciviruses, are very short. Viral replication and expression of virulence factors may overwhelm early defense mechanisms (Xu 1991) and cause acute and severe symptoms. [0003] Group IV RNA viruses contain a single strand of viral mRNA (also known as a positive/plus strand of genomic RNA). Positive sense RNA can be translated directly into protein, without a DNA intermediate and without creating a complementary RNA strand. The positive strand RNA genome is independently infectious, for most Group IV viruses. This means that in the absence of a capsid, envelope, or enclosed proteins, the RNA molecule, when inserted into a cell, is capable of using host cell machinery to construct additional viruses. Six subclasses of the Group IV single-stranded positive-sense RNA viruses include: Picornaviridae, Togaviridae, Coronaviridae, Hepeviridae, Caliciviridae, Flaviviridae, and Astroviridae (Berman (2012) Taxonomic Guide to Infectious Diseases.237-246.). [0004] Coronaviruses are a group of enveloped positive-sense single-stranded RNA viruses that are members of the Coronaviridae family, which are members of Group IV viruses. Since the turn of the millennium, three closely related coronaviruses have infected humans and spread internationally: the 2003 epidemic of Severe Acute Respirator Syndrome (SARS), 2012 Middle East respiratory syndrome (MERS) outbreak and the current Coronavirus Disease 2019 (COVID-19) pandemic. In each instance, these coronaviruses are thought to have originated from an animal reservoir and then ‘jumped’ to humans either directly or through an intermediate species. COVID-19 is caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2 or SARS2). [0005] Noroviruses are a group of non-enveloped, positive-sense single-stranded RNA viruses that are members of the Caliciviridae family, which are members of Group IV viruses. Noroviruses is the most common cause of gastroenteritis and cases result in approximately 200,000 deaths globally per year. [0006] Rhinoviruses have single-stranded positive sense RNA genomes and are not enveloped. They are members of the Picornaviridae family, which are members of Group IV viruses. Rhinoviruses are a predominant cause of the common cold. Rhinoviruses belong to the genus Enterovirus. [0007] Coxsackieviruses are non-enveloped, positive-sense single-stranded RNA viruses that are members of the Picornaviridae family, which are members of Group IV viruses. Coxsackieviruses cause a variety of infections and are among the leading cause of aseptic meningitis. Coxsackieviruses belong to the genus Enterovirus. [0008] To allow administration of a therapeutic compound in a mammal, including humans, the compound should have acceptable pharmacokinetics to reach the site of action with high enough concentration and persist long enough to provide the desired therapeutic outcome. To achieve this the compound should have numerous acceptable parameters to obtain high enough exposure to be efficacious. Examples of such parameters include permeability, solubility and metabolic stability. Identifying compounds with good to excellent metabolic stability is challenging as numerous metabolizing enzymes act on xenobiotics and the metabolizing enzymes can vary depending on structure and physicochemical properties of the compound. One method in vitro to determine metabolic stability is to assess the compound’s stability in the presence of mammalian hepatocytes or microsomes. This provides an indication of how readily the compound is metabolised by the metabolic enzymes in the liver of the mammal. Compounds with low half-lives in the presence of hepatocytes or microsomes will have rapid clearance and low bioavailability resulting from the facile metabolism. Compounds with rapid metabolism will not obtain and sustain high enough concentration to be efficacious in vivo. Adding a metabolism blocker, such as ritonavir that blocks oxidative metabolism by cytochrome P450, can allow increased absorption and reduced clearance to allow efficacy to be obtained in vivo. One example of this is Paxlovid ® , which contains nirmatrelvir and ritonavir. The active ingredient nirmatrelvir relies on the reduced metabolism caused by ritonavir to increase and maintain higher plasma concentrations of the active ingredient nirmatrelvir. However, co-dosing with ritonavir can also affect the metabolism of other xenobiotics, which can lead to severe drug-drug interactions. Finding compounds with higher metabolic stability that do not require co-dosing with a metabolism blocker, such as ritonavir, can significantly reduce the risk of drug-drug interactions for the mammal, including humans, that are taking other medications. Compounds with higher stability may not require co-administration with a metabolism blocker, such as ritonavir, and are more likely to be suitable for combination therapies, for example to treat a viral infection. Combination therapies can prove advantageous to reduce resistance development and have been successfully used to treat HIV and HCV. Compounds that are active against Group IV viruses with improved metabolic stability have promise to treat disease as a mono- or combination therapy. SUMMARY [0009] The present disclosure provides compounds with increased metabolic stability for inhibiting a virus infection, such as a Baltimore Group IV RNA virus infection, such as rhinovirus, coxsackievirus, norovirus and coronavirus. Aspects of the present disclosure also include methods of treating the virus infection in a subject with compounds with increased metabolic stability. [0010] Aspects of the present disclosure include a compound of formula (I): wherein: each R 1 is independently selected from –H, –F, –Cl and –CH 3 ; R 2 is selected from –Cl and –F; R 3 is selected from –CH 2 CH 3 , –CH(CH 3 ) 2 , –C(CH 3 ) 3 , –CF(CH 3 ) 2 , –CF 2 CH 3 , –CH(CF 3 )CH 3 , –CH 2 CCl 2 H, –CH 2 CF 3 , –CH(CF 3 ) 2 , cyclopropyl and cyclohexyl; R 4 is selected from –H, –P(=O)(OH) 2 , –C(=O)CH(NH 2 )CH(CH 3 ) 2 and – C(=O)CH 2 NH 2 ; X is selected from –CH 2 –, –CDH– and –CD 2 –; and Y is –CH 2 – or is absent; or a pharmaceutically acceptable salt, solvate, or hydrate thereof. [0011] In some embodiments, each R 1 is independently selected from –H and –F. [0012] In some embodiments, R 2 is –F. In some embodiments, R 2 is –Cl. [0013] In some embodiments, R 3 is selected from –CH(CH 3 ) 2 , –C(CH 3 ) 3 , –CF(CH 3 ) 2 , –CF 2 CH 3 , –CH(CF 3 )CH 3 ,–CH 2 CF 3 , and cyclopropyl. In some embodiments, R 3 is –CH(CH 3 ) 2 . In some embodiments, R 3 is –C(CH 3 ) 3 . In some embodiments, R 3 is –CF(CH 3 ) 2 . In some embodiments, R 3 is –CF 2 CH 3 . In some embodiments, R 3 is cyclopropyl. [0014] In some embodiments, R 4 is selected from –H, –P(=O)(OH) 2 , and – C(=O)CH(NH 2 )CH(CH 3 ) 2 . In some embodiments, R 4 is –C(=O)CH(NH 2 )CH(CH 3 ) 2 . In some embodiments, R 4 is –H. [0015] In some embodiments, Y is absent. In some embodiments, Y is –CH 2 –. [0016] In some embodiments, X is –CH 2 – or –CD 2 –. In some embodiments, X is – CH 2 –. [0017] In some embodiments, the compound is selected from the following structures:

[0018] In some embodiments, the compound is selected from the following structures:

[0019] Aspects of the present disclosure include a method of inhibiting a Baltimore Group IV RNA virus in a cell infected with a Baltimore Group IV RNA virus, the method comprising contacting the cell with a compound according to the present disclosure. [0020] In some embodiments, the Baltimore Group IV RNA virus is selected from the family of Picornaviridae, Calciviridae and Coronaviridae. [0021] In some embodiments, the Baltimore Group IV RNA virus is selected from rhinovirus, coxsackievirus, norovirus and coronavirus. [0022] In some embodiments, the Baltimore Group IV RNA virus is coronavirus. [0023] In some embodiments, the coronavirus is one that causes disease in mammals. In some embodiments, the coronavirus causes disease in companion animals or livestock. In some embodiments, the coronavirus is a feline coronavirus. In some embodiments, the coronavirus is feline infectious peritonitis. In some embodiments, the coronavirus is a human coronavirus. [0024] In some embodiments, the coronavirus is selected from Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), Severe Acute Respiratory syndrome coronavirus 1 (SARS-CoV-1) and Middle Eastern Respiratory syndrome-related coronavirus (MERS-CoV). [0025] Aspects of the present disclosure include a method of treating a Baltimore Group IV RNA virus infection in a mammal, the method comprising administering to the mammal an effective amount of a compound according to the present disclosure. [0026] In some embodiments, the mammal is selected from a companion animal and livestock. In some embodiments, the mammal is a feline. In some embodiments, the mammal is a human. [0027] In some embodiments, the Baltimore Group IV RNA virus is selected from rhinovirus, coxsackie virus, norovirus and coronavirus. [0028] In some embodiments, the Baltimore Group IV RNA virus is selected from norovirus, and coronavirus. [0029] In some embodiments, the Baltimore Group IV RNA virus is human norovirus. [0030] In some embodiments, the Baltimore Group IV RNA virus is a coronavirus that causes disease in mammals. [0031] In some embodiments, the coronavirus is a feline coronavirus. In some embodiments, the feline coronavirus is feline infectious peritonitis. [0032] In some embodiments, the coronavirus is a human coronavirus. In some embodiments, the human coronavirus is selected from Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), Severe Acute Respiratory syndrome coronavirus 1 (SARS-CoV- 1) and Middle Eastern Respiratory syndrome-related coronavirus (MERS-CoV). BRIEF DESCRIPTION OF THE DRAWINGS [0033] FIGS. 1A and 1B show exemplary concentration response curves for Compound 1 and Compound 2, respectively, screened for SARS-CoV-23CLP inhibition using a Fluorescence resonance energy transfer (FRET) assay, according to embodiments of the present disclosure. [0034] FIGS. 2A to 2F show exemplary percent remaining over time curves for select compounds in the presence of human microsomes, according to embodiments of the present disclosure. DEFINITIONS [0035] The following terms have the following meanings unless otherwise indicated. Any undefined terms have their art recognized meanings. [0036] “Alkyl” refers to monovalent saturated aliphatic hydrocarbyl groups having from 1 to 10 carbon atoms and such as 1 to 6 carbon atoms, or 1 to 5, or 1 to 4, or 1 to 3 carbon atoms. This term includes, by way of example, linear and branched hydrocarbyl groups such as methyl (CH 3 -), ethyl (CH 3 CH 2 -), n-propyl (CH 3 CH 2 CH 2 -), isopropyl ((CH 3 ) 2 CH-), n-butyl (CH 3 CH 2 CH 2 CH 2 -), isobutyl ((CH 3 ) 2 CHCH 2 -), sec-butyl ((CH 3 )(CH 3 CH 2 )CH-), t-butyl ((CH 3 ) 3 C-), n-pentyl (CH 3 CH 2 CH 2 CH 2 CH 2 -), and neopentyl ((CH 3 ) 3 CCH 2 -). [0037] The term “substituted alkyl” refers to an alkyl group as defined herein wherein one or more carbon atoms in the alkyl chain (except the C1 carbon atom) have been optionally replaced with a heteroatom such as -O-, -N-, -S-, -S(O) n - (where n is 0 to 2), -NR- (where R is hydrogen or alkyl) and having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, - SO-alkyl, -SO-aryl, -SO-heteroaryl, -SO 2 -alkyl, -SO 2 -aryl, -SO 2 -heteroaryl, and -NR a R b , wherein R a and R b may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic. [0038] “Alkylene” refers to divalent aliphatic hydrocarbyl groups preferably having from 1 to 6 and more preferably 1 to 3 carbon atoms that are either straight-chained or branched, and which are optionally interrupted with one or more groups selected from -O-, -NR 10 - , -NR 10 C(O)-, -C(O)NR 10 - and the like, where R 10 is chosen from chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic. This term includes, by way of example, methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), n-propylene (-CH 2 CH 2 CH 2 -), iso-propylene (-CH 2 CH(CH 3 )-), (-C(CH 3 ) 2 CH 2 CH 2 -), (-C(CH 3 ) 2 CH 2 C(O)-), (-C(CH 3 ) 2 CH 2 C(O)NH-), (-CH(CH 3 )CH 2 -), and the like. [0039] “Substituted alkylene” refers to an alkylene group having from 1 to 3 hydrogens replaced with substituents as described for carbons in the definition of “substituted” below. [0040] The term “alkane” refers to alkyl group and alkylene group, as defined herein. [0041] The term “alkylaminoalkyl”, “alkylaminoalkenyl” and “alkylaminoalkynyl” refers to the groups R’NHR”- where R’ is alkyl group as defined herein and R” is alkylene, alkenylene or alkynylene group as defined herein. [0042] The term “alkaryl” or “aralkyl” refers to the groups -alkylene-aryl and -substituted alkylene-aryl where alkylene, substituted alkylene and aryl are defined herein. [0043] “Alkoxy” refers to the group –O-alkyl, wherein alkyl is as defined herein. Alkoxy includes, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, t-butoxy, sec-butoxy, n-pentoxy, and the like. The term “alkoxy” also refers to the groups alkenyl-O-, cycloalkyl-O-, cycloalkenyl-O-, and alkynyl-O-, where alkenyl, cycloalkyl, cycloalkenyl, and alkynyl are as defined herein. [0044] The term “substituted alkoxy” refers to the groups substituted alkyl-O-, substituted alkenyl-O-, substituted cycloalkyl-O-, substituted cycloalkenyl-O-, and substituted alkynyl-O- where substituted alkyl, substituted alkenyl, substituted cycloalkyl, substituted cycloalkenyl and substituted alkynyl are as defined herein. [0045] The term “alkoxyamino” refers to the group –NH-alkoxy, wherein alkoxy is defined herein. [0046] The term “haloalkoxy” refers to the groups alkyl-O- wherein one or more hydrogen atoms on the alkyl group have been substituted with a halo group and include, by way of examples, groups such as trifluoromethoxy, and the like. [0047] The term “haloalkyl” refers to a substituted alkyl group as described above, wherein one or more hydrogen atoms on the alkyl group have been substituted with a halo group. Examples of such groups include, without limitation, fluoroalkyl groups, such as trifluoromethyl, difluoromethyl, trifluoroethyl and the like. [0048] The term “alkylalkoxy” refers to the groups -alkylene-O-alkyl, alkylene-O- substituted alkyl, substituted alkylene-O-alkyl, and substituted alkylene-O-substituted alkyl wherein alkyl, substituted alkyl, alkylene and substituted alkylene are as defined herein. [0049] “Alkenyl” refers to straight chain or branched hydrocarbyl groups having from 2 to 6 carbon atoms and preferably 2 to 4 carbon atoms and having at least 1 and preferably from 1 to 2 sites of double bond unsaturation. This term includes, by way of example, bi-vinyl, allyl, and but-3-en-1-yl. Included within this term are the cis and trans isomers or mixtures of these isomers. [0050] The term “substituted alkenyl” refers to an alkenyl group as defined herein having from 1 to 5 substituents, or from 1 to 3 substituents, selected from alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, - SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO 2 -alkyl, -SO 2 -substituted alkyl, -SO 2 -aryl and -SO 2 -heteroaryl. [0051] “Alkynyl” refers to straight or branched monovalent hydrocarbyl groups having from 2 to 6 carbon atoms and preferably 2 to 3 carbon atoms and having at least 1 and preferably from 1 to 2 sites of triple bond unsaturation. Examples of such alkynyl groups include acetylenyl (-C≡CH), and propargyl (-CH 2 C≡CH). [0052] The term “substituted alkynyl” refers to an alkynyl group as defined herein having from 1 to 5 substituents, or from 1 to 3 substituents, selected from alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, - SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO 2 -alkyl, -SO 2 -substituted alkyl, -SO 2 -aryl, and -SO 2 -heteroaryl. [0053] “Alkynyloxy” refers to the group –O-alkynyl, wherein alkynyl is as defined herein. Alkynyloxy includes, by way of example, ethynyloxy, propynyloxy, and the like. [0054] “Acyl” refers to the groups H-C(O)-, alkyl-C(O)-, substituted alkyl-C(O)-, alkenyl- C(O)-, substituted alkenyl-C(O)-, alkynyl-C(O)-, substituted alkynyl-C(O)-, cycloalkyl-C(O)-, substituted cycloalkyl-C(O)-, cycloalkenyl-C(O)-, substituted cycloalkenyl-C(O)-, aryl-C(O)-, substituted aryl-C(O)-, heteroaryl-C(O)-, substituted heteroaryl-C(O)-, heterocyclyl-C(O)-, and substituted heterocyclyl-C(O)-, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. For example, acyl includes the “acetyl” group CH 3 C(O)- [0055] “Acylamino” refers to the groups –NR 20 C(O)alkyl, -NR 20 C(O)substituted alkyl, N R 20 C(O)cycloalkyl, -NR 20 C(O)substituted cycloalkyl, - NR 20 C(O)cycloalkenyl, -NR 20 C(O)substituted cycloalkenyl, -NR 20 C(O)alkenyl, - NR 20 C(O)substituted alkenyl, -NR 20 C(O)alkynyl, -NR 20 C(O)substituted alkynyl, -NR 20 C(O)aryl, -NR 20 C(O)substituted aryl, -NR 20 C(O)heteroaryl, -NR 20 C(O)substituted heteroaryl, -NR 20 C(O)heterocyclic, and -NR 20 C(O)substituted heterocyclic, wherein R 20 is hydrogen or alkyl and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0056] “Aminocarbonyl” or the term “aminoacyl” refers to the group -C(O)NR 21 R 22 , wherein R 21 and R 22 independently are selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 21 and R 22 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0057] “Aminocarbonylamino” refers to the group –NR 21 C(O)NR 22 R 23 where R 21 , R 22 , and R 23 are independently selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 21 and R 22 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0058] The term “alkoxycarbonylamino” refers to the group -NR d C(O)OR d where each R d is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclyl wherein alkyl, substituted alkyl, aryl, heteroaryl, and heterocyclyl are as defined herein. [0059] The term “acyloxy” refers to the groups alkyl-C(O)O-, substituted alkyl-C(O)O-, cycloalkyl-C(O)O-, substituted cycloalkyl-C(O)O-, aryl-C(O)O-, heteroaryl-C(O)O-, and heterocyclyl-C(O)O- wherein alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, heteroaryl, and heterocyclyl are as defined herein. [0060] “Aminosulfonyl” refers to the group –SO 2 NR 21 R 22 , wherein R 21 and R 22 independently are selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 21 and R 22 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0061] “Sulfonylamino” refers to the group –NR 21 SO 2 R 22 , wherein R 21 and R 22 independently are selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 21 and R 22 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0062] “Aryl” or “Ar” refers to a monovalent aromatic carbocyclic group of from 6 to 18 carbon atoms having a single ring (such as is present in a phenyl group) or a ring system having multiple condensed rings (examples of such aromatic ring systems include naphthyl, anthryl and indanyl) which condensed rings may or may not be aromatic, provided that the point of attachment is through an atom of an aromatic ring. This term includes, by way of example, phenyl and naphthyl. Unless otherwise constrained by the definition for the aryl substituent, such aryl groups can optionally be substituted with from 1 to 5 substituents, or from 1 to 3 substituents, selected from acyloxy, hydroxy, thiol, acyl, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, substituted cycloalkyl, substituted cycloalkenyl, amino, substituted amino, aminoacyl, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halogen, nitro, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO 2 -alkyl, -SO 2 -substituted alkyl, -SO 2 -aryl, -SO 2 -heteroaryl and trihalomethyl. [0063] “Aryloxy” refers to the group –O-aryl, wherein aryl is as defined herein, including, by way of example, phenoxy, naphthoxy, and the like, including optionally substituted aryl groups as also defined herein. [0064] “Amino” refers to the group –NH 2 . [0065] The term “substituted amino” refers to the group -NR m R m where each R m is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, and heterocyclyl provided that at least one R is not hydrogen. [0066] The term “azido” refers to the group –N 3 . [0067] “Carboxyl,” “carboxy” or “carboxylate” refers to –CO 2 H or salts thereof. [0068] “Carboxyl ester” or “carboxy ester” or the terms “carboxyalkyl” or “carboxylalkyl” refers to the groups -C(O)O-alkyl, -C(O)O-substituted alkyl, -C(O)O-alkenyl, -C(O)O-substituted alkenyl, -C(O)O-alkynyl, -C(O)O-substituted alkynyl, -C(O)O-aryl, -C(O)O-substituted aryl, -C(O)O-cycloalkyl, -C(O)O-substituted cycloalkyl, -C(O)O-cycloalkenyl, -C(O)O-substituted cycloalkenyl, -C(O)O-heteroaryl, -C(O)O-substituted heteroaryl, -C(O)O-heterocyclic, and -C(O)O-substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0069] “(Carboxyl ester)oxy” or “carbonate” refers to the groups –O-C(O)O- alkyl, -O-C(O)O-substituted alkyl, -O-C(O)O-alkenyl, -O-C(O)O-substituted alkenyl, -O- C(O)O-alkynyl, -O-C(O)O-substituted alkynyl, -O-C(O)O-aryl, -O-C(O)O-substituted aryl, - O-C(O)O-cycloalkyl, -O-C(O)O-substituted cycloalkyl, -O-C(O)O-cycloalkenyl, -O-C(O)O- substituted cycloalkenyl, -O-C(O)O-heteroaryl, -O-C(O)O-substituted heteroaryl, -O-C(O)O- heterocyclic, and -O-C(O)O-substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0070] “Cyano” or “nitrile” refers to the group –CN. [0071] “Cycloalkyl” refers to cyclic alkyl groups of from 3 to 10 carbon atoms having single or multiple cyclic rings including fused, bridged, and spiro ring systems. Examples of suitable cycloalkyl groups include, for instance, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl and the like. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, and the like, or multiple ring structures such as adamantanyl, and the like. [0072] The term “substituted cycloalkyl” refers to cycloalkyl groups having from 1 to 5 substituents, or from 1 to 3 substituents, selected from alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO 2 -alkyl, -SO 2 -substituted alkyl, -SO 2 -aryl and -SO 2 -heteroaryl. [0073] “Cycloalkenyl” refers to non-aromatic cyclic alkyl groups of from 3 to 10 carbon atoms having single or multiple rings and having at least one double bond and preferably from 1 to 2 double bonds. [0074] The term “substituted cycloalkenyl” refers to cycloalkenyl groups having from 1 to 5 substituents, or from 1 to 3 substituents, selected from alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO 2 -alkyl, -SO 2 -substituted alkyl, -SO 2 -aryl and -SO 2 -heteroaryl. [0075] “Cycloalkynyl” refers to non-aromatic cycloalkyl groups of from 5 to 10 carbon atoms having single or multiple rings and having at least one triple bond. [0076] “Carbocycle” refers to non-aromatic or aromatic cyclic groups, such as cycloalkyl, cycloalkenyl, cycloalkynyl, and aryl groups as defined herein. A carbocycle goup may be unsubstituted or substituted as defined herein. [0077] “Cycloalkoxy” refers to –O-cycloalkyl. [0078] “Cycloalkenyloxy” refers to –O-cycloalkenyl. [0079] “Halo” or “halogen” refers to fluoro, chloro, bromo, and iodo. [0080] “Hydroxy” or “hydroxyl” refers to the group –OH. [0081] “Heteroaryl” refers to an aromatic group of from 1 to 15 carbon atoms, such as from 1 to 10 carbon atoms and 1 to 10 heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur within the ring. Such heteroaryl groups can have a single ring (such as, pyridinyl, imidazolyl or furyl) or multiple condensed rings in a ring system (for example as in groups such as, indolizinyl, quinolinyl, benzofuran, benzimidazolyl or benzothienyl), wherein at least one ring within the ring system is aromatic. To satisfy valence requirements, any heteroatoms in such heteroaryl rings may or may not be bonded to H or a substituent group, e.g., an alkyl group or other substituent as described herein. In certain embodiments, the nitrogen and/or sulfur ring atom(s) of the heteroaryl group are optionally oxidized to provide for the N-oxide (N→O), sulfinyl, or sulfonyl moieties. This term includes, by way of example, pyridinyl, pyrrolyl, indolyl, thiophenyl, and furanyl. Unless otherwise constrained by the definition for the heteroaryl substituent, such heteroaryl groups can be optionally substituted with 1 to 5 substituents, or from 1 to 3 substituents, selected from acyloxy, hydroxy, thiol, acyl, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, substituted cycloalkyl, substituted cycloalkenyl, amino, substituted amino, aminoacyl, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halogen, nitro, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, -SO-alkyl, - SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO 2 -alkyl, -SO 2 -substituted alkyl, -SO 2 -aryl and -SO 2 -heteroaryl, and trihalomethyl. [0082] The term “heteroaralkyl” refers to the groups -alkylene-heteroaryl where alkylene and heteroaryl are defined herein. This term includes, by way of example, pyridylmethyl, pyridylethyl, indolylmethyl, and the like. [0083] “Heteroaryloxy” refers to –O-heteroaryl. [0084] “Heterocycle,” “heterocyclic,” “heterocycloalkyl,” and “heterocyclyl” refer to a saturated or unsaturated group having a single ring or multiple condensed rings, including fused bridged and spiro ring systems, and having from 3 to 20 ring atoms, including 1 to 10 hetero atoms. These ring atoms are selected from nitrogen, sulfur, or oxygen, where, in fused ring systems, one or more of the rings can be cycloalkyl, heterocyclyl, aryl, or heteroaryl, provided that the point of attachment is through the non-aromatic ring. Fused ring systems include compounds where two rings share two adjacent atoms. In fused heterocycle systems one or both of the two fused rings can be heterocyclyl. In certain embodiments, the nitrogen and/or sulfur atom(s) of the heterocyclic group are optionally oxidized to provide for the N-oxide, - S(O)-, or –SO 2 - moieties. To satisfy valence requirements, any heteroatoms in such heterocyclic rings may or may not be bonded to one or more H or one or more substituent group(s), e.g., an alkyl group or other substituent as described herein. [0085] Examples of heterocycles and heteroaryls include, but are not limited to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, dihydroindole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, 1,2,3,4-tetrahydroquinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,7-tetrahydrobenzo[b]thiophene, thiazole, thiazolidine, thiophene, benzo[b]thiophene, morpholinyl, 3,4-dihydro-1,4- benzoxazine, thiomorpholinyl (also referred to as thiamorpholinyl), 1,1-dioxothiomorpholinyl, piperidinyl, pyrrolidine, tetrahydrofuranyl, and the like. [0086] Unless otherwise constrained by the definition for the heterocyclic substituent, such heterocyclic groups can be optionally substituted with 1 to 5, or from 1 to 3 substituents, selected from alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO- heteroaryl, -SO 2 -alkyl, -SO 2 -substituted alkyl, -SO 2 -aryl, -SO 2 -heteroaryl, and fused heterocycle. [0087] “Heterocyclyloxy” refers to the group –O-heterocyclyl. [0088] The term “heterocyclylthio” refers to the group heterocyclic-S-. [0089] The term “heterocyclene” refers to the diradical group formed from a heterocycle, as defined herein. [0090] The term “hydroxyamino” refers to the group -NHOH. [0091] “Nitro” refers to the group –NO 2 . [0092] “Oxo” refers to the atom (=O). [0093] “Sulfonyl” refers to the group SO 2 -alkyl, SO 2 -substituted alkyl, SO 2 -alkenyl, SO 2 -substituted alkenyl, SO 2 -cycloalkyl, SO 2 -substituted cylcoalkyl, SO 2 -cycloalkenyl, SO 2 - substituted cylcoalkenyl, SO 2 -aryl, SO 2 -substituted aryl, SO 2 -heteroaryl, SO 2 -substituted heteroaryl, SO 2 -heterocyclic, and SO 2 -substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. Sulfonyl includes, by way of example, methyl-SO 2 -, phenyl-SO 2 -, and 4-methylphenyl-SO 2 -. [0094] “Sulfonyloxy” refers to the group –OSO 2 -alkyl, OSO 2 -substituted alkyl, OSO 2 - alkenyl, OSO 2 -substituted alkenyl, OSO 2 -cycloalkyl, OSO 2 -substituted cylcoalkyl, OSO 2 - cycloalkenyl, OSO 2 -substituted cylcoalkenyl, OSO 2 -aryl, OSO 2 -substituted aryl, OSO 2 - heteroaryl, OSO 2 -substituted heteroaryl, OSO 2 -heterocyclic, and OSO 2 substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0095] The term “aminocarbonyloxy” refers to the group -OC(O)NRR where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein. [0096] “Thiol” refers to the group -SH. [0097] “Thioxo” or the term “thioketo” refers to the atom (=S). [0098] “Alkylthio” or the term “thioalkoxy” refers to the group -S-alkyl, wherein alkyl is as defined herein. In certain embodiments, sulfur may be oxidized to -S(O)-. The sulfoxide may exist as one or more stereoisomers. [0099] The term “substituted thioalkoxy” refers to the group -S-substituted alkyl. [00100] The term “thioaryloxy” refers to the group aryl-S- wherein the aryl group is as defined herein including optionally substituted aryl groups also defined herein. [00101] The term “thioheteroaryloxy” refers to the group heteroaryl-S- wherein the heteroaryl group is as defined herein including optionally substituted aryl groups as also defined herein. [00102] The term “thioheterocyclooxy” refers to the group heterocyclyl-S- wherein the heterocyclyl group is as defined herein including optionally substituted heterocyclyl groups as also defined herein. [00103] In addition to the disclosure herein, the term “substituted,” when used to modify a specified group or radical, can also mean that one or more hydrogen atoms of the specified group or radical are each, independently of one another, replaced with the same or different substituent groups as defined below. [00104] In addition to the groups disclosed with respect to the individual terms herein, substituent groups for substituting for one or more hydrogens (any two hydrogens on a single carbon can be replaced with =O, =NR 70 , =N-OR 70 , =N2 or =S) on saturated carbon atoms in the specified group or radical are, unless otherwise specified, -R 60 , halo, =O, -OR 70 , -SR 70 , -NR 80 R 80 , trihalomethyl, -CN, -OCN, -SCN, -NO, -NO 2 , =N2, -N3, -SO 2 R 70 , -SO 2 O M + , -SO 2 OR 70 , -OSO 2 R 70 , -OSO 2 O M + , -OSO 2 OR 70 , -P(O)(O ) 2 (M + ) 2 , -P(O)(OR 70 )O M + , -P(O)(OR 70 ) 2 , -C(O)R 70 , -C(S)R 70 , -C(NR 70 )R 70 , -C(O)O M + , -C(O)OR 70 , -C(S)OR 70 , -C(O)NR 80 R 80 , -C(NR 70 )NR 80 R 80 , -OC(O)R 70 , -OC(S)R 70 , -OC(O) O-M + , -OC(O)OR 70 , -OC(S)OR 70 , -NR 70 C(O)R 70 , -NR 70 C(S)R 70 , -NR 70 CO 2 M + , -NR 70 CO 2 R 70 , -NR 70 C(S)OR 70 , -NR 70 C(O)NR 80 R 80 , -NR 70 C(NR 70 )R 70 and -NR 70 C(NR 70 )NR 80 R 80 , where R 60 is selected from the group consisting of optionally substituted alkyl, cycloalkyl, heteroalkyl, heterocycloalkylalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each R 70 is independently hydrogen or R 60 ; each R 80 is independently R 70 or alternatively, two R 80’ s, taken together with the nitrogen atom to which they are bonded, form a 5-, 6- or 7-membered heterocycloalkyl which may optionally include from 1 to 4 of the same or different additional heteroatoms selected from the group consisting of O, N and S, of which N may have -H or C 1 -C 3 alkyl substitution; and each M + is a counter ion with a net single positive charge. Each M + may independently be, for example, an alkali ion, such as K + , Na + , Li + ; an ammonium ion, such as + N(R 60 ) 4 ; or an alkaline earth ion, such as [Ca 2+ ] 0.5 , [Mg 2+ ] 0.5 , or [Ba 2+ ] 0.5 (“subscript 0.5 means that one of the counter ions for such divalent alkali earth ions can be an ionized form of a compound of the invention and the other a typical counter ion such as chloride, or two ionized compounds disclosed herein can serve as counter ions for such divalent alkali earth ions, or a doubly ionized compound of the invention can serve as the counter ion for such divalent alkali earth ions). As specific examples, -NR 80 R 80 is meant to include -NH 2 , -NH-alkyl, N-pyrrolidinyl, N-piperazinyl, 4N- methyl-piperazin-1-yl and N-morpholinyl. [00105] In addition to the disclosure herein, substituent groups for hydrogens on unsaturated carbon atoms in “substituted” alkene, alkyne, aryl and heteroaryl groups are, unless otherwise specified, -R 60 , halo, -O-M + , -OR 70 , -SR 70 , -S M + , -NR 80 R 80 , trihalomethyl, -CF 3 , -CN, -OCN, -SCN, -NO, -NO 2 , -N 3 , -SO 2 R 70 , -SO 3 M + , -SO3R 70 , -OSO 2 R 70 , -OSO3 M + , -OSO3R 70 , -PO3 -2 (M + ) 2 , -P(O)(OR 70 )O M + , -P(O)(OR 70 ) 2 , -C(O)R 70 , -C(S)R 70 , -C(NR 70 )R 70 , -CO 2 M + , -CO 2 R 70 , -C(S)OR 70 , -C(O)NR 80 R 80 , -C(NR 70 )NR 80 R 80 , -OC(O)R 70 , -OC(S)R 70 , -OCO 2 M + , -OCO 2 R 70 , -OC(S)OR 70 , -NR 70 C(O)R 70 , -NR 70 C(S)R 70 , -NR 70 CO 2 M + , -NR 70 CO 2 R 70 , -NR 70 C(S)OR 70 , -NR 70 C(O)NR 80 R 80 , -NR 70 C(NR 70 )R 70 and -NR 70 C(NR 70 )NR 80 R 80 , where R 60 , R 70 , R 80 and M + are as previously defined, provided that in case of substituted alkene or alkyne, the substituents are not -O-M + , -OR 70 , -SR 70 , or -S M + . [00106] In addition to the groups disclosed with respect to the individual terms herein, substituent groups for hydrogens on nitrogen atoms in “substituted” heteroalkyl and cycloheteroalkyl groups are, unless otherwise specified, -R 60 , -O-M + , -OR 70 , -SR 70 , -S-M + , -NR 80 R 80 , trihalomethyl, -CF 3 , -CN, -NO, -NO 2 , -S(O) 2 R 70 , -S(O) 2 O-M + , -S(O) 2 OR 70 , -OS(O) 2 R 70 , -OS(O ) 2 O-M + , -OS(O) 2 OR 70 , -P(O)(O-) 2 (M + ) 2 , -P(O)(OR 70 )O-M + , -P(O)(OR 70 )(OR 70 ), -C(O)R 70 , -C(S )R 70 , -C(NR 70 )R 70 , -C(O)OR 70 , -C(S)OR 70 , -C(O)NR 80 R 80 , -C(NR 70 )NR 80 R 80 , -OC(O)R 70 , -OC( S)R 70 , -OC(O)OR 70 , -OC(S)OR 70 , -NR 70 C(O)R 70 , -NR 70 C(S)R 70 , -NR 70 C(O)OR 70 , -NR 70 C(S)O R 70 , -NR 70 C(O)NR 80 R 80 , -NR 70 C(NR 70 )R 70 and -NR 70 C(NR 70 )NR 80 R 80 , where R 60 , R 70 , R 80 and M + are as previously defined. [00107] In addition to the disclosure herein, in a certain embodiment, a group that is substituted has 1, 2, 3, or 4 substituents, 1, 2, or 3 substituents, 1 or 2 substituents, or 1 substituent. [00108] It is understood that in all substituted groups defined above, polymers arrived at by defining substituents with further substituents to themselves (e.g., substituted aryl having a substituted aryl group as a substituent which is itself substituted with a substituted aryl group, which is further substituted by a substituted aryl group, etc.) are not intended for inclusion herein. In such cases, the maximum number of such substitutions is three. For example, serial substitutions of substituted aryl groups specifically contemplated herein are limited to substituted aryl-(substituted aryl)-substituted aryl. [00109] Unless indicated otherwise, the nomenclature of substituents that are not explicitly defined herein are arrived at by naming the terminal portion of the functionality followed by the adjacent functionality toward the point of attachment. For example, the substituent “arylalkyloxycarbonyl” refers to the group (aryl)-(alkyl)-O-C(O)-. [00110] As to any of the groups disclosed herein which contain one or more substituents, it is understood, of course, that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible. In addition, the subject compounds include all stereochemical isomers arising from the substitution of these compounds. [00111] The term “pharmaceutically acceptable salt” means a salt which is acceptable for administration to a patient, such as a mammal (salts with counterions having acceptable mammalian safety for a given dosage regime). Such salts can be derived from pharmaceutically acceptable inorganic or organic bases and from pharmaceutically acceptable inorganic or organic acids. “Pharmaceutically acceptable salt” refers to pharmaceutically acceptable salts of a compound, which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, formate, tartrate, besylate, mesylate, acetate, maleate, oxalate, and the like. [00112] The term “salt thereof” means a compound formed when a proton of an acid is replaced by a cation, such as a metal cation or an organic cation and the like. Where applicable, the salt is a pharmaceutically acceptable salt, although this is not required for salts of intermediate compounds that are not intended for administration to a patient. By way of example, salts of the present compounds include those wherein the compound is protonated by an inorganic or organic acid to form a cation, with the conjugate base of the inorganic or organic acid as the anionic component of the salt. [00113] “Solvate” refers to a complex formed by combination of solvent molecules with molecules or ions of the solute. The solvent can be an organic compound, an inorganic compound, or a mixture of both. Some examples of solvents include, but are not limited to, methanol, N,N-dimethylformamide, tetrahydrofuran, dimethylsulfoxide, and water. When the solvent is water, the solvate formed is a hydrate. [00114] “Stereoisomer” and “stereoisomers” refer to compounds that have same atomic connectivity but different atomic arrangement in space. Stereoisomers include cis-trans isomers, E and Z isomers, enantiomers, and diastereomers. [00115] “Tautomer” refers to alternate forms of a molecule that differ only in electronic bonding of atoms and/or in the position of a proton, such as enol-keto and imine-enamine tautomers, or the tautomeric forms of heteroaryl groups containing a -N=C(H)-NH- ring atom arrangement, such as pyrazoles, imidazoles, benzimidazoles, triazoles, and tetrazoles. A person of ordinary skill in the art would recognize that other tautomeric ring atom arrangements are possible. [00116] It will be appreciated that the term “or a salt or solvate or stereoisomer thereof” is intended to include all permutations of salts, solvates and stereoisomers, such as a solvate of a pharmaceutically acceptable salt of a stereoisomer of subject compound. [00117] “Pharmaceutically effective amount” and “therapeutically effective amount” refer to an amount of a compound sufficient to treat a specified disorder or disease or one or more of its symptoms and/or to prevent the occurrence of the disease or disorder. In reference to tumorigenic proliferative disorders, a pharmaceutically or therapeutically effective amount comprises an amount sufficient to, among other things, cause the tumor to shrink or decrease the growth rate of the tumor. [00118] By “treating” or “treatment” is meant that at least an amelioration of the symptoms associated with the condition afflicting the subject is achieved, where amelioration is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the condition being treated. As such, treatment also includes situations where the pathological condition, or at least symptoms associated therewith, are completely inhibited, e.g., prevented from happening, or stopped, e.g. terminated, such that the subject no longer suffers from the condition, or at least the symptoms that characterize the condition. Thus treatment includes: (i) prevention, that is, reducing the risk of development of clinical symptoms, including causing the clinical symptoms not to develop, e.g., preventing disease progression to a harmful state or prophylactic treatment of a subject; (ii) inhibition, that is, arresting the development or further development of clinical symptoms, e.g., mitigating or completely inhibiting an active disease; and/or (iii) relief, that is, causing the regression of clinical symptoms or alleviating one or more symptoms of the disease or medical condition in the subject. [00119] The terms “polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to a polymeric form of amino acids of any length. Unless specifically indicated otherwise, “polypeptide,” “peptide,” and “protein” can include genetically coded and non- coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones. The term includes fusion proteins, including, but not limited to, fusion proteins with a heterologous amino acid sequence, fusions with heterologous and homologous leader sequences, proteins which contain at least one N-terminal methionine residue (e.g., to facilitate production in a recombinant host cell); immunologically tagged proteins; and the like. [00120] “Native amino acid sequence” or “parent amino acid sequence” are used interchangeably herein to refer to the amino acid sequence of a polypeptide prior to modification to include a modified amino acid residue. [00121] The terms “amino acid analog,” “unnatural amino acid,” and the like may be used interchangeably, and include amino acid-like compounds that are similar in structure and/or overall shape to one or more amino acids commonly found in naturally occurring proteins (e.g., Ala or A, Cys or C, Asp or D, Glu or E, Phe or F, Gly or G, His or H, Ile or I, Lys or K, Leu or L, Met or M, Asn or N, Pro or P, Gln or Q, Arg or R, Ser or S, Thr or T, Val or V, Trp or W, Tyr or Y). Amino acid analogs also include natural amino acids with modified side chains or backbones. Amino acid analogs also include amino acid analogs with the same stereochemistry as in the naturally occurring D-form, as well as the L-form of amino acid analogs. In some instances, the amino acid analogs share backbone structures, and/or the side chain structures of one or more natural amino acids, with difference(s) being one or more modified groups in the molecule. Such modification may include, but is not limited to, substitution of an atom (such as N) for a related atom (such as S), addition of a group (such as methyl, or hydroxyl, etc.) or an atom (such as Cl or Br, etc.), deletion of a group, substitution of a covalent bond (single bond for double bond, etc.), or combinations thereof. For example, amino acid analogs may include α-hydroxy acids, and α-amino acids, and the like. [00122] The terms “amino acid side chain” or “side chain of an amino acid” and the like may be used to refer to the substituent attached to the α-carbon of an amino acid residue, including natural amino acids, unnatural amino acids, and amino acid analogs. An amino acid side chain can also include an amino acid side chain as described in the context of the modified amino acids and/or conjugates described herein. [00123] As used herein the term “isolated” is meant to describe a compound of interest that is in an environment different from that in which the compound naturally occurs. “Isolated” is meant to include compounds that are within samples that are substantially enriched for the compound of interest and/or in which the compound of interest is partially or substantially purified. [00124] As used herein, the term “substantially purified” refers to a compound that is removed from its natural environment and is at least 60% free, at least 75% free, at least 80% free, at least 85% free, at least 90% free, at least 95% free, at least 98% free, or more than 98% free, from other components with which it is naturally associated. [00125] The term “physiological conditions” is meant to encompass those conditions compatible with living cells, e.g., predominantly aqueous conditions of a temperature, pH, salinity, etc. that are compatible with living cells. [00126] As used herein, the term “chronic administration” refers to repeated administration of a compound to a subject. In such treatment, the compound can be administered at least once a week, such as at least once a day, or at least twice or three times a day for a period of at least one month, such as for example five months or more. [00127] As used herein, the term “cysteine protease” refers to a protease having a nucleophilic thiol group in the active site. Cysteine proteases from different organisms can have significantly different cleavage sites. In many RNA class IV viruses, such as coronaviruses, rhinovirus, coxackieviruses and noroviruses, a well-conserved consensus sequence for the 3- chymotrypsin protease (3CP) and 3-chymotrypsin-like protease (3CLP) are observed. For these viruses, this is the main protease (also known as Mpro) responsible for cleaving the polyprotein generated from translation of the viral genome, which liberates the active viral proteins that are critical for viral replication. As this is not a host protease responsible for other critical functions, producing drugs that are highly selective for this viral protease will allow viral replication to be stopped and minimize toxicity for the host. To obtain sufficient inhibition of the protease activity and selectivity over other protease classes, the catalytic mechanism must also be considered in inhibitor design. For cysteine proteases, forming a covalent bond to the catalytic sulfur will ablate activity as it is vital to the cleavage mechanism; however, in some instances, excessive reactivity of the electrophile will also react with serine proteases, other cysteine proteases and other thiols resulting in toxicity. A moiety that forms the covalent bond to the sulfur in the inhibitor is termed the warhead. [00128] Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims. [00129] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention. [00130] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination. All combinations of the embodiments pertaining to the invention are specifically embraced by the present invention and are disclosed herein just as if each and every combination was individually and explicitly disclosed, to the extent that such combinations embrace subject matter that are, for example, compounds that are stable compounds (i.e., compounds that can be made, isolated, characterized, and tested for biological activity). In addition, all sub-combinations of the various embodiments and elements thereof (e.g., elements of the chemical groups listed in the embodiments describing such variables) are also specifically embraced by the present invention and are disclosed herein just as if each and every such sub-combination was individually and explicitly disclosed herein. [00131] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. [00132] It must be noted that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation. [00133] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination. [00134] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed. DETAILED DESCRIPTION [00135] The present disclosure provides compounds with increased metabolic stability for inhibiting a virus infection, such as a Baltimore Group IV RNA virus infection, such as rhinovirus, coxsackievirus, norovirus and coronavirus. Aspects of the present disclosure also include methods of treating the virus infection in a subject with compounds with increased metabolic stability. COMPOUNDS Formula (I) [00136] In certain embodiments, compounds of the present disclosure include a compound of formula (I): wherein: each R 1 is independently selected from –H, –F, –Cl and –CH 3 ; R 2 is selected from –Cl and –F; R 3 is selected from –CH 2 CH 3 , –CH(CH 3 ) 2 , –C(CH 3 ) 3 , –CF(CH 3 ) 2 , –CF 2 CH 3 , –CH(CF 3 )CH 3 , –CH 2 CCl 2 H, –CH 2 CF 3 , –CH(CF 3 ) 2 , cyclopropyl and cyclohexyl; R 4 is selected from –H, –P(=O)(OH) 2 , –C(=O)CH(NH 2 )CH(CH 3 ) 2 and – C(=O)CH 2 NH 2 ; X is selected from –CH 2 –, –CDH– and –CD 2 –; and Y is –CH 2 – or is absent; or a pharmaceutically acceptable salt, solvate, or hydrate thereof. [00137] In certain embodiments, each R 1 is independently selected from –H, –F, –Cl and –CH 3 . In some instances, R 1 is –H. In some instances, R 1 is –F. In some instances, R 1 is –Cl. In some instances, R 1 is –CH 3 . In some instances, each R 1 is independently selected from –H and –F. In some instances, one R 1 is –H and the other R 1 is –F. In some instances, both R 1 are –H. [00138] In certain embodiments, R 2 is selected from –Cl and –F. In some instances, R 2 is –Cl. In some instances, R 2 is –F. [00139] In certain embodiments, R 3 is selected from –CH 2 CH 3 , –CH(CH 3 ) 2 , –C(CH 3 ) 3 , – CF(CH 3 ) 2 , –CF 2 CH 3 , –CH(CF 3 )CH 3 , –CH 2 CCl 2 H, –CH 2 CF 3 , –CH(CF 3 ) 2 , cyclopropyl and cyclohexyl. In certain embodiments, R 3 is selected from –CH(CH 3 ) 2 , –C(CH 3 ) 3 , –CF(CH 3 ) 2 , – CF 2 CH 3 , –CH(CF 3 )CH 3 ,–CH 2 CF 3 , and cyclopropyl. [00140] In some instances, R 3 is –CH 2 CH 3 . In some instances, R 3 is –CH(CH 3 ) 2 . In some instances, R 3 is –C(CH 3 ) 3 . In some instances, R 3 is –CF(CH 3 ) 2 . In some instances, R 3 is – CF 2 CH 3 . In some instances, R 3 is –CH(CF 3 )CH 3 . In some instances, R 3 is –CH 2 CCl 2 H. In some instances, R 3 is –CH 2 CF 3 . In some instances, R 3 is –CH(CF 3 ) 2 . In some instances, R 3 is cyclopropyl. In some instances, R 3 is cyclohexyl. [00141] In certain embodiments, R 4 is selected from –H, –P(=O)(OH) 2 , – C(=O)CH(NH 2 )CH(CH 3 ) 2 and –C(=O)CH 2 NH 2 . In certain embodiments, R 4 is selected from – H, –P(=O)(OH) 2 , and –C(=O)CH(NH 2 )CH(CH 3 ) 2 . [00142] In some instances, R 4 is –H. In some instances, R 4 is –P(O) 3 H2. In some instances, R 4 is –C(=O)CH(NH 2 )CH(CH 3 ) 2 . In some instances, R 4 is –C(=O)CH 2 NH 2 . [00143] In certain embodiments, X is selected from –CH 2 –, –CDH– and –CD 2 –. In certain embodiments, X is selected from –CH 2 – and –CD 2 –. [00144] In some instances, X is –CH 2 –. In some instances, X is –CDH–. In some instances, X is –CD 2 –. [00145] In certain embodiments, Y is –CH 2 – or is absent. In some instances, Y is –CH 2 – . In some instances, Y is absent. [00146] Compounds of the present disclosure (e.g., compounds of formula (I) as described herein) also include an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof. [00147] In addition, compounds of the present disclosure (e.g., compounds of formula (I) as described herein) also include a pharmaceutically acceptable salt, solvate, or hydrate thereof. [00148] In certain embodiments, compounds of the present disclosure (e.g., compounds of formula (I) that find use in the methods of the present disclosure) include compounds selected from: Compound 1 N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxo piperidin-3-yl]butan-2- yl}amino)-1-oxopropan-2-yl]-5,7-difluoro-1H-indole-2-carboxa mide; Compound 2 7-chloro-N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[( 3S)-2-oxopiperidin-3- yl]butan-2-yl}amino)-1-oxopropan-2-yl]-1H-indole-2-carboxami de; Compound 3 N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxo pyrrolidin-3-yl]butan-2- yl}amino)-1-oxopropan-2-yl]-5,7-difluoro-1H-indole-2-carboxa mide; Compound 4 7-chloro-N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[( 3S)-2-oxopyrrolidin-3- yl]butan-2-yl}amino)-1-oxopropan-2-yl]-1H-indole-2-carboxami de; Compound 5 N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxo pyrrolidin-3-yl]butan-2- yl}amino)-1-oxopropan-2-yl]-7-fluoro-1H-indole-2-carboxamide ; Compound 9 N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxo piperidin-3-yl]butan-2- yl}amino)-1-oxopropan-2-yl]-7-fluoro-1H-indole-2-carboxamide ; and

Compound 17 (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L-alanyl]amino}-2-oxo- 4-[(3S)-2-oxopiperidin-3-yl]butyl L-valinate; Compound 19 (3S)-3-{[3-cyclopropyl-N-(7-fluoro-1H-indole-2-carbonyl)-L-a lanyl]amino}-2-oxo-4- [(3S)-2-oxopiperidin-3-yl]butyl L-valinate; and Compound 20 (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L-alanyl]amino}-2-oxo- 4-[(3S)-2-oxopyrrolidin-3-yl]butyl L-valinate. [00149] In certain embodiments, compounds of the present disclosure (e.g., compounds of formula (I) that find use in the methods of the present disclosure) include compounds selected from: Compound 6 5,7-difluoro-N-[(2S)-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxop yrrolidin-3-yl]butan-2- yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2-carboxamide ; Compound 7 7-chloro-N-[(2S)-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxopyrro lidin-3-yl]butan-2- yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2-carboxamide ; Compound 8 7-fluoro-N-[(2S)-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxopyrro lidin-3-yl]butan-2- yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2-carboxamide ; Compound 10 7-fluoro-N-[(2S)-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxopiper idin-3-yl]butan-2- yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2-carboxamide ; Compound 11 5,7-difluoro-N-[(2S)-4-fluoro-1-({(2S)-4-hydroxy-3-oxo-1-[(3 S)-2-oxopiperidin-3- yl]butan-2-yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2- carboxamide; Compound 12 5,7-difluoro-N-[(2S)-4-fluoro-1-({(2S)-4-hydroxy-3-oxo-1-[(3 S)-2-oxopyrrolidin-3- yl]butan-2-yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2- carboxamide; Compound 13 5,7-difluoro-N-[(2S)-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxop iperidin-3-yl]butan-2- yl}amino)-4,4-dimethyl-1-oxopentan-2-yl]-1H-indole-2-carboxa mide; Compound 14 5,7-difluoro-N-[(2S)-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxop yrrolidin-3-yl]butan-2- yl}amino)-4,4-dimethyl-1-oxopentan-2-yl]-1H-indole-2-carboxa mide; Compound 15 7-chloro-N-[(2S)-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxopiper idin-3-yl]butan-2- yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2-carboxamide ; Compound 16 5,7-difluoro-N-[(2S)-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxop iperidin-3-yl]butan-2- yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2-carboxamide ; Compound 18 (3S)-3-{[N-(7-chloro-1H-indole-2-carbonyl)-3-cyclopropyl-L-a lanyl]amino}-2-oxo-4- [(3S)-2-oxopiperidin-3-yl]butyl L-valinate; Compound 21 (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L-alanyl]amino}-2-oxo- 4-[(3S)-2-oxopyrrolidin-3-yl]butyl glycinate; Compound 22 N-[(2S)-4,4-difluoro-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxop yrrolidin-3-yl]butan-2- yl}amino)-1-oxopentan-2-yl]-5,7-difluoro-1H-indole-2-carboxa mide; Compound 23 N-[(2S)-4,4-difluoro-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxop yrrolidin-3-yl]butan-2- yl}amino)-1-oxopentan-2-yl]-7-fluoro-1H-indole-2-carboxamide ; Compound 24 7-chloro-N-[(2S)-4,4-difluoro-1-({(2S)-4-hydroxy-3-oxo-1-[(3 S)-2-oxopyrrolidin-3- yl]butan-2-yl}amino)-1-oxopentan-2-yl]-1H-indole-2-carboxami de;

Compound 25 7-fluoro-N-[(2S)-4-fluoro-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2 -oxopiperidin-3-yl]butan- 2-yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2-carboxami de; Compound 26 N-[(2S)-4,4-difluoro-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxop iperidin-3-yl]butan-2- yl}amino)-1-oxopentan-2-yl]-7-fluoro-1H-indole-2-carboxamide ; and Compound 27 7-fluoro-N-[(2S)-4-fluoro-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2 -oxopyrrolidin-3- yl]butan-2-yl}amino)-4-methyl-1-oxopentan-2-yl]-1H-indole-2- carboxamide. [00150] The compounds described herein can be isolated by procedures known to those skilled in the art. The compounds described herein may be obtained, for instance, by a resolution technique or by chromatography techniques (e.g., silica gel chromatography, chiral chromatography, etc.). As used herein, the term “isolated” refers to compounds that are non- naturally occurring and can be obtained or purified from synthetic reaction mixtures. Isolated compounds may find use in the pharmaceutical compositions and methods of treatment described herein. [00151] The compounds described also include isotopically labeled compounds where one or more atoms have an atomic mass different from the atomic mass conventionally found in nature. Examples of isotopes that may be incorporated into the compounds disclosed herein include, but are not limited to, 2 H (deuterium, D), 3 H (tritium, T), 11 C, 13 C, 14 C, 15 N, 18 O, 17 O, etc. Thus, the disclosed compounds may be enriched in one or more of these isotopes relative to the natural abundance of such isotope. By way of example, deuterium ( 2 H; D) has a natural abundance of about 0.015%. Accordingly, for approximately every 6,500 hydrogen atoms occurring in nature, there is one deuterium atom. Specifically contemplated herein are compounds enriched in deuterium at one or more positions. Thus, deuterium containing compounds of the disclosure have deuterium at one or more positions (as the case may be) in an abundance of greater than 0.015%. In some embodiments, one or more (e.g., 1, 2, 3, 4, 5, 6, 7 or more) hydrogen atoms of a substituent group (e.g., an R-group) of any one of the subject compounds described herein are substituted with a deuterium. [00152] In certain embodiments, the compounds of the present disclosure are in the form of prodrugs that release the active inhibitor in vivo. Stated another way, a compound of the present disclosure, after administration, may undergo a chemical change (e.g., metabolized or converted within the body) into a pharmacologically active agent. For example, as described above, R 4 can be selected from –H, –P(=O)(OH) 2 , –C(=O)CH(NH 2 )CH(CH 3 ) 2 and – C(=O)CH 2 NH 2 . In some instances, when R 4 is –P(=O)(OH) 2 , the compound of formula (I) is a prodrug where the phosphate group (e.g., –P(=O)(OH) 2 group) can be removed by an alkaline phosphatase to produce an active agent where R 4 is H. In some instances, when R 4 is – C(=O)CH(NH 2 )CH(CH 3 ) 2 , the compound of formula (I) is a prodrug where the valinyl ester group (e.g., –C(=O)CH(NH 2 )CH(CH 3 ) 2 group) can be removed by an esterase to produce an active agent where R 4 is H. In some instances, when R 4 is –C(=O)CH 2 NH 2 , the compound of formula (I) is a prodrug where the glycinyl ester group (e.g., –C(=O)CH 2 NH 2 group) can be removed by an esterase to produce an active agent where R 4 is H. METHODS OF USE [00153] The compounds of the present disclosure find use in treatment of a condition or disease in a subject that is amenable to treatment by administration of the compound. Thus, in some embodiments, provided are methods that include administering to a subject a therapeutically effective amount of any of the compounds of the present disclosure. In certain aspects, provided are methods of delivering a compound to a target site in a subject, the method including administering to the subject a pharmaceutical composition including any of the compounds of the present disclosure, where the administering is effective to provide a therapeutically effective amount of the compound at the target site in the subject. [00154] The subject to be treated can be one that is in need of therapy, where the subject to be treated is one amenable to treatment using the compounds disclosed herein. Accordingly, a variety of subjects may be amenable to treatment using the compounds disclosed herein. Generally, such subjects are “mammals”, with humans being of interest. Other subjects can include companion animals or domestic pets (e.g., canine and feline), livestock (e.g., cows, pigs, goats, horses, and the like), rodents (e.g., mice, guinea pigs, and rats, e.g., as in animal models of disease), as well as non-human primates (e.g., chimpanzees, and monkeys). In some instances, the mammal is selected from a companion animal and livestock. In some instances, the mammal is feline. In some instances, the mammal is a human. [00155] The present disclosure provides methods that include delivering a compound of the present disclosure to an individual having a disease, such as methods that include administering to the subject a therapeutically effective amount of a compound of the present disclosure. The methods are useful for treating a wide variety of conditions and/or symptoms associated with a disease. In the context of disease, the term “treating” includes one or more (e.g., each) of: reducing the severity of one or more symptoms, inhibiting the progression, reducing the duration of one or more symptoms, and ameliorating one or more symptoms associated with the disease. [00156] In certain embodiments, methods of the present disclosure include inhibiting a Baltimore Group IV RNA virus in a cell infected with a Baltimore Group IV RNA virus, wherein the method includes contacting the cell with a compound of the present disclosure. In some instances, the contacting includes delivering the compound into the cytosol of the cell by any suitable means. In some instances, the compounds of the present disclosure are effective for inhibiting the viral activity of a Baltimore Group IV RNA virus including any of, e.g., the attachment, penetration, uncoating, replication, assembly, and release of the virus. In some instances, a compound of the present disclosure is effective for treating a Baltimore Group IV RNA virus infection by inhibiting the activity of a protease. The protease may be required for the activity of the virus, e.g., the attachment, penetration, uncoating, replication, assembly, and/or release of the virus. In some instances, compounds of the present disclosure are effective for inhibiting the activity of the protease by inhibiting, e.g., blocking or chemically reacting with, a catalytic domain or catalytic residue(s) of the protease. In some instances, compounds of the present disclosure inhibit the activity of the protease by forming a covalent bond with a catalytic domain or catalytic residue(s). The catalytic domain or catalytic residue(s) may be present in the active site of the protease. In some instances, the protease is a cysteine protease. In some instances, the protease is 3-chymotrypsin protease (3CP). In some instances, the protease is 3-chymotrypsin-like protease (3CLP). [00157] In certain embodiments, methods of the present disclosure include administering a compound of the present disclosure to a subject, where the administering is effective for treating a disease caused by a Baltimore Group IV RNA virus. The methods may include a method of treating a Baltimore Group IV RNA virus infection in a mammal, the method comprising administering to the mammal an effective amount of a compound of the present disclosure. In some embodiments, the methods involve administering an effective amount of a compound according to the present disclosure, a pro-drug thereof or a pharmaceutically acceptable salt thereof to a subject. In certain embodiments, the methods include identifying a subject with a Baltimore Group IV RNA virus infection, e.g., a coronavirus infection, a rhinovirus infection, a coxsackievirus infection, a norovirus infection, and administering a compound of the present disclosure, a pro-drug thereof or a pharmaceutically acceptable salt thereof to the subject. In some instances, the methods include a step (a) of testing a patient for a Baltimore Group IV RNA virus, e.g., before any treatment is administered. The methods may then include step (b) of administering a compound of the present disclosure, a pro-drug thereof or a pharmaceutically acceptable salt thereof to the subject according to any of the embodiments described herein. [00158] A compound of the present disclosure may be administered at any point during a subject’s infection with a Baltimore Group IV RNA virus. In certain embodiments, the subject has, has had, is suspected to have, or is suspected to have had a Baltimore Group IV RNA virus infection. A subject with a Baltimore Group IV RNA virus infection may exhibit one or more symptoms including, e.g., a cough, fever or chills, shortness of breath, fatigue, muscle or body aches, new loss of taste or smell, sore throat, headache, congestion, nasal discharge, nausea, vomiting, diarrhea, stomach pain, chest pain or pressure, confusion, inability to wake or stay awake, and bluish lips or face. In some cases, the subject is asymptomatic. In some cases, a subject with a Baltimore Group IV RNA virus infection exhibits one or more syndromes or acute conditions including, e.g., organ failure, acute respiratory distress syndrome, acute kidney injury, and thrombosis. In certain embodiments, the subject has or is expected to develop symptoms associated with a cytokine response, e.g., a cytokine storm caused by the overproduction of inflammatory cytokines. In some cases, the patient may have signs of respiratory distress, e.g., a cough, but does not have acute respiratory distress syndrome. In these embodiments, the patient may not be in intensive care. In any embodiment, the patient may be 60 years old or more, 70 years old or more, or 80 years old or more. In some instances, the patient may be 60 years old or less, such as 50 years old or less, or 40 years old or less, or 30 years old or less, or 20 years old or less. In some instances, the patient may be immunocompromised, such as immunocompromised due to chemotherapy or radiation therapy. The patient may have or may have had one or more other lung diseases in the past. For example, in some cases, the patient has or has a history of having asthma, pneumothorax, atelectasis, bronchitis, chronic obstructive pulmonary disease, lung cancer or pneumonia. In some cases, the infection is a SARS infection. In some cases, the infection is a MERS infection. In some cases, the infection is a COVID-19 infection. In some cases, the infection is Feline Infectious Peritonitis (FIP). In some instances, the subject receives multiple administrations of a compound over a period including, e.g., days, weeks, or months. [00159] The administering can be done any convenient way. Generally, administration is, for example, oral, buccal, parenteral (e.g., intravenous, intraarterial, subcutaneous), intraperitoneal (i.e., into the body cavity), topically, e.g., by inhalation or aeration (i.e., through the mouth or nose), or rectally systemic (i.e., affecting the entire body). For example, the administration may be systemic, e.g., orally (via injection of tablet, pill or liquid) or intravenously (by injection or via a drip, for example). In other embodiments, the administering can be done by pulmonary administration, e.g., using an inhaler or nebulizer. Compounds of the present disclosure or composition comprising the compounds may be administered in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. The term “topically” may include injection, insertion, implantation, topical application, or parenteral application. [00160] The virus inhibited by the methods may be any of the Baltimore Group IV RNA viruses. In some instances, the Baltimore Group IV RNA virus is selected the family of Picornaviridae, Calciviridae and Coronaviridae. In some instances, the Baltimore Group IV RNA virus is selected from rhinovirus, coxsackievirus, norovirus and coronavirus. In some instances, the Baltimore Group IV RNA virus is selected from norovirus, and coronavirus. In some instances, the Baltimore Group IV RNA virus is human norovirus. In some embodiments, the Baltimore Group IV RNA virus is coronavirus. In certain embodiments, the coronavirus is one that causes disease in mammals. In certain embodiments, the coronavirus causes disease in companion animals or livestock. In certain embodiments, the coronavirus is a feline coronavirus. In certain embodiments, the coronavirus is feline infectious peritonitis. In certain embodiments, the coronavirus is a human coronavirus. In certain embodiments, the coronavirus is selected from Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), Severe Acute Respiratory syndrome coronavirus 1 (SARS-CoV-1) and Middle Eastern Respiratory syndrome-related coronavirus (MERS-CoV). [00161] In some embodiments, by administering a subject the compound of Formula (I), metabolic stability of the compound is increased in the subject by at least 1 fold, 2 fold, 3 fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold, or 10 fold or higher. [00162] In some embodiments, by administering a subject the compound of Formula (I), metabolic stability of the compound in vivo is increased as compared to when the subject is administered a compound other than that of Formula (I), such as a Baltimore Group IV RNA virus inhibitor with a structure other than that of Formula (I). [00163] In some embodiments, the metabolic stability of the compound of Formula (I) in vivo is increased by at least a multiple of 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98 or 100 or more. [00164] In some embodiments, the metabolic stability of the compound of Formula (I), in vivo, is increased by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99%, or a 100% or more compared to the metabolic stability of a compound other than that of Formula (I), such as a Baltimore Group IV RNA virus inhibitor with a structure other than that of Formula (I). [00165] In some embodiments, by administering a subject the compound of Formula (I), the compound exhibits improved efficacy across a range of doses, as compared to a compound other than that of Formula (I), such as a Baltimore Group IV RNA virus inhibitor with a structure other than that of Formula (I). [00166] In some embodiments, due to the improved metabolic stability of the compound of Formula (I), administration of the compound of Formula (I) to a subject can be performed without the co-administration of a metabolism blocker to the subject. For example, in some cases, the compound of Formula (I) can be administered to a subject without co-administration of a metabolism blocker, such as ritonavir. [00167] In some embodiments, due to the improved metabolic stability of the compound of Formula (I), administration of the compound of Formula (I) to a subject can be at a dosage less than the dosage of a compound other than that of Formula (I), such as a Baltimore Group IV RNA virus inhibitor with a structure other than that of Formula (I). In some instances, the same or similar efficacy can be obtained by a compound of Formula (I) at a lower dosage compared to a compound other than that of Formula (I), such as a Baltimore Group IV RNA virus inhibitor with a structure other than that of Formula (I). In some instances, a lower dosage includes a lower concentration of the compound of Formula (I) per unit dose, less frequent dosing, or a fewer number of unit doses per administration, or combinations thereof. Pharmaceutical Compositions [00168] In certain embodiments, the disclosed compounds are useful for the treatment of a disease or disorder. Accordingly, pharmaceutical compositions comprising at least one disclosed compound are also described herein. For example, the present disclosure provides pharmaceutical compositions that include a therapeutically effective amount of a compound of the present disclosure (or a pharmaceutically acceptable salt or solvate or hydrate or stereoisomer thereof) and a pharmaceutically acceptable excipient. [00169] A pharmaceutical composition that includes a subject compound may be administered to a patient alone, or in combination with other supplementary active agents. For example, one or more compounds according to the present disclosure can be administered to a patient with or without supplementary active agents. The pharmaceutical compositions may be manufactured using any of a variety of processes, including, but not limited to, conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, lyophilizing, and the like. The pharmaceutical composition can take any of a variety of forms including, but not limited to, a sterile solution, suspension, emulsion, spray dried dispersion, lyophilisate, tablet, microtablets, pill, pellet, capsule, powder, syrup, elixir or any other dosage form suitable for administration. [00170] A compound of the present disclosure may be administered to a subject using any convenient means capable of resulting in the desired reduction in disease condition or symptom. Thus, a compound can be incorporated into a variety of formulations for therapeutic administration. More particularly, a compound can be formulated into pharmaceutical compositions by combination with appropriate pharmaceutically acceptable excipients, carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, aerosols, and the like. [00171] Formulations for pharmaceutical compositions are described in, for example, Remington’s Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, Pa., 19th Edition, 1995, which describes examples of formulations (and components thereof) suitable for pharmaceutical delivery of the disclosed compounds. Pharmaceutical compositions that include at least one of the compounds can be formulated for use in human or veterinary medicine. Particular formulations of a disclosed pharmaceutical composition may depend, for example, on the mode of administration and/or on the location of the subject to be treated. In some embodiments, formulations include a pharmaceutically acceptable excipient in addition to at least one active ingredient, such as a compound of the present disclosure. In other embodiments, other medicinal or pharmaceutical agents, for example, with similar, related or complementary effects on the disease or condition being treated can also be included as active ingredients in a pharmaceutical composition. [00172] Pharmaceutically acceptable carriers useful for the disclosed methods and compositions may depend on the particular mode of administration being employed. In addition to biologically neutral carriers, pharmaceutical compositions to be administered can optionally contain non-toxic auxiliary substances (e.g., excipients), such as wetting or emulsifying agents, preservatives, and pH buffering agents, and the like. The disclosed pharmaceutical compositions may be formulated as a pharmaceutically acceptable salt of a disclosed compound. [00173] The term “unit dosage form,” as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of a compound calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, excipient, carrier or vehicle. The specifications for a compound depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the subject. [00174] The dosage form of a disclosed pharmaceutical composition may be determined by the mode of administration chosen. For example, in addition to injectable fluids, topical or oral dosage forms may be employed. Topical preparations may include eye drops, ointments, sprays and the like. Oral formulations may be liquid (e.g., syrups, solutions or suspensions), or solid (e.g., powders, pills, tablets, or capsules). Methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. [00175] Certain embodiments of the pharmaceutical compositions that include a subject compound may be formulated in unit dosage form suitable for individual administration of precise dosages. The amount of active ingredient administered may depend on the subject being treated, the severity of the affliction, and the manner of administration, and is known to those skilled in the art. In certain instances, the formulation to be administered contains a quantity of the compounds disclosed herein in an amount effective to achieve the desired effect in the subject being treated. [00176] Each therapeutic compound can independently be in any dosage form, such as those described herein, and can also be administered in various ways, as described herein. For example, the compounds may be formulated together, in a single dosage unit (that is, combined together in one form such as capsule, tablet, powder, or liquid, etc.) as a combination product. Alternatively, when not formulated together in a single dosage unit, an individual compound may be administered at the same time as another therapeutic compound or sequentially, in any order thereof. [00177] A disclosed compound can be administered alone, as the sole active pharmaceutical agent, or in combination with one or more additional compounds of the present disclosure or in conjunction with other agents. When administered as a combination, the therapeutic agents can be formulated as separate compositions that are administered simultaneously or at different times, or the therapeutic agents can be administered together as a single composition combining two or more therapeutic agents. Thus, the pharmaceutical compositions disclosed herein containing a compound of the present disclosure optionally include other therapeutic agents. Accordingly, certain embodiments are directed to such pharmaceutical compositions, where the composition further includes a therapeutically effective amount of an agent selected as is known to those of skill in the art. Methods of Administration [00178] The subject compounds find use for treating a disease or disorder in a subject. The route of administration may be selected according to a variety of factors including, but not limited to, the condition to be treated, the formulation and/or device used, the subject to be treated, and the like. Routes of administration useful in the disclosed methods include, but are not limited to, oral and parenteral routes, such as intravenous (iv), intraperitoneal (ip), rectal, topical, ophthalmic, nasal, intrathecal, and transdermal. Formulations for these dosage forms are described herein. [00179] An effective amount of a subject compound may depend, at least, on the particular method of use, the subject being treated, the severity of the affliction, and the manner of administration of the therapeutic composition. A “therapeutically effective amount” of a composition is a quantity of a specified compound sufficient to achieve a desired effect in a subject (e.g., patient) being treated. For example, this may be the amount of a subject compound necessary to prevent, inhibit, reduce or relieve a disease or disorder in a subject. Ideally, a therapeutically effective amount of a compound is an amount sufficient to prevent, inhibit, reduce or relieve a disease or disorder in a subject without causing a substantial cytotoxic effect on host cells in the subject. [00180] Therapeutically effective doses of a subject compound or pharmaceutical composition can be determined by one of skill in the art. For example, in some instances, a therapeutically effective dose of a compound or pharmaceutical composition is administered with a goal of achieving local (e.g., tissue) concentrations that are at least as high as the EC 50 of an applicable compound disclosed herein. [00181] The specific dose level and frequency of dosage for any particular subject may be varied and may depend upon a variety of factors, including the activity of the subject compound, the metabolic stability and length of action of that compound, the age, body weight, general health, sex and diet of the subject, mode and time of administration, rate of excretion, drug combination, and severity of the condition of the host undergoing therapy. [00182] In some embodiments, multiple doses of a compound are administered. The frequency of administration of a compound can vary depending on any of a variety of factors, e.g., severity of the symptoms, condition of the subject, etc. For example, in some embodiments, a compound is administered once per month, twice per month, three times per month, every other week, once per week (qwk), twice per week, three times per week, four times per week, five times per week, six times per week, every other day, daily (qd/od), twice a day (bds/bid), or three times a day (tds/tid), etc. EXAMPLES [00183] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g. amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Celsius, and pressure is at or near atmospheric. By “average” is meant the arithmetic mean. Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pl, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m., intramuscular(ly); i.p., intraperitoneal(ly); s.c., subcutaneous(ly); and the like. General Synthetic Procedures [00184] Many general references providing commonly known chemical synthetic schemes and conditions useful for synthesizing the disclosed compounds are available (see, e.g., Smith and March, March’s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Fifth Edition, Wiley-Interscience, 2001; or Vogel, A Textbook of Practical Organic Chemistry, Including Qualitative Organic Analysis, Fourth Edition, New York: Longman, 1978). [00185] Compounds as described herein can be purified by any purification protocol known in the art, including chromatography, such as HPLC, preparative thin layer chromatography, flash column chromatography and ion exchange chromatography. Any suitable stationary phase can be used, including normal and reversed phases as well as ionic resins. In certain embodiments, the disclosed compounds are purified via silica gel and/or alumina chromatography. See, e.g., Introduction to Modern Liquid Chromatography, 2nd Edition, ed. L. R. Snyder and J. J. Kirkland, John Wiley and Sons, 1979; and Thin Layer Chromatography, ed E. Stahl, Springer-Verlag, New York, 1969. [00186] During any of the processes for preparation of the subject compounds, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups as described in standard works, such as J. F. W. McOmie, “Protective Groups in Organic Chemistry”, Plenum Press, London and New York 1973, in T. W. Greene and P. G. M. Wuts, “Protective Groups in Organic Synthesis”, Third edition, Wiley, New York 1999, in “The Peptides”; Volume 3 (editors: E. Gross and J. Meienhofer), Academic Press, London and New York 1981, in “Methoden der organischen Chemie”, Houben-Weyl, 4 th edition, Vol. 15/l, Georg Thieme Verlag, Stuttgart 1974, in H.-D. Jakubke and H. Jescheit, “Aminosauren, Peptide, Proteine”, Verlag Chemie, Weinheim, Deerfield Beach, and Basel 1982, and/or in Jochen Lehmann, “Chemie der Kohlenhydrate: Monosaccharide and Derivate”, Georg Thieme Verlag, Stuttgart 1974. The protecting groups may be removed at a convenient subsequent stage using methods known from the art. [00187] The subject compounds, including compounds that are not commercially available, can be synthesized via a variety of different synthetic routes using commercially available starting materials and/or starting materials prepared by conventional synthetic methods. A variety of examples of synthetic routes that can be used to synthesize the compounds disclosed herein are described in the scheme below. [00188] In certain embodiments, compounds of the present disclosure (e.g., compounds of formula (I) are synthesized using conventional methods and conditions, as depicted in the combination of Scheme 1. Scheme 1 wherein R 1 , R 2 , R 3 , R 4 , X, Y and Z are as defined herein. [00189] The starting materials and reagents employed in Scheme 1 may be obtained commercially or through conventional techniques. For example, the lactam starting material can be generated as in Yangyang Zhai et al Journal of Medicinal Chemistry (2015), 58, 9414 – 9420 or Robert L. Hoffman et al Journal of Medicinal Chemistry (2020), 63, 12725 – 12747. The scheme is an example of a method to generate the compounds of the present disclosure where the exact steps and materials will depend on the functional groups present. The selection of the starting materials, reagent, substrates, base, protecting group, solvent and leaving group can be accomplished by one of ordinary skilled in the art. A nitrogen protected, a non-limiting example is a tert-butylcarbonate (Boc) group, an ester of a lactam containing amino acid, a non-limiting example is the methyl ester, is treated with sodium chloroacetate in the present of a base, such as trimethylamine, in an organic solvent, such as THF. To the resulting cooled mixture is added tert-butylmagnesium chloride, or another suitable reagent generating a chloromethylketone. The protecting group on the nitrogen is then removed, such as with HCl or trifluoroacetic acid (TFA) for a Boc protecting group, and the resulting unprotected nitrogen is coupled with a N-protected amino acid, such as with a Boc group, by treating both with a coupling agent, for example 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5- b]pyridinium 3-oxide hexafluorophosphate (HATU), and excess base, such as N- methylmorpholine (NMM), in a suitable solvent, such as N,N-dimethylformamide (DMF) or methylene chloride (DCM). The N-protecting group of the coupled product is removed, such as with HCl or TFA in a suitable solvent like dioxane or dichloromethane for a Boc group. The resulting unprotected nitrogen is coupled with the corresponding indole carboxylic acid using a coupling agent, such as HATU, in the presence of a base, such as NMM, in a suitable solvent, such as DMF. Displacement of the chloride of the chloromethylketone can be achieved by treatment of commercially available ZCO 2 H that was pretreated with a base, such as sodium tert-butoxide, possibly in the presence of sodium iodide, in a suitable solvent, such as DMF. The compound ZCO 2 H that is selected will determine which R 4 group is ultimately generated. For, OR 4 corresponding to L-valinal ester or glycinal ester ZCO 2 H of Boc-L-valine or Bocglycine can be utilized. After chloride displacement, removal of the protecting group, such as Boc with HCl or TFA in a suitable solvent such as dioxane or DCM, will generate the target molecule of formula (I). For R 4 = H or –P(=O)(OH) 2 , or a salt there of, ZCO 2 H such as oxo(phenyl)acetic acid is selected after which transesterification can be performed, such as with cesium fluoride in a tetrahyrdrofuran (THF) and methanol mixture, generating formula (I) compound with R 4 = H. Subsequent conversion of R 4 = H to R 4 = P(=O)(OH) 2 or a suitable salt of Formula (I) can be achieved by a two step process of treatment with di-tert-butyl N,N- dipropan-2-ylphosphoramidite with tetrazole in THF followed by first oxidation with hydrogen peroxide and then treatment with TFA, such as was performed on a different compound in Britton Boras et al., Nature Communications (2021), 12, 6055. [00190] Scheme 1 is meant to be by way of a non-limiting example only, and one of ordinary skill in the art will understand that alternate reagents, solvents, order of reactions or starting materials can be used to make compounds of the present disclosure and/or other intermediates or compounds contained herein. EXAMPLE 1: SYNTHESIS OF COMPOUNDS [00191] All reagents and solvents were used as purchased from commercial sources. Moisture sensitive reactions were carried out under a nitrogen atmosphere. Reactions were monitored by TLC using pre-coated silica gel aluminum plates containing a fluorescent indicator (F-254). Detection was done with UV (254 nm). Alternatively, the progress of a reaction was monitored by LC/MS. Specifically, but without limitation, the following abbreviations were used, in addition to the other ones described herein, in the examples: Boc (tert-butoxycarbonyl); Boc2O (di-tert-butyl dicarbonate); cat. (catalytic amount); DCM (dichloromethane); dioxane (1,4-dioxane); DMF (N,N-dimethylformamide); CDI (1,1′- carbonyldiimidazole); EDCI (N-ethyl-N′-carbodiimide); EtOH (ethanol); ether or Et 2 O (diethyl ether); Et3N (triethylamine); EtOAc (ethyl acetate); HATU (1-[bis(dimethylamino)methylene]- 1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate or N-[(dimethylamino)-1H- 1,2,3-triazolo-[4,5-b]pyridin-1-ylmethylene]-N-methylmethana minium hexafluorophosphate N- oxide); KOtBu (potassium tert-butoxide); hex (hexanes); MeCN (acetonitrile); MeOH (methanol); µW (microwave); N-methylmorpholine (NMM); NaOtBu (sodium tert-butoxide); O/N (overnight); RT or rt (room or ambient temperature); TBS (tert-butyldimethylsilyl); t- BuMgCl (tert-butylmagnesium chloride); TFA (trifluoroacetic acid); TFAA (trifluoroacetic anhydride); THF (tetrahydrofuran). 1 H NMR spectra were recorded at RT with a Bruker Avance III 600 MHz NMR spectrometer equipped with a Bruker’s 5 mm PABBO probe. Chemical shifts are reported in ppm downfield from tetramethylsilane using residual solvent signals as internal reference. NMR data were processed utilizing ACD/Spectrus processor (v2016.1.1, ACD/Labs Inc.). Nomenclature for the naming of compounds, such as for Compound Examples and intermediate compounds, were performed using ACD/Name (Chemists’ Version from ACD/Labs Inc.) or Bruker TopSpin 4.1.3 to generate the IUPAC-style names. Naming of commercial or literature compounds utilized SciFinder, ACD/Names, and common or trivial names known to those skilled in the art. [00192] The LC/MS system used for monitoring the progress of reactions, assessing the purity (absorbance at 254 nm) and identity of the product consisted of Dionex ULTIMATE 3000 uHPLC module and Thermo Scientific LTQ XL mass-spectrometer with electrospray ionization and Ion-Trap type of detector (alternating positive-negative mode). Separation was performed with Thermo Scientific™ Accucore™ aQ C18 Polar Endcapped LC column (100 mm × 2.1 mm; particle size 2.6 μm, 80 Å). The column was maintained at 35 °C. Commercial HPLC-grade methanol and domestic ‘millipore (Milli-Q)’ water used for chromatography were modified by adding 0.1% (v/v) of formic acid. The eluent was delivered with constant flow rate of 0.4 mL/min, column was equilibrated for 5 min with the corresponding eluent prior to injection of the sample (1 μL) and one of the following separation conditions were used: Eluent systems: A - Gradient of Methanol-Water, 45 to 95% in 5.25 min, followed by 5 min of isocratic MeOH – water 95%; and B – Gradient of Methanol-Water, 30 to 65% in 4.75 min, then to 95% in 2.5 min, followed by 4 min of isocratic MeOH – water 95%. Compound 1 Synthesis of N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxo piperidin-3- yl]butan-2-yl}amino)-1-oxopropan-2-yl]-5,7-difluoro-1H-indol e-2-carboxamide, 1

[00193] Compound 1 was synthesized as in Scheme 2. [00194] Preparation of tert-butyl {(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperidin-3-yl]butan-2- yl}carbamate, (3). A mixture of methyl N-(tert-butoxycarbonyl)-3-[(3S)-2-oxopiperidin-3-yl]- L-alaninate (1) (2.5 g, 8.32 mmol) [prepared using method in Journal of Medicinal Chemistry (2015), 58, p 9414-9420 and references within], sodium chloroacetate (2) (2.9 g, 25.0 mmol) and triethylamine (4.1 g, 25.0 mmol) in THF (200 mL) was cooled in an ice bath. To this mixture was added t-BuMgCl (41.6 ml, 83.2 mmol; 2M in ether) dropwise over 1 h via cannula. After addition, the ice bath was removed and the resulting cloudy solution was warmed to room temperature. After overnight, the resulting light brown mixture was cooled again in an ice bath and quenched slowly with 4N HCl until a clear solution was obtained. The resulting mixture was extracted with ethyl acetate (3 ^ 100 mL). The combined organic layer was washed with water (3 ^ 25 mL), brine (25 mL), and then dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The product was purified by silica column chromatography (gradient of 50% to 100% ethyl acetate in hexanes), which produced (3) (2.3 g, 86% yield) as a light brown oil. Preparation of (3S)-3-[(2S)-2-amino-4-chloro-3-oxobutyl]piperidin-2-one hydrochloride salt, (4). A solution of tert-butyl {(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperidin-3-yl]butan-2- yl}carbamate (3) (0.6 g, 1.9 mmol) in CH 2 Cl 2 (20 mL) was cooled in an ice bath and HCl (5 mL, 20 mmol; 4M in 1,4-dioxane) was added. After 30 min, the ice bath was removed and the mixture allowed to warmed to room temperature. After overnight, the mixture was concentrated under reduced pressure (30 ºC water bath temperature), which afforded (4) (2.96 g) as an off- white foam. This material was used without further purification in the next step. [00195] Preparation tert-butyl [(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperidin-3- yl]butan-2-yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]carbamat e, (6). To a mixture of N-(tert- butoxycarbonyl)-3-cyclopropyl-L-alanine (5) (473 mg, 2.06 mmol) and HATU (783 mg, 2.06 mmol) in anhydrous DMF (5 mL) was cooled in an ice bath. After stirring for 15 min, NMM (625 mg, 6.21 mmol) was added followed by (4) (478 mg, 1.88 mmol, in 2 mL of DMF) and stirred for additional 45 min. It was then poured into crushed ice, liquid was decanted. The product was purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which generated (6) (730 mg, 90% yield) as an off-white foam. [00196] Preparation of N-{(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperidin-3-yl]butan-2-y l}-3- cyclopropyl-L-alaninamide, (7). A solution of (6) (730 mg, 1.70 mmol) in CH 2 Cl 2 (25 mL) was cooled in an ice-water bath and then HCl (5 mL, 20 mmol; 4 M in 1,4-dioxane) was added. After 30 min, the ice bath was removed. After overnight, a gummy precipitate formed. The mixture was then concentrated under reduced pressure (bath temperature at 35 ºC), which afforded (7) (598 mg, 96%) as an off-white solid. This material was used without further purification in the next step. [00197] Preparation of N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperidin-3-yl] butan- 2-yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]-5,7-difluoro-1H- indole-2-carboxamide, (9). To a mixture of 5,7-difluoro-1H-indole-2-carboxylic acid (8) (119 mg, 0.60 mmol) and HATU (228 mg, 0.60 mmol) in anhydrous DMF (5 mL) was cooled in an ice bath. After stirring for 15 min, NMM (166 mg, 1.64 mmol) was added followed by (7) (200 mg, 0.55 mmol, in 2 mL of DMF) and stirred for additional 45 min. It was then poured into crushed ice forming a solid that was filtered and air dried. The solid was dissolved in CHCl 3 (5 mL) and product was purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which generated (9) (224 mg, 81% yield) as a white foam. [00198] Preparation of (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl oxo(phenyl)acetate, (11). A mixture of oxo(phenyl)acetic acid (10) (80 mg, 0.53 mmol) and sodium tert-butoxide (51 mg, 0.53 mmol) in anhydrous DMF (3 mL) was stirred at room temperature for 30 min. To this mixture, a solution of (9) (224 mg, 0.44 mmol, in 2 mL of DMF) and NaI (7.5 mg, 0.05 mmol) were added. After 24 h, it was poured into crushed ice to form a solid and filtered. The solid was dissolved in CHCl 3 (5 mL) and purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which afforded (11) (138 mg, 50% yield) as an off-white solid. [00199] Preparation of N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2- oxopiperidin-3-yl]butan-2-yl}amino)-1-oxopropan-2-yl]-5,7-di fluoro-1H-indole-2- carboxamide, 1. To a solution of (11) (100 mg, 0.16 mmol) in THF/methanol (10 mL/10 mL) at room temperature under nitrogen atmosphere was added CsF (5 mg, 0.03 mmol). After overnight, the resulting mixture was concentrated under reduced pressure and the product purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which afforded (41 mg, 52% yield). 1 H NMR (600 MHz, DMSO-d 6 ) δ 12.24 (s, 1H), 8.60 (d, J = 7.5 Hz, 1H), 8.55 (d, J = 7.9 Hz, 1H), 7.46 (br s, 1H), 7.36 – 7.32 (m, 2H), 7.14 – 7.10 (m, 1H), 5.08 (t, J = 6.0 Hz, 1H), 4.60 – 4.57 (m, 1H), 4.56 – 4.52 (m, 1H), 4.28 (dd, J = 6.0, 18.8 Hz, 1H), 4.18 (dd, J = 5.6, 18.8 Hz, 1H), 3.19 – 3.06 (m, 2H), 2.25 – 2.20 (m, 1H), 2.17 – 2.12 (m, 1H), 1.91 – 1.87 (m, 1H), 1.82 – 1.77 (m, 1H), 1.78 – 1.65 (m, 2H), 1.61 – 1.51 (m, 2H), 1.41 – 1.34 (m, 1H), 0.90 – 0.82 (m, 1H), 0.49 – 0.39 (m, 2H), 0.27 – 0.22 (m, 1H), 0.19 – 0.15 (m, 1H). 19 F NMR (565 MHz, DMSO-d 6 ) δ -121.40 (t, J = 9.61, 1F), -127.56 (d, J = 11.0, 1F). LC/MS: Eluent system A (retention time: 4.66 min); ESI-MS: 491 [M + H] + . Compound 2 Synthesis of 7-chloro-N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[( 3S)-2- oxopiperidin-3-yl]butan-2-yl}amino)-1-oxopropan-2-yl]-1H-ind ole-2-carboxamide, 2 [00200] Compound 2 was synthesized as in Scheme 3. Scheme 3 [00201] Preparation of 7-chloro-N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperi din-3- yl]butan-2-yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]-1H-indo le-2-carboxamide, (13). To a mixture of 7-chloro-1H-indole-2-carboxylic acid (12) (180 mg, 0.91 mmol) and HATU (346 mg, 0.91 mmol) in anhydrous DMF (mL) was cooled in an ice bath. After stirring for 15 min, NMM (250 mg, 2.46 mmol) was added followed by (7) (300 mg, 0.82 mmol, in 2 mL of DMF). After 45 min, the mixture was poured into crushed ice, filtered and air dried. The product was purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which generated (13) (387 mg, 93% yield) as a white foam. [00202] Preparation of (3S)-3-{[N-(7-chloro-1H-indole-2-carbonyl)-3-cyclopropyl-L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl oxo(phenyl)acetate, (14). A mixture of oxo(phenyl)acetic acid (10) (140 mg, 0.93 mmol) and sodium tert-butoxide (90 mg, 0.93 mmol) in anhydrous DMF (3 mL) was stirred at room temperature for 30 min. To this mixture was added a solution of (13) (387 mg, 0.77 mmol, in 2 mL of DMF) and NaI (12 mg, 0.08 mmol). After 24 h, the resulting mixture was poured into crushed ice and filtered. The residue was dissolved in CHCl 3 (5 mL) and the product purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which afforded (14) (360 mg, 75% yield) as an off- white solid. [00203] Preparation of 7-chloro-N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[( 3S)-2- oxopiperidin-3-yl]butan-2-yl}amino)-1-oxopropan-2-yl]-1H-ind ole-2-carboxamide, 2. To a solution of (14) (360 mg, 0.58 mmol) in THF/MeOH (20 mL/20 mL) at room temperature under nitrogen atmosphere was added CsF (9 mg, 0.06 mmol). After overnight, the resulting mixture was concentrated under reduced pressure and the product purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which afforded (116 mg, 41% yield). 1 H NMR (600 MHz, DMSO-d 6 ) δ 11.76 (br s, 1H), 8.68 (d, J = 7.9 Hz, 1H), 8.60 (d, J = 8.3 Hz, 1H), 7.67 (d, J = 7.9 Hz, 1H), 7.46 (br s, 1H), 7.35 (d, J = 7.9 Hz, 1H), 7.30 (d, J = 2.3 Hz, 1H), 7.11 (t, J = 7.7 Hz, 1H), 5.08 (t, J = 6.0 Hz, 1H), 4.64 – 4.60 (m, 1H), 4.56 – 4.52 (m, 1H), 4.28 (dd, J = 6.0, 18.8 Hz, 1H), 4.19 (dd, J = 5.6, 18.8 Hz, 1H), 3.19 – 3.04 (m, 2H), 2.27 – 2.19 (m, 1H), 2.25 – 2.20 (m, 1H), 2.18 – 2.12 (m, 1H), 1.91 – 1.84 (m, 1H), 1.81 – 1.75 (m, 1H), 1.75 – 1.68 (m, 1H), 1.63 – 1.58 (m, 1H), 1.57 – 1.51 (m, 1H), 1.41 – 1.34 (m, 1H), 0.94 – 0.85 (m, 1H), 0.50 – 0.40 (m, 2H), 0.27 – 0.22 (m, 1H), 0.20 – 0.16 (m, 1H). LC/MS: Eluent system A (retention time: 4.97 min); ESI-MS: 489 [M + H] + . Compound 3 Synthesis of N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxo pyrrolidin-3- yl]butan-2-yl}amino)-1-oxopropan-2-yl]-5,7-difluoro-1H-indol e-2-carboxamide, 3 [00204] Compound 3 was synthesized as in Scheme 4. Scheme 4 [00205] Preparation of (3S)-3-[(2S)-2-amino-4-chloro-3-oxobutyl]pyrrolidin-2-one hydrochloride salt, (16). A solution of tert-butyl {(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopyrrolidin- 3-yl]butan-2-yl}carbamate (15) (950 mg, 3.12 mmol) [prepared according to Bing Bai et al. Journal of Medicinal Chemistry 2022, 65, 2905 - 2925] in CH 2 Cl 2 (40 mL) was cooled using an ice-water bath and then 4 M HCl in 1,4-dioxane (10 mL) was added. After 30 min, the ice bath was removed and the mixture was warmed to room temperature. After overnight, the mixture was concentrated under reduced pressure (bath temperature at 30 ºC). Co-evaporated with DCM (3 x 10 mL), Et2O (1 x 10 mL), and drying under reduced pressure for 1 h afforded (16) an off-white solid (750 mg). This material was used without further purification in the next step. [00206] Preparation of tert-butyl [(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopyrrolidin-3- yl]butan-2-yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]carbamat e, (17). To a solution of (16) (750 mg, 3.12 mmol) and N-(tert-butoxycarbonyl)-3-cyclopropyl-L-alanine (5) (790 mg, 3.43 mmol) in anhydrous DMF (25 mL) cooled in an ice bath was added HATU (1.30 g, 3.43 mmol). Then NMM (1.03 mL, 9.35 mmol) was added dropwise. After 45 min, ice-water mixture (50 mL) was added and the resulting mixture was extracted with ethyl acetate (3 x 50 mL). The combined organic layer was washed with saturated brine solution (1 x 50 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting residue was dissolved in CHCl 3 (25 mL) and loaded on 80 g silica gel column (Silicycle) and product purified by Biotage ® with a gradient of 0 to 4% MeOH in CHCl 3 , which generated (17) (900 mg, 70% yield) as a white foamy solid. [00207] Preparation of N-{(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopyrrolidin-3-yl]butan-2- yl}-3- cyclopropyl-L-alaninamide hydrochloride salt, (18). A solution of (17) (900 mg, 2.16 mmol) in CH 2 Cl 2 (40 mL) was cooled using an ice-water bath and then 4 M HCl in 1,4-dioxane (10 mL) was added. After 30 min, the ice bath was removed and the mixture was allowed to warm to room temperature. After overnight, the reaction mixture was then concentrated under reduced pressure (bath temperature at 30 ºC). Co-evaporated with DCM (2 x 10 mL), Et2O (1 x 10 mL), and drying under reduced pressure for 1 h afforded (18) as an off-white solid (760 mg). This material was used without further purification in the next step. [00208] Preparation of N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopyrrolidin-3-yl ]butan- 2-yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]-5,7-difluoro-1H- indole-2-carboxamide, (19). To a solution of (18) (156 mg, 0.442 mmol) and 5,7-difluoro-1H-indole-2-carboxylic acid (8) (95.8 mg, 0.486 mmol) in anhydrous DMF (10 mL) cooled in an ice bath was added HATU (185 mg, 0.486 mmol). Then NMM (146 µL, 1.33 mmol) was added dropwise. After 45 min, ice-water mixture (50 mL) was added and the resulting mixture was extracted with ethyl acetate (3 x 50 mL). The combined organic layer was washed with saturated brine solution (1 x 50 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was dissolved in CHCl 3 (10 mL) and loaded on 2 x 12 g silica gel columns (Silicycle) and product purified by Biotage ® with a gradient of 0 to 2% MeOH in CHCl 3 , which generated (19) (140.3 mg, 64% yield) as an off-white solid. [00209] Preparation of (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopyrrolidin-3-yl]butyl oxo(phenyl)acetate, (20). A mixture of oxo(phenyl)acetic acid (10) (255 mg, 1.70 mmol) and sodium tert-butoxide (81.7 mg, 0.850 mmol) in anhydrous DMF (5 mL) was stirred at room temperature for 30 min. To this mixture, a solution of (19) (140 mg, 0.284 mmol, in 5 mL of DMF), followed by NaI (85.0 mg, 0.567 mmol) were added. After overnight at room temperature, the mixture was poured into crushed ice. The resulting precipitation was filtered, washed with water (3 x 2 mL), dried under vacuum. The residue was dissolved in CHCl 3 (20 mL) and loaded on 2 x 12 g silica gel columns (Silicycle) and product purified by Biotage® with a gradient of 0 to 1% MeOH in CHCl 3 , which afforded (20) (112.4 mg, 65% yield) as an off-white solid. [00210] Preparation of N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2- oxopyrrolidin-3-yl]butan-2-yl}amino)-1-oxopropan-2-yl]-5,7-d ifluoro-1H-indole-2- carboxamide, 3. To a solution of (20) (112 mg, 0.185 mmol) in THF (10 mL) at room temperature under nitrogen atmosphere was added a solution of CsF (2.8 mg, 0.018 mmol) in MeOH (10 mL). After overnight at RT, the mixture was concentrated under reduced pressure (bath temperature kept at 30 ºC) then co-evaporated with DCM (1 x 10 mL). The residue was dissolved in CHCl 3 (10 mL) with 5 drops of MeOH and loaded on 2 x 12 g silica gel columns (Silicycle) and product purified by Biotage ® with a gradient of 0 to 2% MeOH in CHCl 3 , the resulting solid was triturated with Et 2 O (5 mL) and hexanes, filtered, and the solid washed with hexanes (3 x 2 mL), dried under vacuum, which generated 3 (63.2 mg, 72% yield) as an off- white solid. 1 H NMR (600 MHz, DMSO-d 6 ) δ 12.25 (d, J = 1.1 Hz, 1H), 8.61 (d, J = 7.7 Hz, 1H), 8.54 (d, J = 7.9 Hz, 1H), 7.66 (s, 1H), 7.36 (d, J = 2.0 Hz, 1H), 7.35 – 7.32 (m, 1H), 7.15 – 7.10 (m, 1H), 5.11 (t, J = 6.0 Hz, 1H), 4.62 – 4.56 (m, 1H), 4.54 – 4.48 (m, 1H), 4.28 (dd, J = 6.1, 18.8 Hz, 1H), 4.18 (dd, J = 5.8, 18.8 Hz, 1H), 3.20 – 3.09 (m, 2H), 2.37 – 2.30 (m, 1H), 2.17 – 2.10 (m, 1H), 1.99 – 1.92 (m, 1H), 1.84 – 1.78 (m, 1H), 1.71 – 1.63 (m, 2H), 1.62 – 1.55 (m, 1H), 0.92 – 0.83 (m, 1H), 0.51 – 0.39 (m, 2H), 0.28 – 0.22 (m, 1H), 0.20 – 0.14 (m, 1H). 19 F NMR (565 MHz, DMSO-d 6 ) δ -121.39 (t, J = 9.2 Hz, 1F), -127.52 (d, J = 10.6 Hz, 1F). LC/MS: Eluent system A (retention time: 4.41 min); ESI-MS: 477 [M + H] + . Compound 4 Synthesis of 7-chloro-N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[( 3S)-2- oxopyrrolidin-3-yl]butan-2-yl}amino)-1-oxopropan-2-yl]-1H-in dole-2-carboxamide, 4 [00211] Compound 4 was synthesized as in Scheme 5. Scheme 5 [00212] Preparation of 7-chloro-N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopyrrol idin-3- yl]butan-2-yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]-1H-indo le-2-carboxamide, (21). To a solution of (18) (162 mg, 0.460 mmol) and 7-chloro-1H-indole-2-carboxylic acid (12) (99.1 mg, 0.507 mmol) in anhydrous DMF (10 mL) cooled in an ice bath was added HATU (193 mg, 0.507 mmol). Then NMM (152 µL, 1.38 mmol) was added dropwise. After 45 min, ice-water mixture (100 mL) was added. The resulting precipitate was collected by filtration, washed the solid with water (3 x 2 mL) and dried under vacuum. The solid was dissolved in CHCl 3 (10 mL) and loaded on 2 x 12 g silica gel columns (Silicycle) and product purified by Biotage ® with a gradient of 0 to 1% MeOH in CHCl 3 , which generated (21) (143 mg, 63% yield) as an off-white solid. Preparation of (3S)-3-{[N-(7-chloro-1H-indole-2-carbonyl)-3-cyclopropyl-L-a lanyl]amino}-2- oxo-4-[(3S)-2-oxopyrrolidin-3-yl]butyl oxo(phenyl)acetate, (22). A mixture of oxo(phenyl)acetic acid (10) (261 mg, 1.74 mmol) and sodium tert-butoxide (83.4 mg, 0.868 mmol) in anhydrous DMF (5 mL) was stirred at room temperature for 30 min. To this mixture, a solution of (21) (143 mg, 0.290 mmol, in 5 mL of DMF), followed by NaI (86.8 mg, 0.579 mmol) were added. After overnight at room temperature, it was poured into crushed ice, the precipitate was filtered, washed with water (3 x 2 mL), and dried under vacuum. The residue was dissolved in CHCl 3 (20 mL) and loaded on 2 x 12 g silica gel columns (Silicycle) and product purified by Biotage ® with a gradient of 0 to 1% MeOH in CHCl 3 , which generated (22) (102 mg, 58% yield) as an off-white solid. [00213] Preparation of 7-chloro-N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[( 3S)-2- oxopyrrolidin-3-yl]butan-2-yl}amino)-1-oxopropan-2-yl]-1H-in dole-2-carboxamide, 4. To a solution of (22) (102 mg, 0.167 mmol) in THF (10 mL) at room temperature under nitrogen atmosphere was added a solution of CsF (2.5 mg, 0.017 mmol) in MeOH (10 mL). After overnight at ambient temperature, the mixture was concentrated under reduced pressure (bath temperature kept at 30 ºC), co-evaporated with DCM (1 x 10 mL). The residue was dissolved in CHCl 3 (10 mL) with 5 drops of MeOH and loaded on 2 x 12 g silica gel columns (Silicycle) and product purified by Biotage ® with a gradient of 0 to 2% MeOH in CHCl 3 , the resulting solid was triturated with Et2O (5 mL) and hexanes, filtered, and the solid washed with hexanes (3 x 2 mL), dried under vacuum, which generated 4 (43 mg, 54% yield) as an off-white solid. 1 H NMR (600 MHz, DMSO-d 6 ) δ 11.74 (d, J = 1.5 Hz, 1H), 8.65 (d, J = 7.9 Hz, 1H), 8.55 (d, J = 8.1 Hz, 1H), 7.63 (s, 1H), 7.62 (s, 1H), 7.31 (dd, J = 0.8, 7.5 Hz, 1H), 7.26 (d, J = 2.1 Hz, 1H), 7.07 (t, J = 7.7 Hz, 1H), 5.07 (t, J = 5.9 Hz, 1H), 4.61 – 4.56 (m, 1H), 4.46 (ddd, J = 3.9, 7.9, 11.4 Hz, 1H), 4.24 (dd, J = 6.1, 18.7 Hz, 1H), 4.15 (dd, J = 5.8, 18.7 Hz, 1H), 3.16 – 3.05 (m, 2H), 2.33 – 2.26 (m, 1H), 2.13 – 2.07 (m, 1H), 1.92 (ddd, J = 4.0, 11.2, 13.8 Hz, 1H), 1.80 – 1.72 (m, 1H), 1.67 – 1.60 (m, 2H), 1.69 – 1.53 (m, 1H), 0.88 – 0.81 (m, 1H), 0.47 – 0.37 (m, 2H), 0.23 – 0.19 (m, 1H), 0.16 – 0.11 (m, 1H). LC/MS: Eluent system A (retention time: 4.77 min); ESI-MS: 475 [M + H] + . Compound 5 Synthesis of N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxo pyrrolidin-3- yl]butan-2-yl}amino)-1-oxopropan-2-yl]-7-fluoro-1H-indole-2- carboxamide, 5 [00214] Compound 5 was synthesized as in Scheme 6. Scheme 6 [00215] Preparation of N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopyrrolidin-3-yl ]butan- 2-yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]-7-fluoro-1H-indo le-2-carboxamide, (24). To a mixture of 7-fluoro-1H-indole-2-carboxylic acid (23) (95 mg, 0.53 mmol) and HATU (203 mg, 0.53 mmol) in anhydrous DMF (5 mL) was cooled in an ice bath. After stirring for 15 min, NMM (151 mg, 1.51 mmol) was added followed by N-{(2S)-4-chloro-3-oxo-1-[(3S)-2- oxopyrrolidin-3-yl]butan-2-yl}-3-cyclopropyl-L-alaninamide hydrochloride salt (18) (171 mg, 0.49 mmol, in 2 mL of DMF). After 45 min., the mixture was poured into crushed ice, extracted with ethyl acetate (3 x 25 mL) and concentrated under reduced pressure. The resulting residue was then dissolved in CHCl 3 (5 mL) and product purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which generated (24) (138 mg, 60% yield) as a white foam. [00216] Preparation of (3S)-3-{[3-cyclopropyl-N-(7-fluoro-1H-indole-2-carbonyl)-L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopyrrolidin-3-yl]butyl oxo(phenyl)acetate, (25). A mixture of oxo(phenyl)acetic acid (10) (99 mg, 0.66 mmol) and sodium tert-butoxide (34 mg, 0.35 mmol) in anhydrous DMF (5 mL) was stirred at room temperature. After 30 min, a solution of (24) (138 mg, 0.29 mmol, in 2 mL of DMF) and NaI (5 mg, 0.03 mmol) were added. After 24 h, the mixture was poured into crushed ice and a solid formed, which was collected by filtration. The collected solid was dissolved in CHCl 3 (5 mL) and product was purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which afforded (25) (102 mg, 60% yield) as an off-white solid. [00217] Preparation of N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2- oxopyrrolidin-3-yl]butan-2-yl}amino)-1-oxopropan-2-yl]-7-flu oro-1H-indole-2-carboxamide, 5. To a solution of (25) (102 mg, 0.17 mmol) in THF/methanol (10 mL/10 mL) at room temperature under nitrogen atmosphere was added CsF (5 mg, 0.03 mmol) as a solid. After overnight, the mixture was concentrated under reduced pressure and the product purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which afforded 5 (38 mg, 48% yield) as an off-white solid. 1 H NMR (600 MHz, DMSO-d 6 ) δ 12.06 (s, 1H), 8.54 – 8.48 (m, 2H), 7.61 (br s, 1H), 7.49 – 7.43 (m, 1H), 7.32 – 7.26 (m, 1H), 7.06 – 6.97 (m, 2H), 5.06 (t, J = 5.8 Hz, 1H), 4.59 – 4.52 (m, 1H), 4.50 – 4.44 (m, 1H), 4.28 – 4.20 (m, 1H), 4.19 – 4.11 (m, 1H), 3.16 – 3.05 (m, 2H), 2.34 – 2.26 (m, 1H), 2.15 – 2.05 (m, 1H), 1.95 – 1.87 (m, 1H), 1.81 – 1.72 (m, 1H), 1.67 – 1.59 (m, 2H), 1.58 – 1.50 (m, 1H), 0.88 – 0.79 (m, 1H), 0.47 – 0.34 (m, 2H), 0.24 – 0.17 (m, 1H), 0.16 – 0.08 (m, 1H). 19 F NMR (565 MHz, DMSO-d 6 ) δ - 131.51 – -131.55 (m, 1F). LC/MS: Eluent system A (retention time: 3.86 min); ESI-MS: 459 [M + H] + . Compound 9 Synthesis of N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2-oxo piperidin-3- yl]butan-2-yl}amino)-1-oxopropan-2-yl]-7-fluoro-1H-indole-2- carboxamide, 9 [00218] Compound 9 was synthesized as in Scheme 7. Scheme 7 [00219] Preparation of N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperidin-3-yl] butan- 2-yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]-7-difluoro-1H-in dole-2-carboxamide, (26). To a solution of N-{(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperidin-3-yl]butan-2-y l}-3-cyclopropyl-L- alaninamide hydrochloride salt (7) (158 mg, 0.430 mmol) and 7-difluoro-1H-indole-2- carboxylic acid (23) (84.9 mg, 0.473 mmol) in anhydrous DMF (5 mL) cooled in an ice bath was added HATU (180 mg, 0.473 mmol). Then NMM (142 µL, 1.29 mmol) was added dropwise. After 45 min, ice-water mixture (25 mL) was added and the resulting mixture was extracted with EtOAc (3 x 25 mL). The combined organic layer was washed with saturated brine solution (1 x 25 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was dissolved in CHCl 3 (10 mL) and loaded on 25 g silica gel column (Silicycle) and product purified by Biotage ® with a gradient of 0 to 2% MeOH in CHCl 3 , which generated (26) (136 mg, 65% yield) as an off-white solid. [00220] Preparation of (3S)-3-{[3-cyclopropyl-N-(7-fluoro-1H-indole-2-carbonyl)-L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl oxo(phenyl)acetate, (27). A mixture of oxo(phenyl)acetic acid (10) (250 mg, 1.67 mmol) and sodium tert-butoxide (80.1 mg, 0.833 mmol) in anhydrous DMF (5 mL) was stirred at room temperature for 30 min. To this mixture, a solution of (26) (136 mg, 0.278 mmol, in 5 mL of DMF), followed by NaI (83.3 mg, 0.556 mmol) were added. After overnight, the mixture was poured into crushed ice-water mixture, extracted with EtOAc (3 x 25 mL). The combined organic layer was washed with saturated brine solution (1 x 25 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was dissolved in CHCl 3 (10 mL) and loaded on 25 g silica gel column (Silicycle) and product purified by Biotage® with a gradient of 0 to 1% MeOH in CHCl 3 , which generated (27) (90.1 mg, 78% yield) as an off-white solid. [00221] Preparation of N-[(2S)-3-cyclopropyl-1-({(2S)-4-hydroxy-3-oxo-1-[(3S)-2- oxopiperidin-3-yl]butan-2-yl}amino)-1-oxopropan-2-yl]-7-fluo ro-1H-indole-2-carboxamide, 9. To a solution of (27) (90.1 mg, 0.149 mmol) in THF (10 mL) at room temperature under nitrogen atmosphere was added a solution of CsF (2.3 mg, 0.015 mmol) in MeOH (10 mL). After overnight, the mixture was concentrated under reduced pressure (bath temperature kept at 30 ºC), then co-evaporated with DCM (1 x 10 mL). The residue was dissolved in CHCl 3 (10 mL) with 5 drops of MeOH and loaded on 2 x 12 g silica gel columns (Silicycle) and product purified by Biotage® with a gradient of 0 to 2% MeOH in CHCl 3 , the resulting solid was triturated with Et 2 O (5 mL) and excess hexanes, filtered, and the solid washed with hexanes (2 x 2 mL), dried under vacuum, which generated 9 (55.1 mg, 78% yield) as an off-white solid. 1 H NMR (600 MHz, DMSO-d 6 ) δ 12.05 (d, J = 1.1 Hz, 1H), 8.52 – 8.50 (m, 2H), 7.47 – 7.44 (m, 1H), 7.42 (br s, 1H), 7.30 – 7.27 (m, 1H), 7.05 – 6.98 (m, 2H), 5.04 (t, J = 6.0 Hz, 1H), 4.58 – 4.53 (m, 1H), 4.52 – 4.47 (m, 1H), 4.24 (dd, J = 6.1, 18.8 Hz, 1H), 4.14 (dd, J = 5.8, 18.8 Hz, 1H), 3.13 – 3.03 (m, 2H), 2.23 – 2.16 (m, 1H), 2.11 (ddd, J = 4.2, 11.5, 13.8 Hz, 1H), 1.89 – 1.82 (m, 1H), 1.79 – 1.72 (m, 1H), 1.72 – 1.63 (m, 2H), 1.58 – 1.47 (m, 2H), 1.38 – 1.30 (m, 1H), 0.88 – 0.78 (m, 1H), 0.47 – 0.35 (m, 2H), 0.24 – 0.18 (m, 1H), 0.16 – 0.10 (m, 1H). 19 F NMR (565 MHz, DMSO-d 6 ) δ -131.52 – -131.61 (m, 1F). LC/MS: Eluent system A (retention time: 4.10 min); ESI-MS: 473 [M + H] + . Compound 17 Synthesis of (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl L-valinate hydrochloride salt, 17 [00222] Compound 17 was synthesized as in Scheme 8. Scheme 8 [00223] Preparation of (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl N-(tert-butoxycarbonyl)-L-valinate, (29). A mixture of Boc-L-valine (28) (80 mg, 0.53 mmol) and sodium tert-butoxide (25 mg, 0.26 mmol) in anhydrous DMF (5 mL) was stirred at room temperature for 30 min. To this mixture, a solution of N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2-oxopiperidin-3-yl] butan-2- yl}amino)-3-cyclopropyl-1-oxopropan-2-yl]-5,7-difluoro-1H-in dole-2-carboxamide (9) (110 mg, 0.22 mmol, in 2 mL of DMF) and NaI (3.2 mg, 0.02 mmol) were added. After 24 h, the resulting mixture was poured into crushed ice to form a solid and filtered. The solid was dissolved in CHCl 3 (5 mL) and product purified by silica column chromatography (gradient of 0 to 10% MeOH in CHCl 3 ), which afforded (29) (82 mg, 55% yield) as an off-white solid. [00224] Preparation of (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl L-valinate-hydrogen chloride, 17. To a solution of (29) (70 mg, 0.10 mmol) in CH 2 Cl 2 (5 mL) at room temperature was added HCl (0.3 mL, 1.2 mmol, 4M in dioxane) slowly. The mixture was stirred overnight, and then concentrated under reduced pressure, and the resulting gum was suspended in ether (10 mL) and filtered which afforded 17 (38 mg, 60% yield) as a white solid. 1 H NMR (600 MHz, DMSO-d 6 ) δ 12.20 (br s, 1H), 8.67 (br d, J = 7.5 Hz, 1H), 8.64 (br d, J = 6.4 Hz, 1H), 8.36 (br s, 3H), 7.46 (br s, 1H), 7.35 – 7.27 (m, 2H), 7.13-7.05 (m, 1H), 5.19 – 4.97 (m, 2H), 4.55 – 4.47 (m, 2H), 4.09 – 4.02 (m, 1H), 3.14 – 3.03 (m, 2H), 2.26 – 2.18 (m, 2H), 2.18 – 2.14 (m, 1H), 1.87 – 1.81 (m, 1H), 1.81 – 1.75 (m, 1H), 1.73 – 1.66 (m, 2H), 1.60 – 1.48 (m, 2H), 1.39 – 1.29 (m, 1H), 1.06 – 0.98 (m, 6H), 0.88 – 0.80 (m, 1H), 0.46 – 0.36 (m, 2H), 0.24 – 0.18 (m, 1H), 0.15 – 0.10 (m, 1H). 19 F NMR (565 MHz, DMSO-d 6 ) δ -121.34 (br t, J = 9.2 Hz, 1F), -127.53 (br d, J = 9.2 Hz, 1F). LC/MS: Eluent system A (retention time: 4.57 min); ESI-MS for free base: 590 [M + H] + . Compound 19 Synthesis of (3S)-3-{[3-cyclopropyl-N-(7-fluoro-1H-indole-2-carbonyl)-L-a lanyl]amino}-2- oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl L-valinate hydrochloride salt, 19 [00225] Compound 19 was synthesized as in Scheme 9. Scheme 9 [00226] Preparation of (3S)-3-{[3-cyclopropyl-N-(7-fluoro-1H-indole-2-carbonyl)-L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl N-(tert-butoxycarbonyl)-L-valinate, (30). A mixture of N-(tert-butoxycarbonyl)-L-valine (28) (727 mg, 3.34 mmol) and sodium tert-butoxide (161 mg, 1.67 mmol) in anhydrous DMF (10 mL) was stirred at room temperature for 30 min. To this mixture, a solution of N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)-2- oxopiperidin-3-yl]butan-2-yl}amino)-3-cyclopropyl-1-oxopropa n-2-yl]-7-fluoro-1H-indole-2- carboxamide (26) (411 mg, 0.837 mmol, in 10 mL of DMF), followed by NaI (125 mg, 0.837 mmol) were added. After overnight, the mixture was poured into crushed ice to form a solid, that was filtered, washed with water (3 x 2 mL), and dried under vacuum. The residue was dissolved in CHCl 3 (10 mL) and loaded on 25 g silica gel column (Silicycle) and the product purified by Biotage ® with a gradient of 0 to 4% MeOH in EtOAc, which generated (30) (195 mg, 35% yield) as an off-white solid. [00227] Preparation of (3S)-3-{[3-cyclopropyl-N-(7-fluoro-1H-indole-2-carbonyl)-L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl L-valinate hydrochloride salt, 19. A solution of (30) (195 mg, 0.290 mmol) in CH 2 Cl 2 (10 mL) was cooled in an ice-water bath and then 4M HCl in 1,4-dioxane (2 mL) was added. After 30 min, the ice-bath was removed. After overnight, a gummy precipitate formed and the mixture was then concentrated under reduced pressure (bath temperature was kept at 30 ºC), and then co-evaporated with DCM (2 x 5 mL). The resulting solid was triturated with Et 2 O (10 mL), filtered, washed with Et 2 O (2 x 5 mL), and dried under vacuum, which generated 19 (163 mg, 92% yield) as an off-white solid. 1 H NMR (600 MHz, DMSO-d 6 ) δ 12.06 (d, J = 1.5 Hz, 1H), 8.68 (d, J = 7.9 Hz, 1H), 8.59 (d, J = 7.5 Hz, 1H), 8.35 (br s, 3H), 7.47 – 7.44 (m, 2H), 7.31 – 7.29 (m, 1H), 7.06 – 6.99 (m, 2H), 5.13 (d, J = 17.2 Hz, 1H), 5.05 (d, J = 17.2 Hz, 1H), 4.55 – 4.48 (m, 2H), 4.09 – 4.04 (m, 1H), 3.13 – 3.03 (m, 2H), 2.25 – 2.14 (m, 3H), 1.87 – 1.81 (m, 1H), 1.81 – 1.75 (m, 1H), 1.74 – 1.65 (m, 2H), 1.59 – 1.48 (m, 2H), 1.38 – 1.30 (m, 1H), 1.04 (d, J = 7.0 Hz, 3H), 1.02 (d, J = 7.0 Hz, 3H), 0.89 – 0.80 (m, 1H), 0.47 – 0.36 (m, 2H), 0.24 – 0.18 (m, 1H), 0.15 – 0.09 (m, 1H). 19 F NMR (565 MHz, DMSO-d 6 ) δ -131.47 – -131.61 (m, 1F). LC/MS: Eluent system A (retention time: 3.98 min); ESI-MS for free base: 572 [M + H] + . Compound 20 Synthesis of (3S)-3-{[3-cyclopropyl-N-(7-fluoro-1H-indole-2-carbonyl)-L-a lanyl]amino}-2- oxo-4-[(3S)-2-oxopiperidin-3-yl]butyl L-valinate hydrochloride salt, 20 [00228] Compound 20 was synthesized as in Scheme 10. Scheme 10 [00229] Preparation of (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopyrrolidin-3-yl]butyl-N-(te rt-butoxycarbonyl)-L-valinate, (31). A mixture of N-(tert-butoxycarbonyl)-L-valine (28) (145 mg, 0.666 mmol) and sodium tert-butoxide (32.0 mg, 0.333 mmol) in anhydrous DMF (10 mL) was stirred at room temperature for 30 min. To this mixture, a solution of N-[(2S)-1-({(2S)-4-chloro-3-oxo-1-[(3S)- 2-oxopyrrolidin-3-yl]butan-2-yl}amino)-3-cyclopropyl-1-oxopr opan-2-yl]-5,7-difluoro-1H- indole-2-carboxamide (19) (110 mg, 0.222 mmol, in 10 mL of DMF), followed by NaI (33.4 mg, 0.222 mmol) were added. After overnight, the mixture was poured into crushed ice to form a solid, and the pH was adjusted to 7 with saturated aq. NaHCO3 solution. The mixture was filtered, and the solid washed with water (3 x 2 mL), and dried under vacuum. The collect solid was dissolved in CHCl 3 (10 mL) and loaded on 2 x 12 g silica gel column (Silicycle) and purified by Biotage ® with a gradient of 0 to 2% MeOH in CHCl 3 , which generated (31) (115 mg, 77% yield) as an off-white solid. [00230] Preparation of (3S)-3-{[3-cyclopropyl-N-(5,7-difluoro-1H-indole-2-carbonyl) -L- alanyl]amino}-2-oxo-4-[(3S)-2-oxopyrrolidin-3-yl]butyl L-valinate hydrochloride salt, 20. A solution of (31) (115 mg, 0.170 mmol) in CH 2 Cl 2 (10 mL) was cooled in an ice-water bath and then 4M HCl in 1,4-dioxane (2 mL) was added. After 30 min, the ice-water bath was removed. After overnight, a gummy precipitate formed. The mixture was then concentrated under reduced pressure (bath temperature was kept at 30 ºC) and co-evaporated with DCM (2 x 5 mL). The resulting solid was triturated with EtOAc (10 mL), filtered, and the solid washed with EtOAc (2 x 2 mL) and after drying under vacuum generated 20 (91 mg, 87% yield) as an off- white solid. 1 H NMR (600 MHz, DMSO-d 6 ) δ 12.22 (s, 1H), 8.67 (d, J = 7.9 Hz, 1H), 8.65 (d, J = 7.5 Hz, 1H), 8.35 (br s, 3H), 7.67 (s, 1H), 7.33 (d, J = 2.1 Hz, 1H), 7.32 – 7.30 (m, 1H), 7.12 – 7.08 (m, 1H), 5.14 (d, J = 17.2 Hz, 1H), 5.05 (d, J = 17.2 Hz, 1H), 4.56 – 4.50 (m, 1H), 4.50 – 4.44 (m, 1H), 4.10 – 4.05 (m, 1H), 3.17 – 3.06 (m, 2H), 2.36 – 2.29 (m, 1H), 2.27 – 2.18 (m, 1H), 2.14 – 2.06 (m, 1H), 2.02 – 1.94 (m, 1H), 1.83 – 1.75 (m, 1H), 1.71 – 1.60 (m, 2H), 1.59 – 1.51 (m, 1H), 1.04 (d, J = 7.0 Hz, 3H), 1.02 (d, J = 7.0 Hz, 3H), 0.88 – 0.80 (m, 1H), 0.47 – 0.36 (m, 2H), 0.25 – 0.18 (m, 1H), 0.15 – 0.09 (m, 1H). 19 F NMR (565 MHz, DMSO-d 6 ) δ - 121.33 (br t, J = 9.2 Hz, 1F), -127.49 (br d, J = 9.2 Hz, 1F). LC/MS: Eluent system A (retention time: 3.78 min); ESI-MS for free base: 576 [M + H] + . EXAMPLE 2: [00231] In this example, methods and results from the development of compounds for use in treating Group IV RNA viruses with improved metabolic stability are provided. METHODS AND MATERIALS SARS-CoV-23-Chymotrypsin-like Protease (3CLP) Expression and Purification [00232] DNA encoding 3CLP from SARS-CoV-2 was obtained from BioBasic Inc. (Ontario, Canada) and codon optimized for expression in Escherichia coli. The gene was cloned into the pET SUMO (small ubiquitin-like modifier) expression vector (Invitrogen). Clones were sequenced to ensure that the SARS-CoV-23CLP protein was in frame with the His-tagged SUMO fusion protein. The resulting plasmid was transformed into E. coli BL21(DE3) and the E. coli transformant was grown in Luria Broth, Miller at 37 °C with shaking (220 rpm) to an OD600 of 0.6-0.7 using kanamycin (50 μg/mL) as selective pressure. Expression of the fusion protein was induced by the addition of 0.5 mM IPTG to the cell culture and the culture was grown for an additional 4-5 h at 37 °C. Cells were harvested by centrifugation (6000 g for 10 min at 4 °C) and suspended in lysis buffer (20 mM Tris-HCl pH 7.8, 150 mM NaCl). Cells were lysed by sonication on ice and the lysate was centrifuged (17000 g for 10 min at 4 °C) to remove cellular debris. The supernatant was isolated and, after adding imidazole (5 mM), mixed with Ni-NTA resin (Qiagen). The mixture was loaded on a fritted column and allowed to flow by gravity at 4 °C. The resin was washed with 10 column volumes (CV) of lysis buffer containing 20 mM imidazole. The fusion protein was eluted using 2 CV of lysis buffer containing increased concentrations of imidazole (40, 60, 80, 100, 200 and 500 mM). Eluted fractions were analyzed by SDS-PAGE and those that contained the fusion protein were pooled together, dialyzed against lysis buffer containing 1 mM DTT at 4 °C and concentrated using Amicon Ultra-15 filter (Millipore) with a MWCO of 10 kDa. The fusion protein was digested by His-tagged SUMO protease (McLab, South San Francisco, CA) at 4 °C for 1-2 h to remove the SUMO tag. The cleavage mixture was added to Ni-NTA resin and loaded on a fritted column. The flow through containing SARS-CoV-23CLP was collected and analyzed by SDS-PAGE. The SARS-CoV-23CLP protein was further purified using size exclusion chromatography (G-100, GE Healthcare, 1 ml/min flow rate, 4°C) in 20 mM Tris, 20 mM NaCl, 1 mM DTT, pH 7.8. Immunoglobulin G, 166 kDa; bovine serum albumin, 67 kDa; ovalbumin, 43 kDa; and lysozyme, 15 kDa were used as calibration standards. Fractions containing the SARS-CoV-23CLP protein were pooled and concentrated using Amicon Ultra- 15 filter with a MWCO of 10 kDa. Mass Spectrometry of SARS-CoV-23CLP [00233] The mass of the free SARS-CoV-23CLP was confirmed by HR-MALDI on a MALDI-TOF (Bruker Ultrafelxtreme, Bruker Daltronics, USA) and LC-MS on an ESI-TOF instrument (Agilent Technologies 6220, California, USA) using electrospray ionization. Determination of Enzyme Inhibition in SARS-CoV-23-Chymotrypsin-like Protease (3CLP)Assay [00234] Compounds described herein were screened for SARS-CoV-23CLP inhibition using a Fluorescence resonance energy transfer (FRET) assay at different compound concentrations. Examples of the resulting concentration response curves are shown in FIGS. 1A and 1B, and examples of the determined 50 percent inhibition concentration (IC 50 ) are provided in TABLE 1. [00235] Synthesis of FRET Peptide Substrate was performed following the procedures Vuong, W. et al., Nature Communications 2020, 11, Article number: 4282. Enzyme Kinetics of SARS-CoV-23CLP [00236] A synthesized fluorescent substrate containing the cleavage site (indicated by the arrow, ↓) of SARS-CoV-23CLP (2-Abz-SVTLQ↓SG-Tyr(NO 2 )-R-NH 2 ) was used for the fluorescence resonance energy transfer (FRET)-based cleavage assay. The protease reaction of SARS-CoV-23CLP towards fluorescent substrate was performed in activity buffer (20 mM Bis Tris, pH 7.8, 1 mM DTT) at 37 °C for 10 min. The final concentration of protease used in the assay was fixed at 80 nM and the concentrations of the substrate were varied from 0.1 to 500 μM. Reaction was started with the enzyme and the fluorescence signal of the Abz-SVTLQ peptide cleavage product was monitored at an emission wavelength of 420 nm with excitation at 320 nm, using an Flx800 fluorescence spectrophotometer (BioTek). Before kinetic calculations, it was verified that the proportionality between the fluorescence emitted and the amount of the substrate used in the assay was linear. The minimal concentration of the enzyme and time of reaction that gave a linear dependence of amount of generated product with time was chosen. Initial velocities in corresponding relative fluorescence units per unit of time (ΔRFU/s) were converted to the amount of the cleaved substrate per unit of time (μM/s) by fitting to the calibration curve of free Aminobenzoyl-SVTLQ. All data are corrected for inner filter effects by an adopted literature protocol. In short, the fluorescence signal (RFU) at each substrate concentration was determined and defined as f(FRET). Then, 5 µL free Aminobenzoyl-SVTLQ at final 5 µM was added to each concentration and fluorescence was taken f(FRET+ Aminobenzoyl-SVTLQ). Simultaneously, a reference reading was taken with the same free Aminobenzoyl-SVTLQ concentration and defined as f(ref). The inner-filter correction was obtained as: corr% = (f (FRET + Aminobenzoyl-SVTLQ) – f (FRET)) / f (ref) x 100% The corrected initial velocity of the reaction was calculated as V = Vo / (corr%). Vo represents the initial velocity of each reaction. [00237] Kinetic constants (vmax and Km) were derived by fitting the corrected initial velocity to the Michaelis-Menten equation, v = vmax × [S] / (Km + [S]) using GraphPad Prism 6.0 software. kcat/Km was calculated according to the equation, kcat/Km = vmax / ([E] x Km). Triplicate experiments were performed for each data point, and the average was determined. Inhibition parameters [00238] Stock solutions of the compounds were prepared with DMSO. For the determination of the IC 50 , 80 nM of SARS-CoV-23CLP was incubated with the compounds at various concentrations from 0 to 100 μM in 20 mM Bis-Tris, pH 7.8, 1 mM DTT at 37 °C for 10 min. The protease reaction was started by addition of 100 μM of the substrate. The GraphPad Prism 6.0 software (GraphPad) was used for the calculation of the IC 50 values. Table 1: SARS-CoV-23CLP activity Evaluation of in vitro inhibition activity of exemplary compounds against SARS-CoV-2 [00239] Compounds described herein were screened for inhibition of SARS-CoV-2 viral replication in an in vitro plaque reduction assay. Examples of the determined effective concentration for 50 percent reduction (EC50) of plaques are provided in TABLE 2. Determination of Inhibition and EC 50 by Plaque Assay [00240] SARS-CoV-2/CANADA/VIDO 01/2020 was a kind gift from Darryl Falzarano (University of Saskatchewan). Vero (Female green monkey kidney) E6 cells were infected with an MOI of .0001 pfu/cell in infection medium consisting of DMEM supplemented with 1x non- essential amino acids (Gibco), 10 mM HEPES, 2% fetal bovine serum, 50 IU/mL penicillin, 50 IU/mL streptomycin with 10 µM or different doses of antiviral drugs. After 1 h, the infecting medium was removed and monolayers were overlaid with MEM supplemented with 10mM HEPES and 1.2% Avicel RC-591 (DuPont). After 48 h, cells were fixed in 10% formaldehyde, and stained using 0.5% (w/v) crystal violet. Plaques were counted and for screening at 10 µM and compounds that did not reduce the plaque numbers by half were assign > 5 µM. The compounds that did reduce viral plaques significantly at 10 µM were tested at multiple concentrations (10, 6, 3, 1, 0.6, 0.3, 0.1, 0.06, and 0.03 µM) and the results were plotted as % inhibition vs the log10[drug] using Prism (GraphPad). EC50’s were determined using a non- linear regression analysis. Experiments were done in triplicate. Measuring Cytotoxicity in A549 and Vero E6 cells [00241] Cell viability was measured using the CellTiter-Glo luminescent cell viability assay (Promega). Separately A549 (male human lung epithelial) cells and VeroE6 cells were seeded at 5x103 cells/well in 96-well plates and incubated overnight before treatment. Compounds were solubilized in DMSO and added to cells in an eight-point four-fold serial dilution (200 µM to 0.0122 µM). Cells were incubated in the presence of compounds for 24 hours before addition of the luminescence substrate and measurement of ATP activity according to manufacturer’s instructions. The percentage of viable cells was calculated relative to cells treated with solvent alone (0.5% DMSO). Table 2: Inhibitor activity against SARS-CoV-2 EXAMPLE 3: Evaluation of in vitro microsomal stability of exemplary compounds [00242] Experiments were performed to assess the stability of the compounds in the presence of mouse and human liver microsomes. The experimental procedures there were used are described below. Examples of the percent remaining of the compound curves in the presence of human microsomes are shown in FIGS. 2A to 2F, and examples of the determined half-life are provided in TABLE 3. Test Compound and Control Working Solution Preparation: [00243] Intermediate solution: 5 μL of compound and control stock solution (10 mM in dimethyl sulfoxide (DMSO)) were diluted with 495 μL of acetonitrile (ACN) (intermediate solution concentration: 100 μM, 99% ACN). [00244] Working solution: 50 μL of compound and control intermediate solution (100 μM) were diluted with 450 μL of 100 mM potassium phosphate buffer (working solution concentration: 10 μM, 9.9% ACN). NADPH Cofactor Preparation: [00245] The appropriate amount of NADPH powder was weighed and diluted into a 10 mM MgCl 2 solution (working solution concentration: 10 unit/mL; final concentration in reaction system: 1 unit/mL). Liver Microsomes Preparation: [00246] The appropriate concentrations of microsome working solutions were prepared in 100 mM potassium phosphate buffer. Stop Solution Preparation: [00247] Cold (4 °C) acetonitrile (ACN) containing 200 ng/mL tolbutamide and 200 ng/mL labetalol as internal standards (IS) was used as the stop solution. Assay Procedure: [00248] Using an Apricot automation workstation, 10 μL/well of compound working solution were added to all 96-well reaction plates except the blank (T0, T5, T10, T20, T30, T60, and NCF60). An Apricot automation workstation was used to add 80 μL/well of microsome solution to all reaction plates (Blank, T0, T5, T10, T20, T30, T60, and NCF60). All reaction plates containing mixtures of compound and microsomes were pre-incubated at 37 °C for 10 minutes. An Apricot automation workstation was used to add 10 μL/well of 100 mM potassium phosphate buffer to reaction plate NCF60. Reaction plate NCF60 was incubated at 37 °C, and timer 1 was started. After pre-incubation, an Apricot automation workstation was used to add 10 μL/well of NADPH regenerating system to every reaction plate except NCF60 (Blank, T0, T5, T10, T20, T30, and T60) to start the reaction. The reaction plates were incubated at 37 °C, and timer 2 was started. An Apricot automation workstation was used to add 300 μL/well of stop solution to each reaction plate at its appropriate end time point to terminate the reaction. Each plate was sealed and shaken for 10 minutes. After shaking, each plate was centrifuged at 4000 rpm and 4 °C for 20 minutes. During centrifugation, an Apricot automation workstation was used to add 300 μL/well of HPLC grade water to eight new 96- well plates. After centrifugation, an Apricot automation workstation was used to transfer 100 μL of supernatant from each reaction plate to its corresponding bioanaylsis plate. Each bioanalysis plate was sealed and shaken for 10 minutes prior to LC-MS/MS analysis. Data Analysis [00249] The equation of first order kinetics was used to calculate T1/2: [00250] Table 3 contains the half-life (t 1/2 or T 1/2 ) in the presence of human and mouse microsomes for select compounds of the invention in comparison to nirmatrelvir [PF-07321332 in Dafydd R. Owen et al, Science 2021, 3741586 - 1593], PBI-0451 [Example 194 in WO2021252644 entitled “Inhibitors of cysteine proteases and methods of use thereof”], AVI- 8004 [Compound 5 in Bing Bai et al, Journal of Medicinal Chemistry 2022, 65, 2905 - 2925] and AVI-8059 [Compound 18a in Bing Bai et al, RSC Medicinal Chemistry 2021, 12, 1722 - 1730].

Table 3: Half-life of the compounds in the presence of liver microsomes [00251] While the present invention has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto.